80
i Optimization of a Manufacturing Process of Parenteral Lyophilised Drugs: Process Validation Cláudia Rego Rodrigues Thesis to obtain the Master of Science Degree in Pharmaceutical Engineering Supervisors: Eng. Samuel Mendes Geraldes Camocho Prof. Carlos Manuel Faria de Barros Henriques Examination Committee Chairperson: Prof. José Monteiro Cardoso de Menezes Supervisor: Eng. Samuel Mendes Geraldes Camocho Member of the Committee: Prof. João Fernandes de Abreu Pinto November 2019

Optimization of a Manufacturing Process of Parenteral

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Optimization of a Manufacturing Process of Parenteral

i

Optimization of a Manufacturing Process of Parenteral

Lyophilised Drugs: Process Validation

Cláudia Rego Rodrigues

Thesis to obtain the Master of Science Degree in

Pharmaceutical Engineering

Supervisors: Eng. Samuel Mendes Geraldes Camocho

Prof. Carlos Manuel Faria de Barros Henriques

Examination Committee

Chairperson: Prof. José Monteiro Cardoso de Menezes

Supervisor: Eng. Samuel Mendes Geraldes Camocho

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

November 2019

Page 2: Optimization of a Manufacturing Process of Parenteral

ii

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

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

Page 3: Optimization of a Manufacturing Process of Parenteral

iii

Page 4: Optimization of a Manufacturing Process of Parenteral

iv

Preface

The work presented in this thesis was performed at department of Liquid an Injectables Division of

Hikma Farmacêutica S.A (Terrugem, Portugal), during the period February – July 2019, under the

supervision of Eng. Samuel Mendes Geraldes Camocho. The thesis was co-supervised at Instituto

Superior Técnico by Prof. Prof. Carlos Manuel Faria de Barros Henriques.

Page 5: Optimization of a Manufacturing Process of Parenteral

v

Page 6: Optimization of a Manufacturing Process of Parenteral

vi

Abstract

Exhibit batches were manufactured in 2011 and an ANDA with two presentations Product Y for

Injection 5 g/vial Product Y for Injection 10 g/vial were submitted to authorities. The approval was

guaranteed to Hikma by FDA on October 2018.

During launching batches of Product Y 5 g/vial it was noticed a high breakage rate, around 13,6%,

representing a non-robust and not validated process. The improvement of Product Y 5 g/vial was

essential and for that it was necessary to investigate the life cycle of Product Y. During the

investigation it was concluded that the root cause of vial breakage was attributed to the fast-freezing

phase. The fast freezing associated with the high filling volume caused a rapid and large expansion of

the cake resulting in breakage problems.

In order to correct this problem, a Risk Assessment for the lyophilisation cycle between Product Y 5

g/vial and 10 g/vial was made. The lyophilisation cycle and the recipe of Product Y 10 g/vial was then

adapted to the 5 g/vial resulting in a decrease to 0,7% of rejected vials resulting in the product’s

validation. After correction the product was successfully launched.

Additionally, process improvement for efficiency purpose proposed a Line Transfer and a scale-up. For

that, a Process Validation Protocol was designed and during these activities a decrease to 0,4% of

broken vials was observed. With these results it was possible to confirm that our modifications were

made successfully resulting in a robust process and independent from the line of the production.

Keywords: Lyophilisation; Process Validation; Life Cycle; Optimization; Technology Transfer; Scale-

up.

Page 7: Optimization of a Manufacturing Process of Parenteral

vii

Page 8: Optimization of a Manufacturing Process of Parenteral

viii

Resumo

Em 2011, após os lotes de exibição, um ANDA com duas apresentações Produto Y para Injeção 5 g/

frasco Produto Y para Injeção10 g / frasco foram submetidos às autoridades. A aprovação foi

acreditada à Hikma pelo FDA em Outubro de 2018.

