development of injectable drugs: technology transfer and ... · development of injectable drugs:...

85
Development of Injectable Drugs: Technology Transfer and Process Validation Anna Carolina Myers da Silva Luzia Thesis to obtain the Master of Science Degree in Pharmaceutical Engineering Supervisors: Professor José Monteiro Cardoso de Menezes Specialist Patrícia Alexandre Horta Antunes Examination Committee Chairperson: Professor Pedro Paulo De Lacerda e Oliveira Santos Supervisor: Professor José Monteiro Cardoso de Menezes Member of the Committee: Professor Helena Isabel Fialho Florindo October 2017

Upload: others

Post on 15-Oct-2019

2 views

Category:

Documents


0 download

TRANSCRIPT

Development of Injectable Drugs: Technology Transfer and

Process Validation

Anna Carolina Myers da Silva Luzia

Thesis to obtain the Master of Science Degree in

Pharmaceutical Engineering

Supervisors: Professor José Monteiro Cardoso de Menezes

Specialist Patrícia Alexandre Horta Antunes

Examination Committee

Chairperson: Professor Pedro Paulo De Lacerda e Oliveira Santos

Supervisor: Professor José Monteiro Cardoso de Menezes

Member of the Committee: Professor Helena Isabel Fialho Florindo

October 2017

i

Acknowledgments

Firstly, I would like to express my gratitude to Hikma Pharmaceuticals for the opportunity of attaining

this internship. A special thank you to my Professor Dr. José Cardoso de Menezes, coordinator of the

Master’s degree, for all the support and wisdom transmitted during the course as well as the help

establishing my introduction to the company together with Eng Samuel Camocho.

For the past year, I have learnt so much about how a pharmaceutical company functions, due to all the

people I have had the privilege to work with. Eng. Raquel Marques from the Technical Services

department deserves an enormous amount of credit, for her tireless effort and patience. Teaching me

and answering all my questions about technology transfers/ process validations and allowing me to

accompany the compounding process of new products being manufactured for validation on the

production line. Her intelligence and experience are in fact admirable. A huge thank you to Raquel.

I would also like to thank the professionalism and help of my Supervisor at the Wet-chemistry department

Eng. Patrícia Antunes. For everything I learned from her and for making it possible to conduct my thesis

with the New Projects and Technical Services departments. I would also like to thank Eng. Isabel

Cordeiro (Manager) and Rita Pereira (Supervisor) of the New Projects department for allowing me to

conduct the analytical studies of the new products I accompanied and for later choosing me to take part

in the team.

I wish to convey my appreciation to all my colleagues from the Wet-chemistry team with whom I worked

every day for six months, and to my colleagues at the New Projects with whom I am presently working.

Thank you also to João Sousa, an experienced production operator at Hikma for helping me understand

the functionality of the production line 5.

Last but not least, I would like to thank my parents, my grandparents and my friends for all their support.

A special mention to Catarina Reto, Mariana Rodrigues, Catarina Serra, Catarina Lima, Andreia Revêz,

Rita Saúde, Pedro Gonçalo, Teófilo São Pedro and Joana Leandro.

ii

iii

Abstract

One of the most important aspects in pharmaceutical industries is the manufacturing process. The aim

of the thesis is to contribute to the knowledge about correct validation of processes and successful

technology transfers. Frequently, it is necessary to transfer a technology from a developing site to a

manufacturing site or from one manufacturing site to another for the validation to be completed.

In order to appropriately plan a manufacturing process, quality by design should be followed. Prior

knowledge has to be gained through research and development studies and risk management tools. By

identifying critical quality attributes and critical process parameters it is possible to evaluate risk

scenarios during production and their level of impact. A suitable control strategy is then established

leading to consistent production of quality products. Innovative approaches such as process analytical

technology enable a real-time monitoring and control of the critical aspects of a manufacturing process.

New products transferred to Hikma Portugal are presented including the process design, risk

assessment, evaluation of the preferable conditions for the process at the new site and scale-up.

Keywords: process validation, technology transfer, quality by design, risk assessment, process

analytical technology, scale-up

iv

v

Resumo

Na indústria farmacêutica, definir um processo de produção adequado é um dos aspetos mais

importantes. O objetivo da tese foca-se em contribuir com conhecimento sobre a validação correta de

processos de produção e transferências de tecnologia. De modo a completar uma validação, pode ser

necessário transferir uma tecnologia de um local de investigação e desenvolvimento para um local de

produção, ou de uma fábrica de produção para outra.

A fim de desenvolver adequadamente um processo de produção, a abordagem “quality by design” deve

ser utilizada. O conhecimento prévio relativamente ao produto e ao processo é obtido através de

estudos de investigação e avaliação de risco. Identificando os atributos de qualidade críticos e os

parâmetros críticos do processo é possível avaliar situações de risco que possam ocorrer durante a

produção e o seu nível de impacto. Um sistema de controlo adequado é então estabelecido resultando

em manufatura de produtos com qualidade consistentemente. Abordagens inovadoras, como a

tecnologia analítica de processo, permitem a monitorização e controlo em tempo real dos aspetos

críticos de um processo de produção.

São apresentados dois novos produtos transferidos para a fábrica Hikma em Portugal, incluindo o

desenho do processo, avaliação de risco, análise das condições preferenciais do processo na nova

instalação e aumento de escala.

Palavras-chave: validação de processo, transferência de tecnologia, “quality by design”, avaliação de

risco, tecnologia analítica de processo, aumento de escala

vi

vii

Table of Contents

Acknowledgments .................................................................................................................................. i

Abstract ................................................................................................................................................. iii

Resumo................................................................................................................................................... v

Table of Contents ................................................................................................................................ vii

List of Acronyms .................................................................................................................................. ix

List of Figures ...................................................................................................................................... xii

List of Tables ....................................................................................................................................... xiv

1. Introduction .................................................................................................................................... 1

1.1. Aim of the thesis .................................................................................................................... 1

1.2. Outline of the thesis ............................................................................................................... 1

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

2.1. Technology transfer ............................................................................................................... 3

2.1.1. Definition ................................................................................................................... 3

2.1.2. Classification ............................................................................................................. 3

2.1.3. From drug discovery and development process to technology transfer ................... 4

2.1.4. Reasons for technology transfer ............................................................................... 6

2.1.5. Importance of technology transfer ............................................................................ 6

2.1.6. Factors influencing technology transfer .................................................................... 7

2.1.7. Team and training ..................................................................................................... 8

2.1.8. Documentation .......................................................................................................... 8

2.2. Process validation ................................................................................................................. 9

2.2.1. Definition ................................................................................................................... 9

2.2.2. Resources required ................................................................................................ 10

2.2.3. Need and importance ............................................................................................. 10

2.2.4. Approaches to validation ........................................................................................ 10

2.2.5. Scale-up and risk assessment ................................................................................ 12

2.2.6. Process validation stages ....................................................................................... 15

2.2.7. Qualification stages ................................................................................................ 18

2.2.8. Validation team and responsibilities ....................................................................... 19

2.2.9. Documentation ........................................................................................................ 20

3. Technology transfer between Hikma R&D and Hikma PT ....................................................... 23

3.1. Analytical method development .......................................................................................... 24

viii

3.2. Product development ........................................................................................................... 24

3.3. Submission batches manufacture ....................................................................................... 25

3.4. Industrial facility and production lines, line 5 operation ....................................................... 26

4. Practical examples: Technology transfer of injectable drugs on line 5 ................................. 31

4.1. Product A ............................................................................................................................. 31

4.1.1. Description .............................................................................................................. 31

4.1.2. Materials and compatibility ..................................................................................... 32

4.1.3. Product and process precautions ........................................................................... 33

4.1.4. Manufacturing process ........................................................................................... 34

4.1.5. Risk assessment ..................................................................................................... 34

4.1.6. Process evaluation activities and results ................................................................ 35

4.1.7. Problems during submission batches ..................................................................... 37

4.2. Product B ............................................................................................................................. 40

4.2.1. Description .............................................................................................................. 40

4.2.2. Product and process precautions ........................................................................... 41

4.2.3. Manufacturing process ........................................................................................... 42

4.2.4. Risk assessment ..................................................................................................... 43

4.2.5. Process evaluation activities and results ................................................................ 44

4.2.6. Scale-up .................................................................................................................. 46

5. Optimization of submission and commercial batches – PAT.................................................. 49

6. Conclusion ................................................................................................................................... 53

6.1. Contributions ....................................................................................................................... 53

6.2. Future work .......................................................................................................................... 55

7. References .................................................................................................................................... 57

8. Annexes ........................................................................................................................................ A1

ix

List of Acronyms

ANDA Abbreviated New Drug Application

API Active Pharmaceutical Ingredient

AR Analytical Research

CMA Critical Material Attribute

CoA Certificate of Analysis

CPP Critical Process Parameter

CQA Critical Quality Attribute

DMF Drug Master File

DOE Design Of Experiments

EMA European Medicines Agency

EMEA European Medicines Evaluation Agency

EP European Pharmacopeia

FDA Food and Drug Administration

FMEA Failure Mode Effects Analysis

FMECA Criticality Analysis for Bulk Compounding and Filling

GMP Good Manufacturing Practices

HPLC High Performance Liquid Chromatography

HVAC Heating, Ventilation and Air Conditioning

ICH International Council for Harmonisation of technical requirements for

pharmaceuticals for human use

IQ Installation Qualification

MBR Master Batch Record

MFC Master Formula Card

MHRA Medicines and Healthcare products Regulatory Agency

NIR Near-Infrared

NMT Not More Than

OOS Out Of Specification

x

OQ Operational Qualification

PAC Process Analytical Chemistry

PAT Process Analytical Technology

PDR Process Development Report

PPE Personal Protective Equipment

PPQ Process Performance Qualification

PQ Performance Qualification

PT Portugal

PV Process Validation

PVDF Polyvinylidene difluoride

QA Quality Assurance

QbD Quality by Design

QC Quality Control

QU Quality Unit

R&D Research and Development

RLD Reference Listed Drug

SOP Standard Operating Procedure

STP Standard Test Procedure

TTR Technology Transfer Report

UK United Kingdom

US United States

USP United States Pharmacopeia

VMP Validation Master Plan

WFI Water For Injection

WHO World Health Organization

xi

xii

List of Figures

Figure 1 – Flow chart of the manufacturing process of product A ......................................................... 34

Figure 2 – Root cause map for oxygen headspace OOS for 5 mL/vial presentation batch .................. 38

Figure 3 - Root cause map for oxygen headspace OOS for 1 mL/vial presentation batch ................... 39

Figure 4 – Flow chart of the manufacturing process of product B......................................................... 42

Figure 5 – Process map for process parameters, quality and material attributes for product B (Madieh

2015) ...................................................................................................................................................... 43

Figure 6 - Scheme of process monitoring: at-line, on-line, in-line and off-line ...................................... 50

Figure 7 – Sampling scheme of product A manufacturing process ....................................................... A2

Figure 8 - Sampling scheme of product B manufacturing process ....................................................... A4

xiii

xiv

List of Tables

Table 1 – Information contained within technology transfer documentation (Dogra 2013) ..................... 9

Table 2 – Responsibilities of the authorities of each department (Keyur 2014) .................................... 20

Table 3 – Information available in the TTR ............................................................................................ 25

Table 4 – Hikma’s facilities, production lines, product presentations and type of products .................. 27

Table 5 – Product A presentation of the submission batches ................................................................ 31

Table 6 – Process conditions for product A ........................................................................................... 33

Table 7 – Volume specifications for filling of product A ......................................................................... 37

Table 8 – Product B presentation of the submission batches ............................................................... 40

Table 9 - Process conditions for product B ............................................................................................ 41

Table 10 - Volume specifications for filling of product B ........................................................................ 45

Table 11 - Rational for the proposed batch size of product B ................................................................ 47

Table 12 – FMEA for product A .............................................................................................................. A1

Table 13 – FMEA for product B ............................................................................................................. A3

Table 14 - Risk assessment for the technology transfer of product B from Bedford to Hikma regarding

compounding ......................................................................................................................................... A5

Table 15 - Risk assessment for the technology transfer of product B from Bedford to Hikma regarding

filtration and filling .................................................................................................................................. A6

Table 16 - Risk assessment for the scale-up to commercial size regarding the preparation tank,

dissolution times, mixing speeds and filtration of product B .................................................................. A7

Table 17 - Risk assessment for the scale-up to commercial size regarding the filtration and actual

production yield of product B ................................................................................................................. A8

xv

1

1. Introduction

1.1. Aim of the thesis

The aim of the thesis is to contribute to the knowledge about development of injectable pharmaceutical

products. In particular, how technology transfer and Process Validation (PV) is developed and performed

in pharmaceutical industries, specifically Hikma Pharmaceuticals Portugal. The purpose is also to

examine the possibility of improving the control strategy of submission batches, so that the risk of failed

batches for validation is reduced as well as the following commercial batches.

1.2. Outline of the thesis

The thesis will begin by a literature review based on platforms from organisations such as International

Council for Harmonisation of technical requirements for pharmaceuticals for human use (ICHs), World

Health Organisation (WHO), Food and Drug Administration (FDA) and European Medicines Agency

(EMA). The literature review is also based on articles regarding technology transfers, PV concept and

scale-up. Hikma’s documentation – Standard Operating Procedures (SOPs) – concerning this matter

will also be used.

The thesis is followed by a description of Hikma’s facilities and lines of production, in more detail line 5,

because the new products in development that will be mentioned were produced in this line.

Two practical examples of products for validation are presented. Starting by a detailed description about

the transfer of the injectable products to Hikma PT from Hikma located in the United States (US)

denominated Bedford. The process is explained, including the compounding and filling on line 5. The

evaluation activities performed during production of the submission batches are mentioned together with

the relevant results. Scale-up plan for product B is presented.

The last part includes an investigation about the possibility of control strategy improvement, so that the

risk of failures during validation and consequently during commercial manufacturing are reduced.

Process Analytical Technology (PAT) is taken into consideration – the Critical Process Parameters

(CPPs), Critical Quality Attributes (CQAs) and Critical Material Attributes (CMAs) of the selected new

products in development at Hikma are analysed.