Durante o lançamento de lotes do Produto Y 5 g / frasco, verificou-se uma percentagem elevada de

frascos rejeitados, cerca de 13,6%, representando um processo não robusto e, portanto, não

validado. A otimização do Produto Y 5 g / frasco foi essencial e, para isso, investigou-se o ciclo de

vida do produto. Durante a investigação, concluiu-se que a principal causa de frascos partidos deve-

se ao rápido congelamento. O congelamento rápido associado a um volume de enchimento alto

provoca uma expansão rápida do produto, resultando em problemas de quebra.

Para corrigir este problema, foi feita uma Avaliação de Risco para o ciclo de liofilização entre o

Produto Y 5 g / frasco e 10 g / frasco. O ciclo de liofilização do Produto Y 10 g / frasco foi adaptado

para a receita de 5 g / frasco, resultando numa diminuição de frascos rejeitados para 0,7% tornando o

processo validado.

Adicionalmente, uma Transferência de Linha e um “scale-up” foram propostos. Para tal, foi elaborado

um Protocolo de Validação de Processo e, durante as atividades, foi observada uma redução para

0,4% de frascos partidos. Com os resultados obtidos, foi possível confirmar que as modificações

foram feitas com sucesso, resultando num processo robusto e independente da linha de produção.

Palavras-chave: Liofilização; Validação de Processo; Ciclo de Vida; Otimização; Transferência de

Tecnologia; “Scale-up”.

Page 9: Optimization of a Manufacturing Process of Parenteral

ix

Page 10: Optimization of a Manufacturing Process of Parenteral

x

Acknowledgments

A tese não é apenas resultado de um empenho individual, mas sim de um conjunto de esforços que o

tornaram possível e sem os quais teria sido muito mais difícil chegar ao fim desta etapa, que

representa um importante marco na minha vida pessoal e profissional. Desta forma, manifesto a

minha gratidão a todos os que estiveram presentes no decorrer destes últimos anos.

Em primeiro lugar gostaria de agradecer à Hikma Farmacêutica pela oportunidade de realizar o meu

estágio nas suas instalações.

Ao Samuel Camocho, orientador externo do estágio, por me ter acolhido e recibo da melhor forma

dando-me a excelente oportunidade de fazer parte da sua equipa, pelos seus conselhos e pela sua

disponibilidade para me ensinar.

Ao Professor Carlos Henriques, orientador interno da tese pela disponibilidade e orientação prestados

nesta fase final.

Não posso deixar de agradecer a todos os meus colegas de trabalho e supervisoras que desde o

primeiro dia estiveram sempre disponíveis para o esclarecimento de dúvidas. Aos que fizeram parte

do meu dia-a-dia na Hikma tornando-o mais fácil.

À minha família. Aos meus pais por serem inalcançáveis comigo, por todos os esforços feitos por

mim, por todos os ensinamentos, valores, educação, amor e incentivo transmitidos, por serem os

meus maiores pilares. Às minhas irmãs por serem as melhores do mundo, por me colocarem à frente

delas, por todo o apoio, amor e carinho. Sem o vosso apoio e presença na minha vida nada disto era

possível. Ao resto da família por sempre terem acreditado em mim e por me depositarem confiança.

Às minhas amigas de sempre pelos momentos partilhados ao longo deste percurso, por todo o apoio

e acima de tudo por estarem sempre presentes nos bons e sobretudo nos maus momentos da minha

vida, sem o amor transmitido por elas seria mais difícil.

Ao João, por teres sido a maior presença e apoio nesta etapa, nos momentos mais difíceis e nas

horas de maior fadiga, por toda a compreensão e paciência. Por todo o amor, dedicação e pelos

momentos partilhados.

Obrigada!