2

3

2. Literature review

2.1. Technology transfer

2.1.1. Definition

The term technology transfer can be defined as the movement of knowledge, skill, organisation, values

and capital from the point of generation to the site of adaptation and application. In the pharmaceutical

industry, technology transfer refers to the processes that are needed for successful progress from drug

discovery to product development, to clinical trials, to full scale commercialization or it can be the

process by which a developer of technology makes its technology available to a commercial partner that

will exploit the technology. In general, technology transfer can be used to refer to movements of

technology from the laboratory to industry, developed to developing countries, or from one application

to another domain. (Feifeit 2008)

The transfer is successful if the receiving unit can routinely reproduce the transferred product, process

or method against a predefined set of specifications as agreed with a sending unit or a development

unit. (Al Ghailani 1995)

2.1.2. Classification

Technology transfer can be classified into vertical and horizontal technology transfer. Vertical transfer

refers to transfer of technology from basic research to applied research, then to development and finally

to production. Horizontal transfer refers to the movement and use of technology used in one place,

organisation, or context to another. (Mansfield 1975)

Vertical tech-transfer can be seen as internal technology transfer and horizontal tech-transfer as external

technology transfer. On this point of view, vertical technology transfer is considered a managerial

process of passing a technology from one phase of its lifecycle to another. Horizontally transfer

reinforces that it may be possible to transfer technology in locality terms at any stage of the technology

lifecycle. (Souder 1990)

The transfer can further be divided in other categories: material transfer, design transfer, and capacity

transfer. Material transfer refers to the transfer of a new material or product while design transfer

corresponds to the transfer of designs and blueprints that can facilitate the manufacturing of the material

or product by the transferee. Capacity transfer involves the transfer of know why and know-how to adapt,

and modify the material or product to suit various requirements. (Steenhuis 2002)

4

2.1.3. From drug discovery and development process to

technology transfer

Technology transfer, as explained previously, is a process to transfer information and technologies

necessary to manufacture quality drug product consistently or it can be the process of taking an

invention from its inception in a laboratory to a commercialized product.

The successful technology transfer from Research and Development (R&D) (the transferring site), to

the commercial production site (the receiving site), is a critical process in the development and launch

of a new medicinal product. It can be extremely costly for a company if something goes wrong during

the transfer process, resulting in delays. Furthermore, it can take increased resource, time and cost to

make corrective actions following an unsuccessful transfer. Progressive pharmaceutical companies are

therefore placing more attention to streamlining and optimising their technology transfer process to

ensure the rapid and successful introduction of a new product to the market. The ideal situation is to

complete the transfer to the production site at an affordable cost. (Ghafaripour 1999)

Technology development has to pass by three stages: the development of a new science, the link

between the new science and technology and the technology being put into products. The drug quality

is designed based on data concerning efficacy and safety obtained from various studies in preclinical

phases and data concerning efficacy, safety and stability of drug products obtained from clinical studies.

The phase I, clinical studies involve small scale studies in patients, they are provided in the form of a

non-optimized formulation, quite different from the intended commercial formulation. There is a high

probability that the studies do not proceed to full development due to toxicity findings or clinical findings

(safety, efficacy and pharmacokinetics/bioavailability). Phase II and III involve longer term safety and

clinical studies in larger groups of patients suffering from the targeted disease. During full development,

the synthetic route for the drug substance is optimized and the manufacturing process is scaled up and

fixed.

If sufficient drug substance is available and the phase III supplies are very large, it may be preferable to

scale-up the manufacturing process to production scale and transfer the process to the commercial

production site and use the supplies from there. A potential risk of transferring early to production is that

all the development work has to be done earlier, the formulation has to be completed as well as the

manufacturing process design. After transferring the process to production it is still possible to perform

adjustments, however it involves documented change controls. Starting the technology transfer before

initiating the phase III studies is also a slight unsafe approach because there is still a relatively high risk

involved during phase II caused by failures related to efficacy and clinical safety.

Regulatory authorities such as FDA, the Medicines and Healthcare products Regulatory Agency

(MHRA) in the United Kingdom (UK) and the European Agency for the Evaluation of Medicinal Products

(EMEA), require three phases of clinical trials and sufficient data to show that the new drug product can

be licensed as safe, effective and of acceptable quality. Once the phase III clinical trials are completed

successfully and the commercial manufacturing process has been transferred from R&D to production

5

site, a regulatory submission can be made. It is important to take into consideration that the technology

transfer, apart from the information related to the process manufacturing, also involves the development

and successful transfer of the analytical and microbiological test methods for the drug substance and

drug product that will be used by the Quality Control (QC) departments at the commercial production

site.

To assure the drug quality, it is desired to make sure what, when and why information should be

transferred to where and by whom and how to transfer, then share knowledge and information of the

drug product between transferring and transferred parties. (Gupta 2012)

As a summary, technology transfers happen mainly between these three points: research phase,

development phase and production phase.

Research phase

The research site is responsible for the correct pharmaceutical design of the drug. This phase includes

the study of the components/product efficacy, guarantee the avoidance of adverse reactions, assure the

drug stability and analyse the data available to achieve a better knowledge about the product. (Gorman

2002)

Development phase

In order to manufacture drugs with qualities as designed, it is required to establish an appropriate

manufacturing process at a small scale and detect variability factors to assure that the scale-up for

submission and validation purposes will be performed without difficulties. The upper and lower limits of

the manufacturing process including composition and parameters should be challenged during

development. (Patel 2009)

Production phase

To assure the consistency between development and manufacturing, the transferring party in charge of

development should fully understand what kind of technical information is required by the receiving party

in charge of manufacturing. An appropriate evaluation method to determine whether a drug to be

manufactured meets the quality of design should be executed. In case the product is similar to others

before produced, it is fundamental to study the information about the process maintained by the

manufacturer. (Gibson 2001)

6

2.1.4. Reasons for technology transfer

A developer of technology makes its technology available to another person to exploit for several

reasons. It can occur that the original inventor of technology may have the resources to conduct early-

stage research such as animal studies and toxicology study, but does not have the resources to take

technology through its clinical phases. Another frequent situation is having the resources to develop the

product, however it might not be sufficient to take the technology through its regulatory phases, for this

reason the developer must collaborate with another organization to take the product into market.

The developer of the technology may have developed the product to a state almost ready to launch,

nevertheless the manufacturing capability and resources available may only be suitable for small scale

production. A partnership with an organization with a large scale capability is necessary.

It can also happen that a full development of the technology has been completed with regulatory

approvals and product registrations for the product to be sold, however it can lack the marketing and

distribution channels. Due to this deficiency a collaboration with another organization which has that

capability is necessary. (Patil 2010)

2.1.5. Importance of technology transfer

It has been recognized that the transfer of technology is essential for the process of economic growth,

and that the progress of both developed and developing countries depends on the efficiency of such

transfer. A firm and its partners collaborating in the technology transfer will gain financial and strategic

benefits, the means not available at one site compensates the resources available in another. Working

in partnership makes it possible to accomplish every stage. The importance of technology transfer has

also stimulated university industry technology transfer.

In the pharmaceutical industry, the intersection between business and science is both essential and

critical to the drug discovery and development process for a new medicinal product. Technology transfer

allows the link between R&D, process development and production for commercialization. If it is

implemented thoughtfully then production process runs efficiently, the risks during production are

minimized and a robust process for routine commercial manufacturing is achieved.

Appropriate technology transfer is essential to upgrade the quality of design to be the quality of product,

and ensure stable and high quality of the product. (Osman 1999)

7

2.1.6. Factors influencing technology transfer

Drivers

Technology transfer has several advantages. As mentioned it allows a promising market scale because

if a specific process is transferred to a larger country the business and marketing potential is greater.

The countries have a supportive environment that includes strong intellectual property and enforcement

to successfully attract imported technology, which facilitates the transfer. Factors as skilled workforce

working together such as engineers and managers also contribute to efficiency in the results. Increased

information exchange including effective systems that identify who is interested in purchasing the

technology and entities willing to transfer their technology are easy ways to facilitate the task.

The prospect of technology transfer can be very desirable to local pharmaceutical manufacturers even

in another viewpoint. The technology, new machinery, training, among other transfer additional benefits

can then be applied profitably for other production purposes. (Donald et al 1995) (Akhavan 1995)

Barriers

Technology transfer can face different difficulties. For example, there is a need of skilled labour however

the unattractive conditions of service are a negative contributor.

In the cases where the knowledge and awareness were not accomplished at a high level by the

developer of the technology, the transfer to the new site is more complex. The lack of government focus

at times towards the technology transfer approach and the high cost for prequalification can also bring

complications, monetarily. The funding on important areas of research should also be higher.

Furthermore, there are controls and restrictions on technology exportation established by national

security which makes it harder to perform the transfer internationally. Another problem found is the

reduced access to online scientific journals, for the R&D site it can raise difficulties during the

development process. (Ortega 2009)

Approaches to overcome obstacles

In order to overcome the problems faced by technology transfer certain measures can be taken. For

instance, it is preferable to commercialize publicly funded technologies so that the costs involved are

not as high. Political stability will influence the rate of inward technology transfer and a good leadership

is essential for a strengthened healthcare system. Incentives designed to encourage technology transfer

such as adequate capital markets should be implemented.

Regarding the difficulty of not having access to important scientific discovers online, research tools for

patents and an international treaty on scientific access could solve the problem. (Madu 1989)

8

2.1.7. Team and training

The technology transfer team is meant to develop and implement a methodology that ensures the

effective and efficient transfer of robust and candidate production processes from development to

manufacturing.

The team is trained on the technology transfer process, so that each member is familiar with both the

business methodology to be used and the technical aspects of the process being transferred. Each

member must understand process tools, to help organize and assemble numerical data, assign action

items and identify areas of weakness or omission. Successful technology commercialization depends

on a skilled workforce in management, production, sales, distribution, and support.

In order to obtain a successful project, it is necessary to implement an effective training for all involved

in the new process, including production operators and regulatory team members which will prepare the

submission. The training is applied on the transfer process and consequently provides a good

knowledge about the process technology. In accordance to the current Good Manufacturing Practices

(GMP) requirements, all training has to be documented and all members which interact with the new

technology have to be trained. (Souder 1990)

2.1.8. Documentation

The documentation for technology transfer includes content for transferring and transferred parties, it

should be always available and traceable. Task assignment and responsibilities should be clarified. The

Quality Assurance (QA) department at the manufacturing site checks and approves the technology-

transfer documentation. Information from the transferring site is essential so that the technology transfer

team can better evaluate options and can distinguish the critical from the incidental. The reasons for the

selection of particular unit operations, equipment, and conditions, should be well described in the

documentation so that every step can be clearly understood. (Ali 2012)

In the cases where the transfer happens from R&D site to manufacturing site, the technology transfer

dossier provided to the production site includes documentation for the transfer of the analytical methods.

It contains information about formulation and drug product such as the Master Formula Card (MFC)

which comprises the product name, strength, generic name, shelf life and markets of interest. It also

includes the master formula (formulation, steps of manufacturing process and environment conditions

required) and the master packaging card (packaging type, material used for packaging, stability and

shelf life). The specifications and Standard Test Procedure (STP) have to be included together with

information about the Active Pharmaceutical Ingredient (API) and excipients profile, in-process

parameters and finished product details. (Dogra 2013)

Table 1 presents the data included in the existing types of documents for technology transfer.

9

Table 1 – Information contained within technology transfer documentation (Dogra 2013)

R&D report Product

specification file

Technology

transfer plan

Technology

Transfer Report

(TTR)

Info

rmati

on

Development of the new

drug

Product

manufacturing

Items and contents

of technology to be

transferred

Activities and results

performed by

transferring party

Raw materials, API and

impurities

Assurance of

operation safety

Procedures of

transfer and transfer

schedule

Knowledge to

achieve a

successful transfer

Synthetic route, formula

design

Environmental

impact assessment

Judgment criteria for

the completion of

the transfer

Stability data Costs

Specifications and test

methods

2.2. Process validation

2.2.1. Definition

Validation means “action of proving effectiveness”. There are several similar definitions, on the first

stages it can be defined as collecting and evaluating data. (Sarvani 2013)

The European commission, FDA and ICH consider PV as documented evidence with high degree of

assurance that a process, operated within established parameters, can perform effectively and

reproducibly to produce a medicinal product meeting its predetermined specifications and quality

attributes. (WHO 1996)

10

2.2.2. Resources required

A validation requires three main aspects: time, financial means and skilled labour. It can take a lot of

time to complete all the stages, however rigorous time schedules will have to be accomplished to assure

the availability of the product in the market when planned. A high financial plafond must be available by

the manufacturing site because validation requires specialized personnel and expensive technology.

The need of a multidisciplinary team depends on the process and product to be validated, it comprises

quality assurance, engineering, manufacturing and other disciplines. (WHO 2006)

2.2.3. Need and importance

Validation in itself does not improve processes however it confirms the efficiency of the developed

processes and proves that they are under control. It can help to reduce the quality costs, which are

divided into four categories: preventive costs, appraisal costs, internal failure costs and external failure

costs.

A scientifically studied and controlled validated process makes it less likely that defective products are

sent to the market and also reduces customer complaints. It is proven that batches fail less, as a

consequence the output is increased and productivity is higher. Another important quality that results

from validation is the increase in safety. Tested and approved equipment and materials during

qualification and validation assure that the product is produced safely. The obligation by regulatory

governments to calibrate certain equipment, and to perform periodic maintenance also improves the

security. Furthermore, there is an advantage for employees, their execution is improved due to the

previous awareness of the process during the validation.

Government regulation obliges compliance with validation requirements to obtain approval for

manufacture and to introduce new products, this helps to assure that all processes implemented are

controlled and suitable. (Keyur 2014)

2.2.4. Approaches to validation

Validation can be prospective, concurrent, retrospective or revalidation depending on when it is

performed in relation to production.

Prospective validation

Prospective validation is adopted for when new drug products are introduced. It is carried out in the

development stage, a risk analysis of the production process is made. The process is broken down into

individual steps which are evaluated based on data from experimentation to determine if they might lead

to critical situations. If critical points are found, the risk is evaluated and the potential causes are

11

investigated. Trials are performed, and if the results are acceptable, the process is suitable. (Sarvani

2013)

Unsatisfactory processes are modified and improved until a validation demonstrates that they are

acceptable. This type of validation limits the risk of errors occurring on the production scale. (WHO 1996)

Concurrent validation

Concurrent validation takes place during normal production. This method is effective if the development

stage has resulted in sufficient knowledge and understanding of the process. The first three production

scale batches must be well monitored, the specifications of subsequent in-process and final tests are

based on the monitoring results. (WHO 1996)

With the documented results the process is proven to be in a state of control. Concurrent validation with

a trend analysis including stability should be done throughout the lifecycle of the product. (Sarvani 2013)

Retrospective validation

Retrospective validation is chosen for established products with stable processes. This type of validation

comprises experience of production, it assumes that composition, procedures, and equipment remain

unchanged. The experience and the results of in-process and final product testing are then evaluated.

Failures that occur in production are analysed to determine the limits of process parameters. (Sarvani

2013)

A trend analysis is conducted to determine the extent which the process parameters are within the

permissible range. This mean of validation should never be applied to new processes or products, it is

used only in special circumstances. Retrospective validation can be useful in establishing priorities for

the validation programme. If the results of a retrospective validation are positive, it indicates that the

process does not need immediate consideration and may be validated normally. (WHO 1996)

Revalidation

Revalidation is used when it is necessary to prove that intentional or unintentional changes in the

process or in the process environment, do not adversely affect process characteristics and product

quality. Revalidation can be executed after a change with impact on product quality, or periodically at

scheduled intervals.

When the revalidation occurs after a change, it is necessary to implement the alterations affecting the

manufacturing or standard procedure and perform again the same tests made in the first validation.

Each modification should be reviewed by a qualified validation group to conclude if it is sufficient to opt

12

by revalidation, and if it is, in which extent. Unexpected changes and deviations can be observed during

self-inspection or audit, or during the continuous trend analysis of process data.

The changes are usually on starting materials, physical properties (density, viscosity, particle size

distribution and crystal type of the active ingredients or excipients), packaging material (for example

replacing plastics by glass), process (changes in mixing time, drying temperature and cooling regime

can affect process steps and product quality), equipment, production area and support system. The

rearrangement of manufacturing areas or support systems such as ventilation may result in changes in

the process, revalidation may be necessary predominantly in sterile products manufacturing.

When the revalidation is periodic, scheduled times for analysis are organised even if no changes have

been made (just for caution). This happens because process changes may occur gradually even if

experienced operators work correctly according to established methods, so as equipment which can

also have continuing changes.

For scheduled revalidation the following points have to be checked: changes in the master formula,

methods, batch sizes, analytical control methods and if it impacts the product; confirmation that

calibrations and preventive maintenance has been made in accordance with the established programme

and time schedule; cleaning programmes have been carried out and SOPs have been updated correctly

and implemented.

Periodic revalidation is based on a review of historical data (in-process and finished product testing after

the latest validation) to verify that the process is under control.

In some processes, such as sterilization, additional process testing is required to complement the

historical data. (Sarvani 2013)

So basically, there are two approaches to validation, one based on evidence obtained through testing

(prospective and concurrent validation), and one based on the analysis of accumulated/ historical data

(retrospective validation). Whenever possible, prospective validation is preferred. Retrospective

validation is no longer encouraged and is, in any case, not applicable to the manufacturing of sterile

products. (WHO 1996)

2.2.5. Scale-up and risk assessment

Scale-up of pharmaceutical manufacturing processes demand a combination of experience, science

and engineering. In order to achieve a successful scale-up between the different phases, it is essential

that the Critical Material Attributes (CMAs) and Critical Quality Attributes (CQAs) of those materials

involved in the formulation of the product together with the Critical Process Parameters (CPPs) of the

manufacturing process are well defined since the beginning. Risk management tools can help controlling

the critical steps of the process.

13

Risk assessment can be defined as the identification of hazards and the analysis and evaluation of risks

associated with exposure to those hazards. It is commonly understood that risk is the combination of

the probability of occurrence of harm and the severity of that harm, risk scenarios are the events that

identify the risks associated with the use of a system. The adoption of effective risk management is

indispensable to create awareness about the critical steps of the process with most probability to cause

failure, and how to control them.

Process mapping should be undertaken to ensure that all possible risks are considered, it is a map

performed by interdisciplinary teams which provides a better understanding of the process and assists

in providing a structured methodology for risk identification (failure modes identification). After identifying

the risk and its scenario, the effects are considered. The next stage is to determine the likelihood of an

adverse event to happen and assigning a value to that estimate. The parameters probability, severity

and detectability should be identified for each risk. Frequency of probability is rated from 1 to 3 in

accordance to its likelihood:

- Level 1 (low): the frequency of the event occurring is once per ten thousand transactions;

- Level 2 (medium): the frequency of the event occurring is once per thousand transactions;

- Level 3 (high): the frequency of the event occurring is once per hundred transactions.

Severity of the potential effect of the failure requires the team to consider which impact the event has

on the product quality or data integrity. The impact of the consequence is rated also 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 can be

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

Having assigned the probability of the risk and the level of impact that such an event may have, the risk

can be classified. The risk is a multiplication of probability and severity. It can range from:

- Level 1 (high): the probability that this failure appears is high and that the impact on product

quality or data integrity is high or medium;

- Level 2 (medium): moderate impact on product quality or data integrity;

- Level 3 (low): practically no impact on product quality or data integrity.

The detectability stage is performed to identify if the risk can be recognized or detected by other means

in the system. The probability of a risk being detected is rated 1, 2 or 3 according to its detection

possibilities:

- Level 1 (high): 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);

14

- Level 2 (medium): a reliable detection procedure is systematically applied however it gives

delayed results, as another option an indirect measuring device can be used, or a direct

measuring device in line with no alarm (insufficient secured);

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

operation or monitoring (not secured).

The risk evaluation can be done by the comparison against qualitative or quantitative criteria. Historical

data and trends should be evaluated. For quantitative evaluation, risks prioritization can be followed.

Risk priority number is a multiplication of three parameters taking into account the potential failure

associated with the potential effect and its detectability.

- High risk priority number: function or component is critical, validation measures are to be taken;

- Medium risk priority number: function or component is potentially critical, validation measures

are to be taken;

- Low risk priority number: function or component is not critical and there are no validation actions

or measures to be taken.

The risk management strategy has been completed and correctly applied as soon as the risk reduced

to an acceptable level and when the risk control process has been accepted. If the evaluation reached

to the conclusion that controls are not enough the risk reduction process should be evaluated. (ICH

2005)

Application of statistical methods such as Design Of Experiments (DOE) together with advances in

measurement tools such as PAT allow improvements in process understanding and control. DOE are a

mean to gain knowledge about the process and design an effective control strategy by allowing the

establishment of multivariate interactions between the variable inputs (material attributes and process

parameters for example) and the outputs (in-process material, intermediates or final product). It makes

it easier to apply risk management. PAT is an innovative mean of control that includes timely analysis

and control loops to adjust the processing conditions so that the output remains constant. Manufacturing

using this system can provide a higher degree of process control, reducing the risks. (FDA 2011)

These advances are expected to cause a shift from trial and error to rational process scale-up following

Quality by Design (QbD) initiative. QbD is a systematic approach to development that begins with

predefined objectives and emphasizes product/process understanding simultaneously with process

control, based on science and quality risk management. (ICH 2009)

The first studies performed to validate a product start at a laboratory scale, following the pilot batches

and finally the scale-up to production batches. (Gibson 2001)

Laboratory scale batches

These batches are produced at the research and early development laboratory stage, they have a very

small size, approximately one tenth of a normal production batch. The production of these batches is

15

helpful to support formulation and packaging development, clinical and pre-clinical studies. The data

that these batches provide are useful to define the products characteristics and enable the choice of

appropriate manufacturing process. (Nandhakumar 2011)

Pilot batches

Pilot batches may be used in the process development or optimisation stage to support formal stability

studies and support pre-clinical and clinical evaluation. The size of the batches corresponds to at least

10% of the production scale batch, the multiplication factor for the scale-up does not exceed ten. The

objective of pilot scale batches is to provide data predictive of the production scale product. To continue

developing and to optimise the manufacturing process, pilot scale batches are essential. They make it

possible to analyse and evaluate the difficulties and critical points of the process together with the

methods most appropriate for large scale production.

The production of pilot batches should assure that the product and manufacturing process are

achievable on an industrial scale. (Nandhakumar 2011)

The batches produced for submission of the product to the authorities depend on the risk of the

product/process. However, the usual situation is three batches for each presentation (strength and

size/quantity). A bracketing approach can be used in certain occasions, for example if different sizes are

produced for the same strength it is only necessary to produce three batches of the biggest size and

three batches of the smallest size, the intermediate quantities require the production of only batch.

Production scale batches

The batches have the normal size for production of the routine commercialization of the product. The

evaluation and characterization of the critical process parameters at laboratory or pilot scale made,

followed by the manufacture of submission batches, is completed by a formal validation programme on

production scale batches for which the validation scheme has been applied. (Nandhakumar 2011)

2.2.6. Process validation stages

Process design

Process design is defining a process suitable for routine commercial manufacturing that will be reflected

in planned master production and control records. The objective is to manufacture a product that meets

its quality attributes. An important step in process design is to build and capture process knowledge and

understanding. For the first design experiments good documentation practises and scientific methods

have to be followed though it is not necessary to follow the current GMP conditions. However, for drugs

that are manufactured during stage 2 (process qualification) and stage 3 (continued process verification)

16

it is obligatory. All information must be documented and reviewed, the data should be used for adaptation

throughout the lifecycle of the product.

During the product development, important aspects which are defined such as quality attributes, dosage

form and manufacturing steps, contribute to the process design. Manufacturing equipment, production

operators, contributors to variability and environmental conditions are considered for the design

Computer simulations of certain manufacturing steps can help avoiding problems during commercial

production, however it is important to take into consideration the differences that might exist between

the models and reality. All information which results in process understanding should be documented, it

is used for process qualification and continued process verification.

In order to guarantee quality production, there are different options of process control, for example

material analysis and equipment monitoring at certain processing points. The type and extent of process

controls are decided by previous risk assessments and improved as process experience is gained.

There is a need of special in-process control when the product attribute is not readily measurable due

to limitations of sampling or detectability, or when intermediates/products cannot be highly characterized

and well-defined quality attributes cannot be identified. The controls are listed in the master production

and control records.

The designed commercial process which includes the operational limits and process control strategy,

should be passed to the next stage for confirmation. (FDA 2011)

Process qualification

The objective of the process qualification stage is to evaluate if the process design is capable of

reproducible commercial manufacture. It is necessary to complete this stage successfully in order to

proceed with commercial distribution, GMP-compliant procedures must be followed.

The process qualification is divided in two fundamentals:

- Design of the facility and qualification of utilities and equipment;

- Process Performance Qualification (PPQ).

A correct design of a manufacturing facility is required by the current GMP regulations on buildings and

facilities. The term qualification refers to the activities undertaken to demonstrate that utilities and

equipment are suitable for their intended use, they are completed before starting production at a

commercial scale. The activities include defining construction materials for utilities and equipment,

operating principles, and testing performance characteristics. Utilities and equipment are built and

installed in compliance with the design specifications. The function of the equipment is challenged to

the expected conditions during routine production including interventions, stoppage, and start-up. The

defined operating ranges should be shown as capable of being used.

17

The project plan for qualification should include timing of qualification activities, studies/tests to be

performed, appropriate criteria to evaluate outcomes, responsibilities of relevant departments,

procedures for documenting and approving the qualification and requirements for the evaluation of

changes. The QC unit must review and approve the qualification plan and report.

The PPQ stage confirms the design stage, it happens after the facility, utilities and equipment are

qualified. The personnel are trained to execute the commercial manufacturing process as designed and

to be aware of the control procedures to produce commercial batches. Before the commercial

distribution of the drug product, data regarding all relevant studies such as designed experiments and

laboratory/pilot/commercial batches are evaluated to establish the manufacturing conditions in the PPQ.

Experience gained with similar products and processes can also be helpful. It is advised that PPQ

documentation include description on how to address deviations from expected conditions and handling

of nonconforming data.

The PPQ has a higher level of sampling, additional testing, and greater examination of process

performance than typical routine commercial production in order to confirm uniform product quality

throughout the batch and to establish frequency of sampling and monitoring for the product and process.

In case the process uses PAT, the approach is slightly different. The design and process qualification

stages will have to focus on the measurement system and control loop of the critical defined attributes,

however the scientific basis is the same: the process will have to consistently deliver quality products.

A PPQ protocol (denominated PV protocol in Hikma) has to be written specifying the manufacturing

conditions. It should include operating parameters, controls, processing limits, samples to be collected,

tests to be performed and acceptance criteria. The sampling plan includes sampling points, frequency

of sampling for each unit operation or attribute and number of samples (should be adequate to provide

sufficient statistical confidence of quality within and between batches). The PPQ protocol should also

include information about the design of the facilities, the qualification of utilities and equipment,

personnel training, verified components, verified containers/closures and the adopted validated

analytical methods. The protocol is reviewed and approved by the Quality Unit (QU).

The execution of the PPQ protocol involves manufacturing the product as described in the document,

under normal conditions (of environment and materials for example) by the personnel routinely expected

to perform each step of each unit operation in the process.

After performing the protocol, a report is written with a discussion of all aspects. The data is presented

and analysed, additional observations are mentioned such as deviations or strange test results and

correspondent corrective actions/changes that should be made to procedures or controls. The report

presents a conclusion specifying if the process met the conditions established and whether the process

is considered to be in a state of control. In case it is not, it should be mentioned what can be done for

improvements with a justification for the approval of the process. The report is reviewed and approved

by the relevant departments and QU. (FDA 2011)

18

Continued process verification

The objective of this stage is to guarantee that the process remains in a state of control - the validated

state - during commercial manufacture. Good process design and development should anticipate

sources of variability and establish appropriate detection, control and mitigation strategies.

Nevertheless, a process is likely to encounter sources of variation that were not previously detected or

to which the process was not previously exposed.

Collecting data and performing statistical trend analysis can be taken in order to verify that the quality

attributes are being appropriately controlled throughout the process and to apply corrective actions in

case problems are detected. Continued monitoring of process parameters and quality attributes during

the process qualification stage is essential until sufficient data is available to generate significant

variability estimates and basis for establishing frequency of routine sampling and monitoring for the

process.

It is also important to be aware of variability through defect complaints, out-of-specification findings,

process deviation reports, batch records, incoming raw material records, and adverse event reports.

Production line operators and QU staff should be encouraged to provide feedback on process

performance. The QU ought to meet periodically with production staff to evaluate data, discuss possible

trends or undesirable process variation, and coordinate any correction or follow-up actions by

production.

The data gathered during this stage can help improve and optimize the process/product, such as the

operating conditions (ranges and set-points), process controls, component, or in-process material

characteristics. When changes are made, a document describing the changed plan must be written with

a well justified rationale for the change, an implementation plan, and QU approval. Depending on how

the proposed change might affect product quality, additional process design and process qualification

activities might be necessary.

It is equally essential to guarantee maintenance of the facility, utilities, and equipment to ensure that the

process remains in control. (FDA 2011)

2.2.7. Qualification stages

Validation and qualification are components of the same concept. The term qualification is normally used

for equipment, utilities and systems, and validation for processes. In this sense, qualification is part of

validation.

All SOPs for operation, maintenance and calibration should be prepared during qualification. Training

should be provided to operators and training records should be maintained. There are four stages of

qualification. (WHO 2006)

19

Design Qualification (DQ)

The first stage of qualification is the DQ which provides documented evidence that the design

specifications were met. (Sharma 2013)

Installation Qualification (IQ)

On this stage evidence must be documented proving that the installation was complete and satisfactory.

The purchase specifications, drawings, manuals, spare parts lists and vendor details should be verified

during IQ. Control and measuring devices are calibrated. (Sharma 2013)

Operational Qualification (OQ)

OQ provides documented evidence that utilities, systems or equipment and all its components operate

in accordance with operational specifications. Tests should be designed to demonstrate satisfactory

operation over the normal operating range as well as at the limits of its operating conditions (including

worst case conditions). Operation controls, alarms, switches, displays and other operational

components should be tested. (Sharma 2013)

Performance Qualification (PQ)

PQ is the final step in qualification processes for equipment, this step involves verifying and documenting

that the equipment is working reproducibly within a specified working range. Rather than testing each

instrument individually, they are all tested together as part of a partial or overall process. (Sharma 2013)

2.2.8. Validation team and responsibilities

The validation team comprises several departments which work together at different stages. Table 2

describes the responsibilities of the authority in each department that actively takes part in the validation

of the process (the department names were actualized to the Hikma denomination).

20

Table 2 – Responsibilities of the authorities of each department (Keyur 2014)

Department Designation Responsibility

Technical

services Executive/Officer

To coordinate the entire validation process by scheduling

meetings and discussions with production, AR, and QA.