Page 11: Optimization of a Manufacturing Process of Parenteral

xi

Page 12: Optimization of a Manufacturing Process of Parenteral

xii

Table of Contents

Preface.................................................................................................................................................... iv

Abstract .................................................................................................................................................. vi

Resumo ................................................................................................................................................ viii

Acknowledgments .................................................................................................................................. x

Table of Contents ................................................................................................................................. xii

List of figures ....................................................................................................................................... xiv

List of Tables......................................................................................................................................... xv

List of Acronyms ................................................................................................................................ xvii

1. Introduction ................................................................................................................................... 20

1.1. Hikma Farmacêutica S.A. .................................................................................................... 20

1.2. Regulatory Authorities .......................................................................................................... 21

1.2.1. Documentation ........................................................................................................ 21

2. Lyophilisation ............................................................................................................................... 22

2.1. Desired Characteristics of lyophilised products: .................................................................. 23

2.2. Advantages and disadvantages of lyophilisation process ................................................... 27

3. Process Validation ........................................................................................................................ 29

3.1. Different Methods of Validation ............................................................................................ 29

3.2. Process Validation Phases ................................................................................................... 31

3.3. Qualifications Steps ............................................................................................................. 33

4. Process Validation, Quality System and Quality Risk Management (QRM) ........................... 35

4.1. Risk Assessment .................................................................................................................. 35

4.1.1. Risk Assessment Methodology ............................................................................... 36

5. Technology Transfer..................................................................................................................... 39

5.1. Scale-up ............................................................................................................................... 39

6. Manufacturing Process of Lyophilised Products .................................................................... 41

6.1. Manufacturing Process ........................................................................................................ 41

6.2. Description of the Line and Facilities ................................................................................... 43

Page 13: Optimization of a Manufacturing Process of Parenteral

xiii

7. Related Work: Improvement of Lyophilised Pharmaceutical Form ........................................ 46

7.1. Description of Product Y 5 g/vial and 10 g/vial .................................................................... 46

7.2. Problem Reported ................................................................................................................ 48

7.3. Life Cycle of Product Y for Injection, USP 5 g/vial and 10 g/vial ......................................... 50

7.4. Practical Case ...................................................................................................................... 58

7.5. Line Transfer and Scale-up for Product 5 g/vial – a Process Validation .............................. 61

7.5.1. Process Evaluation Activities and Results .............................................................. 62

7.6. Launching ............................................................................................................................. 67

8. Conclusion .................................................................................................................................... 68

8.1. Future work .......................................................................................................................... 70

9. References .................................................................................................................................... 71

10. Appendix ....................................................................................................................................... 74

Page 14: Optimization of a Manufacturing Process of Parenteral

xiv

List of figures

Figure 1 - Steps Involved in Lyophilisation (Lyophilisation Cycle).

Figure 2 - Lyophilisation Process Cycle.

Figure 3 - Phase Diagram of Water.

Figure 4 - Design of a Lyophilizer.

Figure 5 - The Different Phases of Process Validation.

Figure 6 - Classification of the Risk Level.

Figure 7 - Evaluation of the Risk Priority.

Figure 8 - General Steps Involved in the Manufacture of Lyophilised Products.

Figure 9 - Lyophilised Manufacturing Process and Facilities.

Figure 10 - General View of the Filling Machine.

Figure 11 - General GLT Layout.

Figure 12 - General Load/Unload Line Description.

Figure 13 - Freeze-dried Product Y bulk solution at -4.0 ºC.

Figure 14 - Freeze-dried Product Y bulk solution at -0.7 ºC.

Figure 15 - Freeze-dried Product Y bulk solution at -0.3 ºC.

Figure 16 - Typical Bottomless Breakage of the Product Y 5 g/vial Presentation During

Lyophilisation.

Figure 17 - Life Cycle of Product Y for Injection, USP 5 g/vial and 10 g/vial.

Figure 18 - Squeme of Loading positions for PV batches and samples collection.

Figure 19 - Life Cycle of Product Y for Injection after Launching.

Page 15: Optimization of a Manufacturing Process of Parenteral

xv

List of Tables

Table 1 - Parameters for Production of Product Y.

Table 2 - First Submission Lyophilisation Cycle Parameters.

Table 3 - First R&D Trial Lyophilisation Cycle Parameters.

Table 4 - Second R&D Trial Lyophilisation Cycle Parameters.

Table 5 - Fourth R&D Trial Lyophilisation Cycle Parameters.