Preparation of validation protocol, master formula record,

monitoring the process, analysing data and test results. To

review the validation documents. Preparing the final report.

QA Officer

To coordinate the validation process by scheduling meetings

and discussions with the team. Monitoring the process,

analysing data and test results. To review validation

documents.

Production Officer To participate in performing the validation steps during

manufacturing processes. To assist in collection of data.

AR Officer To test and report the results. To review validation documents.

Production General manager To approve the PV protocol and report. To review of validation

documents. To approve the process.

2.2.9. Documentation

Qualification and validation should be done according to written procedures. The documents include the

SOPs, specifications, plans, protocols, reports, risk assessment outcomes, process flow charts,

operator manuals, training records, calibration procedures and records, sampling plans, testing plans

and methods, statistical methods and results, history of qualification or validation and the plan for

ensuring the maintenance of a validation status.

Data from validation should be available for all products to demonstrate the effectiveness of the process.

It should be held at the manufacturing location and accessible for verification by concerned authorities.

(Nandhakumar 2011)

Validation Master Plan (VMP)

The VMP is one of the key documents in the current GMP regulated pharmaceutical industry. It is a

summary document, brief concise and clear. The VMP should not repeat information documented

elsewhere but has a duty to refer to existing documents such as policy documents, SOPs, validation

protocols and reports.

21

VMP gives an overview of the entire validation operation, its organizational structure, content and

planning. It also provides a written program for achieving and maintaining a qualified facility with

validated processes. All validation/qualification activities related to critical technical operations relevant

to product and process controls within a company are included. (Saudi Food & Drug Authority 2010)

The VMP should start with an introduction describing the validation policy, scope, location and schedule.

The document includes information about the product or products to be validated, process description,

critical steps, microbiological monitoring, cleaning validation approach and acceptance criteria. The

building and plant layout of the facility is included together with information regarding construction

materials. The equipment used is described and calibration/maintenance operations too. Utilities such

as heating, ventilation and air-conditioning (HVAC), water, clean steam, compressed air, gases and

vacuum system are installed in the facility and described in the VMP. The responsibilities of the trained

personnel should be well defined. Suppliers inspection, internal audits, time plans of each validation

project and sub-project, re-validation activities, actual status and future planning are also included in the

document. (Nandhakumar 2011)

The VMP should be reviewed at regular intervals and kept up to date according to current GMP. (WHO

2016)

Validation report

A report is written regarding the validation performed. Reports should reflect the protocols and

procedures followed and include at least the title and objective of the study, make reference to the

protocol and to the appropriate risk assessment, details of materials, equipment, programmes and

cycles used, procedures and test methods. Results have to be recorded and in compliance with good

data and record management practices, they are reviewed, analysed and compared against the justified

predetermined acceptance criteria, interpreted and statistically analysed where appropriate. The results

must meet the acceptance criteria. Deviations, out-of-specification and out-of-limit results are

documented and investigated according to appropriate procedures. If the deviations are accepted, they

have to be justified. When necessary, further studies should be performed.

The conclusion of the report states whether the outcome of the qualification or validation was considered

successful, and should make recommendations for future monitoring and define alerts and action limits

in necessary cases.

The departments responsible for the qualification and validation approve the report, production manager

approves the report after the final review. The criteria for approval should be in accordance to the quality

assurance system of the company. Any deviations found during the validation process are managed and

documented. Corrective actions should be considered. (WHO 2016)

The report has the following structure: title and objective of study, reference to protocol, details of

material, equipment, conditions used, details of procedures, information regarding analytical methods

and results. (WHO 1992)

22

23

3. Technology transfer between Hikma

R&D and Hikma PT

The first step of a technology transfer between Hikma’s R&D site and the manufacturing site of Hikma

pharmaceuticals PT is to select the product. A R&D plan is made annually by the management team

specifying which products will be developed each year. Later on, R&D in US or Jordan and technical

services from Portugal coordinate the dates for the products defined in the plan, together with the project

manager at each site, who decide when each step per product has to be completed.

For the API selection it is essential to analyse the Drug Master Files (DMF) of each manufacturer. If a

DMF is not available, a technical package should be requested with information about the synthetic

route, stability data, specifications, justification of specifications, analytical procedures, impurities profile,

origin of each impurity, certificate of analysis and a quality commitment if required.

When choosing the manufacturer, it is important to make sure that the API is being produced with

consistent quality and in a GMP facility. A material sample from the supplier should be requested to

evaluate the API quality, stability and degradation profile. In case the API is to be used as sterile, a sterile

representative sample ought to be provided to test for sterility and bacterial endotoxin. It should be

confirmed that the packaging meets current plant requirements, if it is going to be used inside the sterile

core. In case it is different in any way, a handling SOP modification is required, new sanitization study

and monitoring. The impurities have to be evaluated so it is important that they are available for analytical

activities. However, the API manufacturer should provide a statement for residual solvents, genotoxic

impurities and elemental impurities.

After selecting the API supplier, which previously has been audited by corporate Hikma compliance

group, it is necessary to send to Hikma PT the most recent DMF/technical package of the API and the

CoA (Certificate of Analysis) which is in compliance with the EU or US guidelines. The price of the API

has to be mentioned for R&D purposes. The necessary quantities for development have to be ordered,

and later on for stability/submission batches.

The TTR is a complete document sent by R&D site at Jordan or USA. It provides a description about

the product that will be submitted, including the description of the analytical studies performed with its

results and the final conclusion: an ideal manufacturing process defined after all the studies. This

document also describes a risk assessment and presents the results, the critical parameters are

mentioned as are the strategies of controlling them.

The PV protocol is developed by technical services Portugal and is based on the TTR. It starts with a

brief description of the product including the strength, the presentation, and where it was developed. It

refers also to the purpose of the protocol: how to produce the necessary batches for validation.

24

Regarding the manufacturing process designed by R&D, several adjustments are made and described

in the protocol, in accordance to the available equipment and means at Hikma PT site. In this document,

each activity is explained in detail as are the sampling requirements needed to complete the evaluation.

A sampling log covering the full testing is included in the protocol appendices. The parameters that are

verified or established during the study, such as specific processing time limits, will be added to the

Master Batch Record (MBR) following the conclusion of the PV. (Marques 2015)

3.1. Analytical method development

The Analytical Research (AR) department in Hikma PT evaluates the validation of the analytical methods

developed in R&D for the API and excipients. United States Pharmacopeia (USP) and European

Pharmacopeia (EP) are taken into account in the transfer. All the methods required for in-process and

finished product testing should be developed and validated/co-validated before the manufacture of the

product. The validated analytical methods are used during the testing of the submission batches and if

everything works as expected, they will continue to be used after the product is approved by the

authorities for commercialization.

The R&D team has to provide a list of the evaluated raw-materials to the technical services department

so that they can be ordered in Portugal by the logistic team.

As part of the development work, Hikma R&D has to provide technical services with information required

by validation department to evaluate if the new product fits within the existing cleaning validation matrix.

A cleaning method should be developed and validated taken into consideration the cleaning limit

established by the manufacturing site. (Marques 2015)

3.2. Product development

After developing a new product in the R&D site, a TTR is written as mentioned. This document includes

information regarding the following aspects described in Table 3.

25

Table 3 – Information available in the TTR

Raw Materials and

API Product Formulation

Manufacturing

process Compounding

Classification of raw-

material and physical

properties of the API

Intellectual property

assessment Equipment description

Product formula and

calculations for the

quantities to weigh

Analysis and

specifications

Excipient supplier,

analysis and

specification

Physiochemical

compatibility studies

with contact surfaces

Sequence of material

addition

Degradation study

towards pH, light,

humidity and oxygen

Free inorganic ions

that are part of the

drug molecule

Results for light,

sensitivity, viscosity

and conductivity

Process temperature

and mixing

time/ranges

Toxicity evaluation and

impurity profile studies Potential degradants

Maximum limit for

dissolved oxygen on

the bulk solution

Dissolution and

appearance of the

product per step

Retest times and shelf-

life Comparison between

Hikma product and

other generics on the

market

Maximum holding

times

Equations, mass

balance, pH ranges,

solubility with

temperature

Material Safety Data

Sheet

Terminal sterilization

evaluation

Sensitivity of product

to atmospheric gases

3.3. Submission batches manufacture

Before submitting a batch, technical services Portugal has access to API DMF, audit report and letter of

access. A list of the originator and all the generics on the market, including strengths, presentations and

container closure is also necessary. The technology transfer or development report has to be sent to

tech-services together with the analytical methods transfer for the AR department. If applicable the

microbiology analytical methods are also included.

Hikma PT is responsible for defining the batch size and perform the scale-up of the batches for

validation. It should follow the TTR or Process Development Report (PDR) sent by R&D, in a way that

26

it mimics as much as possible the industrial scale, using the same equipment when possible. The PV

protocol is prepared by technical services and the submission MBRs regarding the production of the

batches too. The PV report includes a side by side evaluation between submitted batch scale and

proposed batch scale.

Before manufacturing the submission batch or batches the analytical methods validation for the new

product have to be completed by the AR department. All conditions used at the production line where

the validation takes place have to be conform. The containers, closures, filters and equipment have to

be sterilized and in cases where the processes are aseptic a media fill validation is necessary. If

applicable terminal sterilization validation for drug product has to be done too.

After manufacturing the submission batches they are placed in stability for further studies, the product

is then submitted to the regulatory authorities. After their evaluation, it happens that deficiency letters

are sent to improve the product (usually in terms of analytical methods adjustments or process

manufacturing). If no adversities are found, eventually, the product can be manufactured for the first

time in accordance to the commercial scale-up, and approved for routine commercial manufacturing.

(Marques 2015)

3.4. Industrial facility and production lines, line 5

operation

Hikma is a multinational pharmaceutical group with a Jordan background, it has a dimension at a global

scale and is specialized in production and commercialization of branded and non-branded generic and

in-licensed pharmaceutical products for exclusive hospital use. Hikma Pharmaceuticals operations span

twenty-nine manufacturing plants in eleven countries: US, Portugal, Italy, Jordan, Saudi Arabia, Algeria,

Germany, Egypt, Morocco, Tunisia and Sudan.

Portugal is the centre of production in the segment of injectables within the group. The industry is located

in Sintra and is divided in two separate industries, one of them is composed by eight production lines

for injectable manufacturing and the other has three lines for the production of antibiotics in powder

form. At the moment, a new industry is in construction specifically for the production of oncologic drugs.

(Hikma website 2017)

Table 4 resumes the type of products manufactured in each line.

27

Table 4 – Hikma’s facilities, production lines, product presentations and type of products

Facilities Production lines Product presentation Type of products

Hikma 1

1 Lyophilized

Anti-infectives

Cardiovascular and diabetes

Controlled substances

Gastro-intestinal

Central nervous system

Respiratory

Antibiotics

2 Vials

3 Ampoules

4 Bags

5 Vials

7 Bags

9 Lyophilized

10 Lyophilized

Hikma 2

1

Powders Antibiotics 2

3

New Hikma To be defined To be defined Oncological

The two products which will be presented as examples of technology transfers and PVs were produced

on line 5. This line is specific for the production of solutions in vials to be used for injections (hospital

use).

Line 5 usually uses a 1000L and 2000L stainless steel tank for the compounding. It happens a lot that

when the mixture has finished the solution needs to be put on hold because another product has to be

compounded, in these situations the second tank is used as a transference tank if applicable. However,

for the two examples which will be presented, since they were submission batches, smaller tanks were

used.

The API and the excipients are introduced in the compounding vessel together with the WFI which

occupies the major area. A propeller is installed in the tank and assures the complete homogenization

of the solution.

28

There are four machines which work simultaneously when the bulk solution leaves the tank: the vials

washing machine, depyrogenation tunnel, filling machine and capper machine.

The tanks have an outlet where the tubbing fits, the tube carries the bulk solution until the filling machine.

At this point in the manufacturing process the formulated drug product enters the Class A clean room. It

remains under these conditions until the product is filled into the vails and stoppered.

As soon as the bulk solution enters de filling machine they pass by filters which sterilize the product.

There are four primary types of filters used in the parenteral and biopharmaceutical industry, the type of

filter chosen depends on the type of material to be removed. The types of filters include the following:

- Clarifying filters – for large particles;

- Microfilters – suitable for bacteria and yeasts (used for injectable drug products);

- Ultrafilters – used to retain viruses;

- Nanofilters – for small organic compounds and ions.

The injectable drug industry uses microfilters to remove particles in the 0.1 to 10 µm size range from

the formulated drug product. Many types of membranes are available in this pore size range for different

types of formulations, including water based formulations (hydrophilic) and solvent based formulations

(hydrophobic). The development site conducts studies for filter compatibility in order to determine the

correct filter and filter surface area for the particular product. For most parenteral products, a hydrophilic

filter is used, it can be made of: cellulose acetate, cellulose nitrate, regenerated cellulose, modified

regenerated cellulose, polyamide (nylon), polycarbonate, polyethersulfone, polysulfone or

polyvinylidene difluoride (PVDF).

The filters are available as either flat disks or as cartridge filters, which significantly increase the filter

surface area when extremely large volumes need to be filtered. To ensure that the filter membrane is

completely intact with no holes, integrity testing is performed both before and after filtering the product.

This is accomplished through a process known as bubble point testing, a non-destructive integrity test

measuring diffusive flow or water intrusion over the filter membrane. As soon as the filter is chosen and

installed, the solution can be sterilized. If a product is a suspension or large particle-sized emulsion it

cannot be sterile filtered and has to be aseptically formulated, all components are pre-sterilized

individually and then brought together in a sterile environment.

On line 5 the filters which are installed in the filling machine usually have a pore size of 0.2 µm. In the

cases when products need a pre-filter, the size used is normally 1.2 µm. Once the product has been

filtered into a sterile filling container and the filter passes the post fill integrity test, it is ready to fill into

its primary container which was previously washed and pre-sterilized in the depyrogenation tunnel.

Sterile tubing is placed into the tank with the sterile solution, which leads first to pumps and then to filling

needles. The needles descend into the vials and slowly rise as the required amount of product is

dispensed. This method of filling minimizes splashing of product on the sides of the container.

The type of pump used depends on the type of product being filled: piston or peristaltic are used for

either liquids or gases, gravity is used for solids and liquids.

29

The product is generally filled into glass vials using one of three main methods:

- Volumetric – a fixed volume is added;

- Time/pressure – a fixed pressure is administered over a certain amount of time;

- Net weight – each container is weighed while being filled.

For the examples which will be presented the types of pumps chosen were piston and the products were

filled using the volumetric method.

Once the vials have been filled, they travel down the filling line to have pre-sterilized stoppers inserted.

Line 5 has an auto-clave which sterilizes equipment parts and the rubber stoppers. After the stoppering,

the vials leave the sterile room (class A) with laminar flux, and are led to the clean area to be capsulated

in the capper machine.

Caps are comprised of a plastic flip-off and an aluminium skirt, they are used to secure the stopper in

the neck of the vial to prevent the stopper from coming out either over time or during handling. The caps

are loosely placed on the top of each vial, then travel to the crimping station where rotating blades crimp

the bottom of the aluminium skirt around the lip on the neck of the vial, producing a tight fit that locks

the stopper into the neck of the vial. At the time of use the flip-off is removed, exposing the top of the

stopper which is pierced with a needle to remove the content inside the vial. After capsulation, the vials

are ready for inspection and final packaging.

30

31

4. Practical examples: Technology

transfer of injectable drugs on line 5

4.1. Product A

4.1.1. Description

Injection A is a new product developed by West-Ward Pharmaceuticals (Hikma USA) – at the Bedford

development centre, where the R&D is performed for this country. It is an Abbreviated New Drug

Application (ANDA) of a generic transferred to the Hikma PT manufacturing site. This product is

indicated for the treatment of heart complications, it is in a liquid form, aseptically filled however non-

terminally sterilized. Table 5 summarizes the information regarding the product and manufactured

submissions batches.

Table 5 – Product A presentation of the submission batches

Product/

presentation

Target fill

volume

Bulk

concentration

Number of

batches

Batch size Vial size

Injection A

(1 mL/vial)

1.20 mL

0.2 mg/mL

2 50 L (100 L tank) 6 mL vial,

neck 13 mm 1 30 L (30 L tank)

Injection A

(5 mL/vial)

5.60 mL

2 50 L (100 L tank) 2 mL vial,

neck 13 mm 1 30 L (30 L tank)

Since the product is an ANDA it was compared to the Referenced Listed Drug (RLD) in terms of

analytical results. However, the data provided from R&D site was subjected to slight alterations in

accordance to the manufacturing conditions available at Hikma PT manufacturing site, this information

will be further gathered in the product development report.

Injection A has five main components, including the API. For the pH adjustment 1N sodium hydroxide

solution and 1N hydrochloric acid are used. To complete the volume, Water for Injection (WFI) is added.

The active raw material has some special precautions when handling, since it is hygroscopic. The

production conditions requires an inert atmosphere (nitrogen). Storage conditions is a tight, light

resistant container away from moisture at below 25ºC.

32

4.1.2. Materials and compatibility

In order to choose the container closure system, a study of material compatibility was performed. For

the 1 mL/vial presentation a clear type I tubular vial with 2 mL capacity was chosen, with a 13mm rubber

stopper and an aluminium capsule with a flip-off. For the 5 mL/vial the capacity of the vial is 6 mL, the

rubber stopper and the aluminium capsule are the same.

Because of the product components the probability of delamination of the vial increases, the metal ions

in the glass can be leached. Additional studies were performed in order to improve this medium risk

factor. The responsible raw-materials for the delamination were stored in the vials for a defined time in

different concentrations and temperatures. The results showed that higher temperatures increased the

levels of elements especially on 5 mL filled vials, the concentration did not have influence on the

problem. However, R&D suggested additional tests during stability studies and submission batches in

order to evaluate the risk of glass delamination.

The rubber stoppers may contain extractables, however there is no damage for the product even though

it is an acidic injectable. The protection comes from the contact surface of the rubber which is coated

with resin free from metal ions.

To assure that the product is compatible with all contact materials, the production tools were also

studied, including the tubing where the mixture will pass through and the stainless steel from the tanks.

The tubbing was filled with bulk solution and stored at room temperature under yellow lightning for 48

hours. In a stainless-steel vessel, a batch of bulk holding solution was maintained also for 48 hours at

room temperature, the vessel was sealed with a nitrogen overlay. No significant changes were verified,

the data showed that the drug product is compatible with the tubing and stainless steel. However, with

an extra study of increased holding time of the bulk solution in the vessel, a slightly growth in the iron

level was verified. This means that leaching from the stainless steel can happen, due to the composition

of the product in contact with the vessel for a long time. This aspect will be taken into consideration

during submission batches.

Filters are used during the passage of the solution between tubbing from the tanks to the filling

department. A study was performed to determine among other factors, the filters acceptable size. The

results show that the drug substance does not absorb to the filter membrane in the initial stage of bulk

solution filtration. Regarding extractables and leachables from the filters, no significant risk was

detected. The filter was shown to have an effective microbial retention, with no contaminating

microorganisms downstream.

The filter which was chosen and validated after the tests has the following characteristics: a PVDF

membrane with a safety factor of 1.8 and a maximum achievable flow rate of 60L/min. The minimum

flow rate of 38mL/min with a filter membrane area of 13.8cm2 results in a validated flow rate of 2.8

mL/min/cm2. (Colvard 2016)

33

4.1.3. Product and process precautions

This product needs some precautions with light, compounding and filling must be performed under

yellow light. The containers used must be opaque. However, studies conducted under white light proved

that it is acceptable and has no harm on the product for a limit of seven days. Regarding oxygen, nitrogen

protection is required to protect the product from its high sensitivity to oxygen. The acceptable

temperature range is between 20°C and 25°C.

The ideal pH range (3.5-4.0) was chosen in accordance to the results of the tests performed, certain pH

values increased the impurities. The increase in oxygen in the vials also intensified the pH value, which

meant that the impurities also grew. It was noticed that in-process solution changes to pink as impurities

increase, so a yellowish colour is the acceptable if it is within the limits.

To determine if the product should be terminally sterilized or aseptically filled a heat sterilization was

performed. When terminally sterilized the assay results were critically impacted, which means that the

product is affected by excessive heat from sterilization.

In Table 6 it is possible to find a resume of the most important process conditions for product A.

Table 6 – Process conditions for product A

Under nitrogen overlaying Yes (during compounding and filling)

Tubing to be used Teflon

Filling pumps to be used Piston

Terminal Sterilization No

Use pre-filter No

Maximum bulk holding time 48h

Maximum contact time of filter with product 48h

Product sensitive to light Yes (light protection during compounding and

filling activities by using yellow lights)

Product sensitive to oxygen Yes

Product sensitive to heat sources No

Finished product storage conditions Store at 20ºC - 25ºC

34

4.1.4. Manufacturing process

The production process for product A is described on Figure 1. The mixing times required for dissolution

were defined as ten minutes for the API and excipients, and five minutes for the acid/base addition to

adjust the pH. The packaging includes a pack of twenty-five vials with one insert for the 1 mL/vial

presentation and ten vials one insert for the 5 mL/vial presentation.

Figure 1 – Flow chart of the manufacturing process of product A

4.1.5. Risk assessment

The batches manufactured for validation used qualified facilities, systems and equipment, and were

tested using validated analytical methods. All raw-materials and primary package materials were

analysed and released before production. On line 5, the manufacturing process of aseptically filled

products was evaluated through media fill. These measures reduce the probability of hazards. (Marques

2017)

The critical parameters identified as potential process failure causes were evaluated based on risk

management tools – Failure Mode Effect Analysis (FMEA). Detailed description of CPPs, cause of the

failure, potential effect on the system, severity, probability and control is presented in Annex A – Risk

management tools (FMEA) for product A. Severity and occurrence/probability are categorized in level 1

Add initial WFI 60% of final

weight

Sparge WFI with nitrogen

Begin mixing and maintain temperature

Discontinue purge. Add excipient 1

Rinse with WFI, reinstate purge and mix

Discontinue purge. Add excipient 2

Rinse with WFI, reinstate purge and mix

Add excipient 3 and WFI.

Add to vessel

Rinse solution container with WFI and mix

Prepare a solution with API and WFI. Add to vessel

Rinse solution container with WFI and mix

Discontinue purge. Add excipient 4

Rinse with WFI, reinstate purge and mix

Check and adjust pH with 1N NaOH or

1N HCl

Complete solution with WFI and mix

Check pH

Place a nitrogen

headspace on bulk solution

Filter

Fill solution into vials,

stopper and seal

35

(low), level 2 (medium), level 3 (high). Detection follows the opposite scale: level 3 (low), level 2

(medium), level 1 (high). Severity x occurrence x detection is categorized as: critical – 12, 18 and 27;

medium – 6, 8 and 9; low – 1, 2, 3 and 4.

It was concluded that the calculations for API and excipients are very critical for the process having a

high severity risk. A mistake can cause low or high assay and result in batch failure. As a control, the

calculations are double verified. Other failures that can also result in higher or lower concentration

results include errors in the final adjustment of the solution with WFI and incomplete dissolution of API

or excipients. Controls can consist of pre-dissolution before addition of the materials to the preparation

tank to assure total homogenization and calibration of the balances before compounding to guarantee

that the final volume is correct.

4.1.6. Process evaluation activities and results

In order to validate the process for injection A at Hikma Portugal site, three submission batches were

produced for the 1 mL/vial presentation and other three batches were manufactured for the 5 mL/vial

presentation. Different activities were performed during the manufacturing of these batches, mainly in

the important steps of the process, to evaluate quality consistency and so that it could be possible to

adjust certain parameters of the process, for example mixing times. Annex B – Sampling scheme for

product A includes the samples necessary to be taken at the critical process steps to perform the

analytical studies.

Effects of mixing time and speed on the quality of the bulk solution

The dissolution/homogenization of the bulk solution should be complete and uniform. The range of

mixing speeds and agitation times to be used were evaluated during compounding to assure that

complete dissolution occurs under all approved compounding conditions. The values were chosen in

accordance to experience, and were adjusted during the manufacture to achieve proper dissolution. The

speeds were defined as 200-300 rpm for the 30 L batches and 300-400 rpm for the 50 L batches.

Samples were collected from the top and bottom of the compounding vessel of all batches for

physiochemical analysis to ensure uniformity of the compounded solution, microbiological analysis were

also performed. The results were conform.

Effects of elapsed compounding (bulk holding time)

The bulk solution for the volume of 30L or 50L is manufactured in a stainless-steel compounding tank.

However, it might be necessary to transfer the bulk into another vessel for scheduling production needs.

For this reason, an evaluation of the maximum bulk holding time in a temporary tank is needed.

36

Taken into consideration that all batches have the same bulk concentration, the product is expected to

behave similarly during holding times studies. Therefore, the data generated from the evaluation of one

submission batch was considered applicable to both presentations.

The maximum bulk holding time was studied in a sample of bulk solution left in the preparation tank up

to 48 hours at room temperature. Samples were taken from the bottom of the tank following an elapsed

time period of 24 hours and 48 hours from the end of compounding for physiochemical and

microbiological testing. Results were compared with time zero (0 hours) bulk results. All samples met

in-process physiochemical and microbiological specifications which means that the product can be left

during 48 hours on hold inside the tank.

Effects of initial set up (dead volume) and line stoppages

The purpose of the “dead volume” study (one batch of each presentation tested) is to determine the

minimum volume of solution necessary to be discarded before having manufactured product in-

specification to be kept. The first units are discarded as part of initial line set-up. The procedure includes

priming the pumps and purging the system. The first and last vials filled from a 1st stage were collected

and tested. Since they were not within specifications or aligned with time zero bulk results, which

happens frequently, a 2nd and 3rd stage was performed in the same way. At the 3rd stage the results were

conform.

The line stoppage test is performed on one batch of each presentation, in order to establish acceptable

duration of stoppages, and to determine the minimum quantity of vials to be discarded before restarting

the filling. After two hours of line stoppage, 300mL were filled into vials, the first and last vials of three

stages were collected. Physicochemical tests were performed. It was only necessary to pass to stage 2

which means that the quantity to be discarded before restarting the filling corresponds to the volume

used to fill the vials from the first two stages.

Filling machine speed

To define the ideal speed for the filling machine the maximum and minimum speed was tested during

the filling of the vials. The analysis performed for the control was volume variation of the vials.

No problems were detected. Table 7 shows the requirements for the volumes in the vials of the two

presentations for this product.

37

Table 7 – Volume specifications for filling of product A

Presentation

Individual minimum

(mL)

Average (mL) Individual maximum

(mL)

Minimum

action

Minimum

alert

Minimum Target Maximum Maximum

action

Maximum

alert

1 mL/vial 1.10 1.10 1.15 1.20 1.25 1.30 1.35

5 mL/vial 5.30 5.45 5.50 5.60 5.70 5.75 5.80

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

Filled vials after capping were collected for physicochemical analysis from the beginning, middle and

end of the filling process for all batches. Beginning of the filling is considered the point in which the

filtered bulk is no longer discarded and line flush and set up activities have been completed. At every

hour, visible particles and oxygen headspace were checked.

There were no detected physicochemical or particle deviations, however oxygen Out Of Specification

(OOS) were confirmed.

Vial inspection

After capping the vials, they were all inspected visually to detect defects and particles, using a particle

automatic inspection machine. Although the product is sensitive to light, white light is acceptable for the

inspection. All results were in-specification.

4.1.7. Problems during submission batches

As mentioned, injection A is an oxygen sensitive product filled with nitrogen flushing in the vials

headspace. The oxygen headspace is controlled hourly during the manufacturing. The limit is Not More

Than (NMT) 2.5% for 1 mL/vial presentation and NMT 5.0% for 5 mL/vial presentation as per relevant

MBRs and SOPs.

Three batches for the 5 mL/vial presentation were produced with oxygen values found to be very above

the limit of 5.0%. As per the TTR sent by West-Ward Pharmaceuticals, oxygen headspace was also

challenged during R&D stages.

In order to determine the root cause and to investigate the deviation detected, the root cause analysis

methodology was followed as shown on Figure 2.

38

Figure 2 – Root cause map for oxygen headspace OOS for 5 mL/vial presentation batch

The container closure 6 mL is a worst case for the oxygen headspace measurement in the lighthouse

system due to its limited free space between the neck of the vials and the solution within the vial. The

difficulty faced during the oxygen headspace analysis was considered the main cause for the results

obtained. Moreover, several oxygen headspace results were obtained at 0%, the technicians did not

recognize these values as abnormal since line 5 is known to be highly capable. A human error is

considered in this situation in addition to the inadequacy of the equipment towards the 6 mL vials. The

followed procedures and the filling machine capability were not considered to be involved with the

problem.

A decision was made to reject the batches since R&D stability data demonstrated that the product does

not support a higher oxygen headspace specification.

Another three batches were manufactured, this time for the 1 mL/vial presentation. During filling start-

up of the first two batches, vials with oxygen headspace out of the limits were obtained. Adjustments

were done to the needle holders/supports of the nitrogen flushing part and to the nitrogen flows to

improve the oxygen headspace levels until results were conforming. During in-process control vials with

oxygen headspace OOS were still found, consequently the batches were placed on retention. Figure 3

presents the root cause map for the deviated results.

39

Figure 3 - Root cause map for oxygen headspace OOS for 1 mL/vial presentation batch

After further investigation, using new stability data provided by the USA site, a worst-case oxygen

headspace of 3.5% might be considered acceptable in exceptional situations.

However, filling machine from line 5 is highly capable of achieving low oxygen headspace levels.

Qualification studies were conducted before and demonstrated a highly capable process for 2 ml vials.

For this reason, no problems were anticipated with the filling of injection A. The capability of the machine

is not considered the root cause.

It could be considered that the procedures were not clear and affected the system, however this

hypothesis was ruled out because all SOPs and MBR were followed correctly. There was only a slight

alteration on the nitrogen flow to a value out of the range specified in the MBR, nevertheless the

adjustment during filling start-up was done to meet the required oxygen headspace and was considered

to have no impact.

The most probable root cause is equipment related because one of the needle holder/support got broken

a month before the mentioned batches were produced. The holder was replaced by a new one, since

then it is probable that it has been misaligned.

After disassembling the nitrogen system and re-checking all supports one by one, the misalignment was

fixed and the 3rd batch of the campaign was filled without any observations and with consistent oxygen

headspace results.

The statistical analysis of the data generated demonstrated a highly capable process for an oxygen

headspace limit of NMT 3.5% - this new specification was applied. For the 1st and 2nd batch produced,

the trays where the low amount of outliers were found were rejected.

40

Following the completeness of the 1 mL/vial validation, three batches of the 5 mL/vial were

manufactured once again. A new lighthouse system was obtained and optimized for the reading of 6 mL

vials, with improved results and provided less analytical error. The implemented alterations were

successful. A slight problem was detected during production of one of the batches, the nitrogen flushing

part moved from its original fitting position, which had impact on some results. However, not significant.

As a conclusion, frequent maintenance and substitute of parts of the equipment must be made so that

problems as the mentioned do not happen.

4.2. Product B

4.2.1. Description

Injection B is an aseptically filled product for administration during heart surgeries, developed by

Bedford. The API of this product is an isolated protein from salmon. It has only one excipient (excipient

A) that adds to the API, sulfuric acid 0.1N and dibasic sodium phosphate 0.1N are used to adjust the

pH. WFI completes the quantity of the solution.

Table 8 summarizes the information regarding the product and manufactured submissions batches.

Table 8 – Product B presentation of the submission batches

Product/

presentation

Target fill

volume

Bulk

concentration

Number of

batches

Batch size Vial size

Injection B (5

mL/vial) 5.60 mL

10 mg/mL

1 50 L

(100 L tank)

6 mL vial,

neck 13 mm

Injection B (25

mL/vial) 26.30 mL 1

200 L

(200 L tank)

30 mL vial,

neck 20 mm

Injection B is an example of a product which is not “new”, it has been submitted in 2007 by Bedford to

the regulatory authorities, however it was not approved. A major deficiency letter related to the analytical

methods of the API and the final drug product was sent by FDA.

Bedford sent the amended application of the drug, to remove chemistry deficiencies as per the agencies

correspondence. As a response, FDA sent a minor deficiency letter in 2010 regarding some extra small

modifications.

When the characterization studies were conducted it was observed that the proposed ANDA drug

demonstrated foaming when shaken, greater than seen in the RLD. A slight foaming is expected since

the API is a peptide, however the observed foaming was considered unacceptable and undesired. The

41

cause was attributed to the manufacture of the drug substance (reliable). The biopharmaceutical

“reliable” agreed to reduce fatty acid constituents that were identified as the most likely source of

foaming in the formulated drug product. (Madieh 2015)

Bedford developed a TTR so that injection B could be transferred to Portugal manufacturing site,

however the analytical alterations solicited by FDA would have to be conducted in order to get the

product approved. AR department completed the required analytical validation tests and manufactured

the product for validation.

4.2.2. Product and process precautions

This product requires some precautions. Table 9 describes the most important process conditions. In

terms of temperature the API is stored between 2ºC and 8ºC, before opening the container it is

necessary to keep it at room temperature approximately one hour. The product is formulated between

21ºC-23ºC, however it is not necessary to maintain temperature control throughout filtration and filling.

The final drug product is stored between 20ºC to 25ºC.

Injection B is a non-cytotoxic, non-genotoxic product assigned with a hazard category of 3A. When

handling drug substance and product during formulation it is necessary to use a Personal Protective

Equipment (PPE) such as eye protection, gloves and a half mask cartridge respirator. (Madieh 2015)

Table 9 - Process conditions for product B

Under nitrogen overlaying No

Tubbing to be used Teflon

Use pre-filter No

Filter for product filtration Yes

Integrity test for the filter Bubble point > 3180 bar

Filling pumps to be used Piston

Terminal sterilization No

Maximum contact time of filter with product 16h

Product sensitive to: light, oxygen, heat No

Finished product storage conditions Store at 20ºC – 25ºC

42

4.2.3. Manufacturing process

One submission batch of each presentation was produced in Hikma Portugal on line 5 to validate the

process in the new manufacturing site. A 50L batch was manufactured to validate the 5 mL/vial

presentation and a 200L batch was manufactured to validate the 25 mL/vial presentation. Both with the

same bulk concentration (10mg/mL).

The manufacturing process includes the compounding of the bulk solution formula followed by filtration

through a 0.2 µm membrane filter. The filters are integrity tested before sterilization, before filtration and

after filling. The pre-sterilization integrity test is conducted with WFI, pre-filtration and post-filling are

conducted with drug product. Microbial retention, microbial viability, bubble point, filter compatibility and

filtration time duration were included in the filter validation by R&D site. Leachables and extractables

were validated at the manufacturing site.

The filtration is followed by aseptic filing into the respective vials (6 mL and 30 mL vials), stoppering,

capping and inspection. The packaging includes ten labelled vials and one insert per coded shelf carton

for the 5 mL/vial presentation and one labelled vial with one insert per coded unit carton for the 25

mL/vial presentation. Figure 4 presents a flow chart with each step of the process.

Figure 4 – Flow chart of the manufacturing process of product B

Add 80% WFI of the final weight of the vessel. Adjust to temperature

Add excipient A. Rinse weighing container with

WFI and add to the vessel. Mix

Add API. Rinse weighing container with WFI and add to the vessel. Mix

Check pH 6.3-6.7. If pH > 6.7 adjust with 0.1N

sulfuric acid solution, if pH < 6.3 adjust with

0.01N Sodium Phosphate Dibasic

Solution. Mix

Discontinue mixing, seal vessel and transfer to

filtration area

Filling of solution into vials, stopper and

cappsulate.

43

4.2.4. Risk assessment

The TTR includes the process map for the manufacture of injection B, with the material attributes,

process parameters and quality attributes for each unit operation listed in sequence of the manufacturing

process as shown on Figure 5. This information can be used to define the specific CQAs, CMAs and

CPPs.

The CPPs are verified during the manufacturing process to ensure the manufactured product

consistently meets the desired quality specifications. The critical product quality attributes are

considered critical since they can be impacted by the formulation and manufacturing process variables

– the CQAs include the product appearance, assay and degradation products, bacterial endotoxin level,

product sterility and sub-visible particulate matter. Identity, residual solvents and microbial limits are less

critical. These CQAs are ensured through a good pharmaceutical quality system and an effective control

strategy. (Marques 2016).

Figure 5 – Process map for process parameters, quality and material attributes for product B (Madieh 2015)

API

44

The parameters identified as potential process evaluation failures were evaluated based on risk

assessment: Annex C – Risk management tools (FMEA) for product B

The API calculations and weighing were considered critical stages of the process as well as the

dissolution of the bulk solution. Since the product is not light, heat or oxygen sensitive, the packaging

step has a low severity risk.

The side-by-side evaluation regarding the risk of technology transfer from Bedford to Hikma PT is

presented on Table 14 and Table 15, which can be found on Annex E - Risk assessment for the

technology transfer of product B from Bedford to Hikma. This analysis was essential for the transfer to

the new manufacturing conditions at the new site and for the scale-up which will be presented further

on.

4.2.5. Process evaluation activities and results

The following activities were performed during the production of both validation batches in order to define

certain process details and to guarantee that the manufacturing process is suitable for the production of

this injection at Hikma facilities. The sampling scheme for the manufacturing process of product B can

be found on Annex D – Sampling scheme for product B.

Effects of mixing time and speed on the quality of the compounded bulk

solution

The range of mixing speeds and processing durations to be used were evaluated during compounding

to assure that complete dissolution occurred. The speeds were defined as 400 rpm for the 5 mL/vial

presentation and 1200 rpm for the 25 mL/vial presentation.

After the final mixing step was complete, samples from the top and bottom of the compounding vessel

were taken for physiochemical analysis (in plastic sealed bottles) and only from the bottom for

microbiological analysis (in pyrogen free bottles). The samples met in-process physiochemical and

microbiological specifications per the in-process bulk analysis.

Effects of elapsed compounding (bulk holding time)

The bulk solution of one batch presentation was evaluated for the maximum bulk holding time in the

preparation tank. Since all batches have the same bulk concentration, the product is expected to behave

similarly during holding times studies.

At West-Ward Pharmaceuticals the validated maximum holding time from the end of WFI addition to the

tank until the end of filling is 48h at room temperature. For the validation in Hikma PT on line 5, samples

were taken from the bottom of the tank after 24 hours, 48 hours and 72 hours from the end of

compounding for physiochemical testing (in glass vials stoppered and capped or plastic bottles properly

45

sealed) and following 48 hours and 72 hours for microbiological testing (in sterile pyrogen free bottles).

All samples met in-process physiochemical and microbiological specifications. The maximum validated

time for the bulk solution to be left in the tank was defined as 72 hours.

Effects of initial set up (dead volume) and line stoppages

Samples were collected from each batch before starting the filling, after line set up, so that line

adsorption effect could be evaluated.

The objective of the dead volume sampling and correspondent tests was to determine the quantity of

volume to be discarded in order to meet specifications, as explained also for injection A. The system

was purged with 1000 mL following the sampling of the first and last vials of stage 1. Since the results

were within specifications however not aligned with time 0 hours bulk solution, stage 2 and 3 were also

analysed. No OOS was verified, however an extra purge can be defined so that the results approximate

more with the results of 0 hours bulk solution.

To establish an acceptable line stoppage duration and to determine the minimum quantity of vials

needed to be discarded before restarting the filling, a line stoppage of two hours was performed.

Samples were collected for analysis, the first and last vials of the first stage were tested. The results

were within specifications, therefore it was not necessary to analyse the 2nd stage. Two hours was

validated as the maximum line stoppage duration.

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

The filtration and the filling uniformity were evaluated in both submission batches. Filled units were

collected for physicochemical analysis from the beginning, middle and end of the filling process, after

capping. The volumes obtained were within the specifications presented in Table 10 for the filling of

vials. No deviations were detected which means that the defined filtration and filling conditions are

suitable for the process. At every hour visible particles were also analysed, the results were conform.

Table 10 - Volume specifications for filling of product B

Presentation

Individual minimum

(mL)

Average (mL) Individual maximum

(mL)

Minimum

action

Minimum

alert

Minimum Target Maximum Maximum

action

Maximum

alert

5 mL/vial 5.30 5.45 5.50 5.60 5.70 5.80 5.75

25 mL/vial 25.75 25.85 26.10 26.30 26.50 27.20 26.85

46

Vials inspection

All the vials after being capped were inspected using the particle automatic inspection machine, the light

box will be used for stability samples. All results were conform.

4.2.6. Scale-up

At the moment, the data compilation report resulting from the validation is being developed. The defined

parameters for the compounding of the 5 mL/vial presentation batch includes a 10 minute dissolution

time after the addition of excipient A as defined in the MBR, the agitation speed was challenged and

established to 400 rpm. After the addition of the API only 1 hour was necessary for complete dissolution,

even though the MBR had 180 minutes registered. The agitation speed was also defined as 400 rpm. In

order to adjust the pH to the range 6.3-6.7, 0.1N sulfuric acid was added (8mL) – the optimal value of

6.5 was achieved. For the final homogenization, the defined 10 minutes of the MBR was followed and

sufficient. Agitation speed of 400rpm continued to be used. The total compounding time was 7 hours

and 24 minutes.

Regarding the manufacturing of the 25 mL/vial presentation batch, a 200 L preparation tank was used

as established on the MBR. Dissolution time after excipient A addition took 10 minutes, the information

registered on the record was a minimum of 10 minutes. No information about the agitation speed was

given, however after challenging the speeds and according to knowledge from production of other similar

injections, 1200 rpm was defined for all stages. After adding the API the same situation happen

compared to the 5 mL/vial batch, after 1 hour total dissolution was achieved, contrarily to the minimum

180 minutes defined on the MBR. To adjust the pH, 40 mL of 0.1N sulfuric acid was added and a pH of

6.3 was obtained. The final homogenization required a minimum of 10 minutes of mixture, it was

sufficient with only 10 minutes. Compounding time took a total time of 4 hours and 55 minutes.

For both batches, the bulk holding time was tested until 72 hours and was stable. The maximum contact

with the filter was defined as 16 hours in the MBR, however the necessary time for filtration did not

overcome 5 hours. The filling duration was 35 minutes for the 5 mL/vial presentation batch and 3 hours

and 30 minutes for the 25 mL/vial presentation batch – including the 2 hours of line stoppage as indicated

in the protocol. The volume to be purged before filling (dead volume) defined in the MBR was established

as superior to 1000 mL. For the 5 mL/vial presentation it was defined as 192 vials (equivalent to around

1075 ml using a fill volume of 5.60 mL per vial) and for the 25 mL/vial presentation 71 vials were defined,

equivalent to around 1867 mL using a fill volume of 26.30 mL per vial.

Based on the results and parameters obtained for the validated batches, it was possible to plan the

scale-up. The proposed commercial batch sizes were established based on the number of viable vials

obtained on the submission batches. It was decided to slightly increase the proposed batch sizes to

accommodate routine bulk losses and to be within 10 times the theoretical submission batch sizes.

47

Table 11 summarizes the rational used for the definition of the proposed commercial batch size of each

presentation. The risk assessment for product B scale-up can be found on Table 16 and Table 17 in

Annex F – Risk assessment for the increase of batch size from submission scale to a proposed

commercial scale (product B)

Table 11 - Rational for the proposed batch size of product B

Presentation

5 mL/vial

(10 mg/ml)

25 mL/vial

(10 mg/ml)

Su

bm

iss

ion

sc

ale

Batch size 50L (8928 units) 200L (7604 units)

Viable vials

(after visual inspection+ QA samples)

6853 6925

0.1N H2SO4 solution added 8 mL 40 mL

Filling speed used 100%

(≈ 466 vials/min)

100%

(≈ 216 vials/min)

Co

mm

erc

ial

scale

Maximum size (based on viable vials)1 383 L 1821 L

Proposed size 380L 1800L

Target fill volume 5.60 mL 26.30 mL

Number of vials 2 67857 vials 68441 vials

Approximately filling duration 3 ≈ 03h15min ≈ 07h00min

Maximum holding time 72 h

Maximum qualified time for aseptic

filling (by Media Fill)

26h25min

0.1N H2SO4 solution needed 60.8 mL 360 mL

1 Maximum proposed batch size for US market (mL): viable vials x batch average fill volume x 10.

2 Based on the target fill volume.

3 Assuming a filling speed of 350 vials/min for the 6 mL vials and a filling speed of 162 vials/min for the 30 mL vials

(average between the maximum and minimum filling speed on line 5).

48

49

5. Optimization of submission and

commercial batches – PAT

The quality of submission batches is important. If a batch is manufactured correctly and without

unexpected difficulties during validation, the authorities approve the new product without requiring

improvements. Repeating batches which do not have the expected output lead to unpredicted costs by

the manufacturing site and entities involved. Equally, a process which faced several difficulties during

validation and eventually is approved is most likely to become problematic when applied to commercial

production, once again increasing costs and in this case with a probability of a public health risk.

As a solving measure, the development stage has to be effective, the technology transfer if applied must

be done correctly and the control during the submission process has to be well executed. However, it is

important to notice that even if the validated process is suitable for the manufacture of the product and

has a positive result, variability/unpredictability between commercial batches can still occur and face

problems. This can happen because raw materials used to manufacture pharmaceutical products might

vary in their attributes (for example: moisture content, crystal structure etc.) and manufacturing

equipment can have functionality complications, consequently the product is occasionally produced with

variability too. Slight problems during production might seem to have no importance at an initial stage,

nevertheless stability studies occasionally detect unexpected results which lead to withdrawn

medication from the market. If PAT is implemented during validation and for commercial manufacturing,

a wider flexibility for improvements during production is acceptable. FDA is implementing a 21st century

approach to encourage these manufacture progresses.

PAT is intended to support innovation and efficiency in pharmaceutical development, manufacturing,

and quality assurance. It is a mechanism to design, analyse, and control pharmaceutical manufacturing

processes through the measurement of CPPs which affect the CQAs. The concept aims at

understanding the processes by defining their CPPs, and accordingly monitoring them in a timely

manner (preferably in-line or on-line). This way of monitoring is extremely more advantageous than off-

line and at-line analyses characterized by manual sampling followed by discontinuous sample

preparation, measurement and evaluation. Figure 6 shows a scheme with the difference between the

different kinds of sampling. With a consistent control during manufacturing, adjustments can be applied

at real time without a need of extra validations if working within the design space (limits defined for the

parameters). PAT tools can implement a dynamic manufacturing process that compensates for

variability to produce a consistent product, a real-time adjustment is able to save the quality of the

products being manufactured and avoid batch rejection.

50

Figure 6 - Scheme of process monitoring: at-line, on-line, in-line and off-line

A combination of three main PAT tools is essential in order to implement an efficient PAT project:

- Multivariate data acquisition and data analysis software: advanced programs which aid in DOE,

collection of raw data and statistically analysing the data to determine which parameters are

CPPs.

- Process Analytical Chemistry (PAC) tools: in-line and on-line analytical instruments such as

Near-infrared (NIR) spectroscopy, biosensors, raman spectroscopy, fiber optics among others,

which are used to measure parameters that have been defined as critical.

- Continuous improvement and knowledge management tools: paper systems or softwares which

accumulate QC data acquired over time with the aim of defining process weaknesses and

implementing process monitoring initiatives. (FDA 2004)

The practical examples presented – technology transfer and submission of injection A and B, were

subjected to studies in the R&D site in USA as mentioned previously. The research involved the

establishment of the critical quality attributes of the materials and critical process parameters which had

impact on those CQAs. The parameters identified as critical and responsible for potential batch failure

causes were further evaluated based on risk management tools - FMEA. In this point of view, Hikma

Pharmaceuticals follows a QbD approach in order to obtain a better knowledge of the processes and

improve the control of critical steps.

Concerning the example of injection A, the oxygen headspace is one of the most critical parameters of

the process, it is essential to control this CPP in order to guarantee a quality product manufacturing.

OOS results were detected by the In-Process Control (IPC) team when the vials were analysed. To

monitor the oxygen levels, a lighthouse oxygen headspace analyser was used. This equipment can be

considered a PAT tool because it follows the required definitions, it is a process analyser system which

provides non-destructive measurements and delivers information on a critical product attribute.

The instrument utilizes a patented laser absorption technique. Light from a near-infrared diode laser is

tuned to match an internal absorption frequency of the oxygen molecule and passes through the empty

51

container headspace above the product. The amount of laser light absorbed is proportional to the

oxygen concentration in the headspace. Since the sample is not destroyed there are no waste disposal

issues and potentially valuable products can be saved. These characteristics allow multiple

measurements on the same sample enabling the accurate monitoring of the oxidation of a single sample

over time.

Regarding injection B, there were no difficulties during the production of the submission batches. It is a

simple and stable manufacturing process with no concerns about light, oxygen or other parameters.

However, it was noticed that after adding the API to the mixture, a homogenous solution was obtained

after about one hour. The TTR received by Bedford required a three hour duration to obtain the

dissolution of the mixture. The difference was figured out to be related to the API which was improved

by the reliable after the initial studies at the developing site, the foaming verified at Bedford was not

observed in our validation process. Foaming could be interfering with the dissolution rate. As it was

proven, alterations in the materials used have a great impact on the dissolution duration.

It can happen that between commercial batches the raw-materials can present slight alterations or the

equipment can face problems (the propeller in the compounding tank can have difficulties with its

rotation for example), these differences can have an effect on the time needed for complete solution

homogenization. If a one hour duration is always followed the product quality can occasionally be in risk.

Taking into consideration the variability/unpredictability that can occur, an implementation of a PAT tool

such as a NIR probe in the compounding tank can help controlling the dissolution and assure a

consistent quality product output.

The NIR is an analytical technique implemented when an industrial process needs a monitoring system

in real time for the mixing steps. NIR spectroscopy is a precise, fast, mechanically simple and non-

destructive technique that studies the interaction of infrared radiation with matter. This PAT tool

associated to chemometrics techniques, enables the construction of models to monitor the mixture

process. (Rodrigues, 2015)

52

53

6. Conclusion

6.1. Contributions

The literature review presented provides information about technology transfers and PVs. Technology

transfers in the pharmaceutical industry are described as movements of technology from drug discovery

to product development, clinical trials or full-scale commercialization. It can also be the process by which

a manufacturing site makes its technology available to another production site that will exploit the

technology. Appropriate technology transfer is essential to upgrade the quality of design to be the quality

of product.

In order to obtain a successful project, it is necessary to implement an effective training for all involved

in the new process, including production operators and regulatory team members which will prepare the

submission. The information sent by the transferring site is also very important, so that the

understanding of the technology is clear and well implemented.

PVs are considered a mean of proving effectiveness, by assuring that a process operated within

established parameters, can perform effectively and reproducibly to produce a medicinal product

meeting its predetermined specifications and quality attributes.

Before validating a pharmaceutical manufacturing process and defining a commercial process, the R&D

team creates the product at a smaller scale to gain knowledge about the process/product. The first

studies start at a laboratory scale, following pilot batches and finally production scale batches.

The first step of a technology transfer between Hikma’s R&D site and the manufacturing site of Hikma

pharmaceuticals PT is the selection of the product. Hikma PT is responsible for defining the batch size

and perform the scale-up of the batches for submission and validation. It should follow the TTR or PDR

sent by R&D using the same equipment when possible. The PV protocol is prepared by technical

services and the submission MBRs regarding the production of the batches too. The PV report includes

a side by side evaluation between submitted batch scale and proposed batch scale. The batches

manufactured for validation use qualified facilities, systems and equipment, and are tested using

validated analytical methods. All raw-materials and primary package materials are analysed and

released before production.

Hikma’s industrial facility is located in Sintra and is divided in two separate industries, one of them is

composed by eight production lines for injectable manufacturing and the other has three lines for the

production of antibiotics in powder form. Two new products were presented in this dissertation as

examples of technology transfers and submission processes produced on line 5. This line is specific for

the production of solutions in vials to be used for injections.

Injection A is an ANDA indicated for the treatment of heart complications and transferred to the Hikma

PT manufacturing site. Three batches were produced for each presentation (1 mL/vial in 2 mL vials and

5 mL/vial in 6 mL vials), the strength is 0.2 mg/mL for both presentations. The manufacturing conditions

54

defined include compounding and filling under yellow light. Regarding oxygen, nitrogen protection is

required to protect the product from its high sensitivity to oxygen. The ideal pH range (3.5-4.0) was

chosen in accordance to the results of the tests performed, certain pH values increased the impurities.

Different process parameters were challenged during validation in order to establish the best conditions

at the transferred site. Effects of mixing times and speeds were tested as well as effects of initial set up

and line stoppage, the filling machine speed was studied and also the effects of filling on the solution.

The speeds were defined as 200-300 rpm for the 30 L batches and 300-400 rpm for the 50 L batches.

The maximum bulk holding time was established as 48 hours and 2 hours for line stoppage. OOS results

were obtained for oxygen values during filling, for the 5 mL/vial presentation batches, the reason was

attributed to a human error in addition to the inadequacy of the oxygen reading equipment towards 6

mL vials. For the batches produced for the 1 mL/vial presentation batch the most probable root cause

was related to the misalignment of the needle holder/support from the filling machine. Corrections were

implemented and the results were favourable. The physicochemical and microbiological results were in-

specification. The PV report is still in construction for product A, the scale-up plan for commercial sizes

and submission to the authorities has not been concluded.

Injection B is an aseptically filled product transferred to Hikma Portugal. It is not considered a new

product because it has been developed and submitted before by Bedford. One batch of each

presentation was manufactured for the same bulk concentration of 10 mg/mL (5 mL/vial – 6 mL vial

capacity and 25 mL/vial – 30 mL vial capacity). No special concerns are necessary for the manufacturing

process, the product is not sensitive to light, heat or oxygen. The same activities performed for the

evaluation of the submission manufacturing of product A were applied to the manufacturing of product

B. The mixing speeds were defined as 400 rpm for the 5 mL/vial presentation and 1200 rpm for the 25

mL/vial presentation. For both batches the bulk holding time was tested until 72 hours and was stable.

The maximum contact with the filter was defined as 16 hours in the MBR, however the necessary time

for filtration did not overcome 5 hours. The filling duration was 35 minutes for the 5 mL/vial presentation

batch and 3 hours and 30 minutes for the 25 mL/vial presentation batch – including the 2 hours of line

stoppage as indicated in the protocol. The volume to be purged before filling (dead volume) defined in

the MBR was established as superior to 1000 mL. For the 5 mL/vial presentation it was defined as 192

vials (equivalent to around 1075 mL using a fill volume of 5.60 mL per vial) and for the 25 mL/vial

presentation 71 vials were defined, equivalent to around 1867 mL using a fill volume of 26.30 mL

per vial.

Based on the results and parameters obtained for the validated batches, it was possible to plan the

scale-up. The proposed commercial batch sizes were established based on the number of viable vials

obtained on the submission batches. It was decided to slightly increase the proposed batch sizes to

accommodate routine bulk losses and to be within 10 times the theoretical submission batch sizes. The

5 mL/vial presentation batch of 30 L was scaled up to 380 L and the 200 L batch correspondent to the

25 mL/batch presentation was scaled up to 1800 L. For product B the submission to the authorities is

almost ready to be done.

55

Advances in measurement tools (PAT) and quantitative analysis/modelling enables real-time monitoring

of critical steps and the scale-up of pharmaceutical processes with more reliability. These advances are

expected to cause a shift from trial and error to rational process scale-up following a QbD initiative.

The practical examples presented – injection A and B, were subjected to studies in the R&D site in USA.

The research involved the establishment of the CQAs and CPPs which had impact on those CQAs. The

parameters identified as critical and responsible for potential failure causes were further evaluated

based on risk management tools - FMEA. Risk assessment consists of the identification of hazards and

the analysis and evaluation of risks associated with exposure to those hazards.

Concerning the example of injection A, the oxygen headspace is a CPP, to monitor the oxygen levels, a

lighthouse oxygen headspace analyser was used. This equipment can be considered a PAT tool

because it follows the required definitions, it is a process analyser system which provides non-

destructive measurements and delivers information on a critical product attribute.

Regarding injection B, there were no difficulties during the production of the submission batches. It is a

simple and stable manufacturing process. However, it was noticed that after adding the API to the

mixture, a homogenous solution was obtained after about one hour. The TTR received by Bedford

required a three hour duration to obtain the dissolution of the mixture. It can happen that between

commercial batches the raw-materials can present slight alterations or the equipment can face

problems. Taking into consideration the variability/unpredictability that can occur, an implementation of

a PAT tool such as a NIR probe in the compounding tank can help controlling the dissolution and assure

a consistent quality product output.

6.2. Future work

Following the above described work, it would be interesting in future terms, to develop a more in-depth

study of the following aspects:

- Trend analysis of the results and deviations observed during validation and commercial batch

manufacturing at Hikma PT in order to identify the most frequent problems observed;

- Investigation about which could be the most necessary PAT tools to implement at Hikma PT in

accordance to the characteristics of the kind of products manufactured in this site and the most

regular problems detected.

56

57

7. References

Akhavan A. Technology Transfer to Developing Countries: The Iranian Experience, Ph.D. Dissertation,

University of Bradford 1995, UK

Al Ghailani HH, Moor WC. Technology Transfer to developing countries. International Journal of

Technology Management 1995; 10 (7/8): 687-703

Ali S, Pandit V, Chander S. Technology Transfer in Pharmaceuticals. Int Res J Pharm 2012; 3(6): 43-

48.

Colvard B, Jarmusik K, Brown D. Technology Transfer of Injection A. West-Ward Pharmaceuticals 2016;

1-46

Dogra R, Garg R and Jatav P: Technology Transfer in Pharmaceutical Industry: Transfer of process from

Development to Commercialization. Int J Pharm Sci Res 2013; 4(5); 1692-1708.

FDA. Guidance for Industry PAT — A Framework for Innovative Pharmaceutical Development,

Manufacturing, and Quality Assurance; 2004

FDA. Guidance for Industry Process Validation: General Principles and Practices; 2011; 8-16.

Feifei YUE, Yigming YUE. Research on Technology Transfer in the Pharmaceutical Industry, Technology

Transfer Journal of Tongji University. Natural Science 2008; 11-14.

Ghafaripour A. Fundamental Concepts for Technology Transfer Process, Industries Ministry, Tehran

1999.

Gibson M. Pharmaceutical Preformulation and Formulation: A Practical Guide form Candidate Drug

Selection to Commercial Dosage Form, Interpharm/CRC Press, A division of Taylor and Francis Books

2001, Inc, FL, USA.

Gibson M. Pharmaceutical Preformulation and Formulation: A Practical Guide form Candidate Drug

Selection to Commercial Dosage Form, Interpharm/CRC Press, A division of Taylor and Francis Books

2001, Inc, FL, USA.

Gorman ME. Types of knowledge and their roles in technology transfer. Journal of Technology Transfer

2002; 27: 219-231.

Gupta et al. Technology Transfer in pharmaceutical industry: An overview. Novel Science International

Journal of Pharmaceutical Science 2012; 1(7): 430-434

ICH. Guideline for pharmaceutical development Q8(R2); 2009

ICH. Quality Risk Management Q9; 2005

58

Keyur B. Ahir, Khushboo D. Singh, Sushma P. Yadav, Hetal S. Patel, Chetan B. Poyahari Overview of

Validation and Basic Concepts of Process Validation. Sch. Acad. J. Pharm., 2014; 3(2): 178-190

L. Nandhakumar, G. Dharmamoorthy, S. Rameshkumar and S. Chandrasekaran An overview of

Pharmaceutical Validation: Quality Assurance view point. IJRPC 2011; 1(4)

Madieh S, Bucur B, Ali M. Technology Transfer of Injection B. West-Ward Pharmaceuticals 2015; 1-17

Madu CN. Transferring technology to developing countries – Critical factors for success. Long Range

Planning 1989; 22(4): 115-124.

Mansfield E. East-West technological transfer issues and problems, International technology transfer:

forms, resource requirements, and policies. American Economic Review 1975; 65(2): 372-376.

Marques R, Lopes A, Dana L. Technology Transfer between Hikma R&D and Hikma Farmacêutica

Portugal. Hikma Sintra 2015; 1-16

Marques R, Cordeiro I, Campos R. Data Compilation Protocol for Product A (10 mg/mL). Hikma Sintra

2016; 1-30.

Marques R, Cordeiro I, Campos R. Process Validation Protocol for Product B (0.2 mg/mL). New ANDA.

Hikma Sintra 2017; 1-25.

Marques R. Data Compilation Report Draft for Product B (0.2mg/mL). Hikma Sintra 2017; 15-21.

Ortega AJ, Arce NA, Sequeda F, Gribenchenko I. Management of Innovation and Technology Transfer

Process Between University and Industry. The Mater Res Cent Case in Universidad del valle, Colombia

2009; 2: 7-8.

Osman Gani AAM. International technology transfer for competitive advantage: A conceptual analysis

of the role of HRD. Competitiveness Review 1999; 9: 9

Patel DS, Nadiad. Technology Transfer an Overview of Pharmaceutical Industry. Int Biopharm

Association Publ 2009; 2(4): 2-8.

Patil RP. Technology Transfer in Pharmaceutical industry: Objective, Issues and Policy Approaches. Int

J Pharma Res Dev 2010; 2(10): 43-48

Rodrigues, A.; Almeida, J.; Lopes, L.; Menezes, J. Analysis of a pharmaceutical process powder mixture

with NIR spectroscopy and chemometric tools. Instituto Superior Técnico da Universidade de Lisboa,

Faculdade de Farmácia da Universidade de Lisboa, Generis Farmacêutica, S.A.; 2015

Sarvani V, Elisha Raju P, Sreekanth Nama, Leela Madhuri Pola, Chandu Babu Rao Process Validation:

an essential process in pharmaceutical industry, International Journal of Medicinal Chemistry & Analysis,

Vol 3 Issue 2 2013; 49-52

Sharma Sumeet, Singh Gurpreet. Process validation in pharmaceutical industry: an overview. Journal

of Drug Delivery & Therapeutics; 2013, 3(4), 184-188

Souder WE, Nashar AS, Padmanathan V. A guide to the best technology transfer practices. Journal of

Technology Transfer 1990; 15: 1-2.

59

Souder WE, Nashar AS, Padmanathan V. A guide to the best technology transfer practices. Journal of

Technology Transfer 1990; 15: 1-2.

Steenhuis HJ, De Boer SJ. Differentiating between types of technology transfer: the technology building.

International Journal of Technology Transfer and Commercialization 2002; 1(1/2): 187-200.

WHO Expert Comitee on Specifications for Pharmaceutical Preparations WHO technical report

Guidelines on the validation for manufacturing processes. Series no. 863-34th report annex 6 1996; 4-7

WHO Good manufacturing practices for pharmaceutical products. Comitee on specification for

pharmaceutical preparation, report series no. 823, Geneva; 1992, 14-96

WHO Guidelines on Validation. Working document QAS/16.666; 2016

WHO Supplementary guidelines on good manufacturing practices: validation, technical report series.

No. 937, 2006 Annex 4

Saudi Food & Drug Authority. Guidelines for Process Validation of Pharmaceutical Dosage Form Version

2; 2010

http://www.hikma.com/en/about-us/global-presence.html (Accessed at August 2017)

A1

8. Annexes

Annex A - Risk management tools (FMEA) for product A

Table 12 – FMEA for product A

ID Process step Cause failure Potential effect Effect Severity Probability Criticality Detection SPD Control

A API and

excipients

calculation

Incorrect added amount Low or high assay Batch failure 3 1 3 1 3 Calculation of the quantity of API is double verified before weighing.

B API handling Handling instruction

missing

API quality Batch failure 3 1 3 1 3 API is weighed in a glass flask. API is pre-dissolved with WFI before adding to

the compounding tank. The API is weighed in a controlled humidity glove box,

up to a maximum of 24 hours before being used.

C Dissolution of

API/ excipients

Incorrect visual

evaluation

Low or high assay,

homogeneity

Batch failure 3 1 3 1 3 Dissolution steps are double verified. A pre-dissolution is performed before

addition to the preparation tank to avoid powder losses.

D Protect bulk from

light exposure

Human error, handling

instruction missing

Increase of impurities,

product degradation

Batch failure 3 1 3 2 6 The filling room uses yellow lights.

E Protect bulk

solution from

oxygen exposure

Human error, excessive

exposure to ambient

conditions

Increase of impurities,

product degradation

Batch failure 3 2 6 2 12 WFI is continuously sparged with filtered nitrogen throughout the

compounding. Oxygen sensor is calibrated before compounding.

F pH

measurement

and pH

adjustment

pH meter malfunction,

error in pH adjusting

solution quantity

Higher or lower pH

measurement result

Batch failure 3 2 6 1 6 pH meter is calibrated daily to cover the pH range on the MBR. pH adjustment

is performed with acid and basic solutions and checked by IPC pH meters

G Final QS Human error, scale

malfunction

Low or high assay Batch failure 3 1 3 2 6 Balance is calibrated and verified prior to each compounding, the step is

double verified. Final volume is verified with calibrated ruler.

H Filtration Product incompatibility

with filter membrane,

filter clogging

Low assay or high

impurity level

OOS results 3 1 3 1 3 The filter membrane used is the same as tested and validated at the R&D

site.

I Filling Improper nitrogen

headspace purging

Product degradation Batch failure 3 2 6 2 12 Oxygen headspace is monitored by IPC. The product is filled with nitrogen

flushing. The filling machine is qualified to meet product oxygen headspace.

specifications. Error in weights entry or

machine malfunction

Incorrect fill volume Low or high

assay, content

uniformity

affected

2 1 2 1 2 Filling start-up with volume verification by IPC. In-process control of fill volume

by IPC.

Washing machine, tunnel

or filling mal function

Product degradation Quality of the

batch

3 2 6 1 6 Critical holding time study, including the evaluation of the impact of 2 hours

line stoppage during filling.

J Protect product

from light

Human error, handling

instructions missing

Product degradation OOS results 3 2 6 1 6 The product was challenged under normal lights and did not significantly

impact the product quality for an exposure period up to 7 days.

K Inspection Product defects/particles Product degradation Batch failure 1 2 2 1 2 Inspection of the batches for particles. Samples are placed on stability.

A2

Annex B – Sampling scheme for product A

Figure 7 – Sampling scheme of product A manufacturing process

A3

Annex C – Risk management tools (FMEA) for product B

Table 13 – FMEA for product B

ID Process step Cause failure Potential effect Effect Severity Probability Criticality Detection SPD Control

A API calculations Added amount of

API is > or <

Low or high assay Batch failure 3 1 3 1 3 Calculation of the quantity of API is double verified before weighing.

B API handling Handling

instructions missing

API handling, low

or high assay

Batch failure 3 1 3 1 3 The weighing process is performed in a plastic bag.

C pH measurement

and adjustment

pH meter

malfunction, error

in pH adjusting

Higher or lower pH

measurement

result

pH OOS results,

product degradation

3 1 3 1 3 pH meter is calibrated daily to cover the pH range. pH adjustment is performed with

acid or basic solutions. Sample is checked by IPC pH meters.

D API dissolution Incorrect visual

evaluation

Low or high assay,

homogeneity of the

bulk solution

Batch failure 3 1 3 1 3 Dissolution of API is double verified. A minimum dissolution time is stated. A sample

from the bottom of the preparation tank is collected to verify API dissolution.

E Final QS Human error, scale

malfunction

Low or high assay Batch failure 3 1 3 2 6 Balance is calibrated and verified prior to each compounding. This step is double

verified.

F Filtration Product

incompatibility with

filter membrane,

filter clogging

Low assay, high

impurity level

Batch failure 3 1 3 1 3 Filter membranes to be used (0.2 µm) are the same as previously used in West-Ward

(Bedford). Filter validation reports are available.

G Filling Error in filling

weights entry or

machine

malfunction

Incorrect fill

volume;

Low or high

assay

2 1 2 1 2 Filling start-up with volume verification by IPC. In-process control of fill volume by IPC.

Washing machine,

tunnel or filling

machine,

malfunction leading

to line stoppages

Product

degradation

Compromise

quality of the

batch

3 2 6 1 6 The product is not oxygen or light sensitive. Maximum bulk hold time and the impact of

a 2 hours line stoppage is studied.

H Inspection Product defects;

colour or particles

Product

degradation or

process related

Batch failure 3 2 6 1 6 Inspection of the batches. Samples are placed on stability after inspection.

L Labelling and

Packaging

Uncontrolled

exposure to

ambient conditions

Product

degradation

Batch failure 3 1 3 1 3 The product is not light or heat sensitive, submission batches are labelled and packed

under normal lights to challenge the product.

A4

Annex D – Sampling scheme for product B

Figure 8 - Sampling scheme of product B manufacturing process

A5

Annex E - Risk assessment for the technology transfer of product B from Bedford to Hikma

Table 14 - Risk assessment for the technology transfer of product B from Bedford to Hikma regarding compounding

Item Current New Justification for change Risk Categorization

Batch size 80 L and 250 L 50L and 200 L As part of the technology

transfer to the new

manufacturing site. To

generate sufficient vials for

stability

Low

Compounding evaluated during submission

batches. Commercial sizes defined based on

market demand and during process validation.

Compounding

tank

75 gallons stainless

steel tank with a

propeller

100L or 200L stainless

steel

tank with a 4 blade

magnetic stirrer

As part of the Technology

Transfer to the new

manufacturing site

Low

Same material of construction (stainless

steel).

Compounding process evaluated during

submission batches manufacture.

Temperature

Mixing speed

and time

21-23ºC. 8-12 minutes

for excipient

dissolution, 178-182

minutes for API

dissolution. Mixing

speed: 550-600 rpm

20-25ºC (room

temperature). Average

mixing times and speeds

challenged on each step

As part of the Technology

Transfer to the new

manufacturing site

Low

Compounding process evaluated during

submission batches manufacture; New mixing

speeds/times evaluated to ensure efficient

mixing in the new tank without splashing and

without excessive foaming (if any).

API &

Excipients

API from Reliable.

Excipients from

Bedford suppliers

Same API supplier. Hikma

excipients suppliers

As part of the Technology

Transfer to the new

manufacturing site

Low

New mixing speeds/times evaluated during

submission to ensure efficient mixing

dynamics in the new tank without splashing

and without excessive foaming (if any).

A6

Table 15 - Risk assessment for the technology transfer of product B from Bedford to Hikma regarding filtration and filling

Item Current New Justification

for change

Risk categorization (low/medium/high)

Filtr

ati

on

Filtration

train

design

Two filters in parallel for

clarification and two

redundant filters used for

final filtration (0.2 µm)

One final filter inside the

sterile core (0.2 µm)

As part of the

technology

transfer to the

new

manufacturing

site

Low

Two filters in series inside the sterile core does not mean higher

sterility assurance. All the internal standard sterility assurance

controls are in place during submission. Same filter membrane

is used with the same porosity and from the same supplier. Less

extractables are expected by using one filter instead of two.

Fillin

g

Tubing Platinum cured silicone or

teflon

Teflon

Low

Teflon is considered as an inert material. Teflon has lower

extractables.

Automatic weighing is efficient. The filling machine is capable of

filling the product within the specified limit.

Fill volume consistency along the filling process is evaluated

during the manufacture of the submission batch. A proper

extended data compilation is performed during submission and

PV batches in order to evaluate the impact of each

manufacturing step on product quality, which includes extended

in-process analysis during filling in order to validate the entire

filling process and prove that it is not affecting the quality of the

product.

Fill

volume

Target fill weights are

checked every 15 minutes

gravimetrically using the

theoretical density to

ensure that vials are filled

correctly

Fill weight, based on

actual density. Filling line 5

has an automatic IPC.

Manual volume

verification is also

performed

IPC of

bulk

solution

Appearance, assay, pH,

density, osmolality and

bioburden were tested

before filtration

Bulk solution is tested

(before filtration). Routine

IPC tests are performed

(particles)

A7

Annex F – Risk assessment for the increase of product B batch size from submission scale to a proposed commercial

scale

Table 16 - Risk assessment for the scale-up to commercial size regarding the preparation tank, dissolution times, mixing speeds and filtration of product B

Item Submission batch sizes

(50L, 200L)

Proposed batch sizes

(380L, 1800L)

Rational/Risk evaluation

Preparation

tank

100L stainless steel tank.

200L stainless steel tank

600L stainless steel tank

2000L stainless steel tank

Low

Same material of construction (stainless steel).

Dissolution

times and

mixing

speeds

The minimum times were

challenged

The mixing times will be

challenged based on the data

collected on the submission

batches

Low

Typical mixing ranges for the selected tank will be used and evaluated

to assure proper dissolution/homogenization, having as indicative the

data collected during submission batches.

Filtration 0.2 µm PVDF filter

0.2 µm PVDF filter

Low

Same filter reference will be used.

Filter validation studies are available, covering the following

parameters: 16 hours as the maximum contact time of the product with

the filter, a maximum pressure of 2000 mbar and a maximum flow of

11.59 mL/min.cm2 of filter area.

Dead volume (which includes any filter adsorption) was evaluated and

conclusions are applicable to the new batch size.

Maximum contact of the

product with the filter: NA

Maximum contact of the product

with the filter: 16 hours

Filtration pressure ≤ 2.0 bar Filtration pressure ≤ 2.0 bar

A8

Table 17 - Risk assessment for the scale-up to commercial size regarding the filtration and actual production yield of product B

Item Submission batch sizes

(50L, 200L)

Proposed batch sizes

(380L, 1800L)

Rational/Risk evaluation

Filling

The filling duration was less than 2

hours (for both batches without

considering line stoppage

challenges)

The approximate filling

duration is between 4 hours

and 8 hours, assuming an

average filling speed for

each presentation

Maximum/minimum filling

speeds will be challenged

Low

The estimated filling times are well within the current maximum

qualified time for aseptic filling (26 hours 25 minutes) and well within

the maximum holding time between the start of API addition and the

end of filling (established as 72 hours at room temperature).

The 2 hours line stoppage was challenged and showed no impact on

the quality of the finished product.

The filling speed was 100% for both

batches (466 vials/minute for the

6 ml vials and 216 vials/minute for

the 30 mL vials)

Actual

production

yield

(based on

quantity for

stock and

total

samples)

77% – 91% *

Expected yield is ≤99% *

%991002000000

100002000000

ml

mlml

%98100500000

10000500000

ml

mlml

Not applicable

The manufacturing process yield limits will be established after

manufacture of at least 30 batches.

* Considering worst case fixed bulk/product losses for commercial batches of around 10L for residual volume in the filtration / filling system; for IPC (bulk testing

and samples during filling) and for finished product testing – and without considering rejected vials during inspection.

A9