Table 6 - Final Lyophilisation Cycle Parameters to be Applied for FDA Submission.

Table 7 - Approved Freezing Phase Parameters.

Table 8 - Lyophilisation Cycle Parameters Comparison for Product Y 5 g/vial (Current and

Improved Freezing Phase) and 10 g/vial.

Table 9 - Side-by-side Inspection Results of Batch of Product Y for Injection USP 5 g/vial.

Table 10 - Percentage of cracked vials reject after Scale-up and Line Transfer.

Table 11 - Launching for Product Y for Injection USP.

Table 12 - Approved Batch Sizes for Product Y for Injection, USP 5 g/vial.

Page 16: Optimization of a Manufacturing Process of Parenteral

xvi

Page 17: Optimization of a Manufacturing Process of Parenteral

xvii

List of Acronyms

ANDA

API

cGMP

CMAs

CPPs

CQAs

DQ

EMA

FDA

FLC

FMEA

FMECA

GMP

HEPA

INFARMED

IPC

IQ

MENA

OQ

PAT

PPQ

PQ

PV

QRM

R&D

Abbreviated New Drug Application

Active Pharmaceutical Ingredient

Current Good Manufacturing Process

Critical Material Attributes

Critical Process Parameters

Critical Quality Attributes

Design Qualification

European Medicines Agency

Food and Drugs Administration

Filling Machine BOSCH

Failure Modes Effect Analysis Method

Failure Mode Effect Critical Analysis

Good Manufacturing Process

High Efficiency Particulate Arrestance

Autoridade Nacional do Medicamento e Produtos de Saúde

In Process Control

Installation Qualification

Middle East and North Africa region

Operational Qualification

Process Analytical Technology

Process Performance Qualification

Performance Qualification

Process Validation

Quality risk management

Research and Development

Page 18: Optimization of a Manufacturing Process of Parenteral

xviii

RPN

SA

USP

WFI

Risk Priority

Active Substance

United States Pharmacopoeia

Water for Injection

Page 19: Optimization of a Manufacturing Process of Parenteral

xix

Page 20: Optimization of a Manufacturing Process of Parenteral

20

1. Introduction

This dissertation under the theme, " Optimization of a Manufacturing Process of Parenteral Lyophilised

Drugs: Process Validation", was designed to obtain a master's degree under the Master of

Pharmaceutical Engineering, in which for six months, I did a practical internship at Hikma

Farmacêutica S.A.

I was part of the production team of lyophilised injectable products where I had the pleasure to follow

all production processes, documentation for each batch, people management, process optimization

and the opportunity to support the commissioning and validation of a new manufacturing line

production.

The purpose of this thesis is to contribute to the knowledge about development of injectable

pharmaceutical products and the improve/optimization of the process and how technology transfer and

Process Validation is performed at Hikma Farmacêutica.

1.1. Hikma Farmacêutica S.A.

In 1978, Hikma company was founded in Jordan by Samih Darwazah where it has established itself as

a leading supplier of branded generics and in-licensed products. After two years, on the 80’s, Hikma

established the first FDA-inspected manufacturing plant in the region of MENA (Middle East and North

Africa region). In this decade, Hikma extended beyond the Middle East and acquired land in Portugal

for the construction of a sterile manufacturing plant for injectable pharmaceutical products. The land

acquired in Sintra, Terrugem was the first step into a new region and major turning point for the

company.

After the successful expansion in Portugal and the continued effective delivering medicines in the

MENA, Hikma entity expressed interest in the US market and acquired West-Ward Pharmaceuticals,

in 1990s. The continuous growth of the company came with the acquisition in Tunisia and Saudi

Arabia who rename Hikma as a leading provider of generics in the MENA region but also

internationally.

In 2000, Portugal and Saudi Arabia manufacturing plants had successfully passed FDA inspections. It

was in this decade that Hikma expanded into the lyophilised market - variant of injectable products-

with the investment of a specialized manufacturing plant in Italy. Not yet satisfied, they invested on the

oncology market by buying Ribosepharm GmbH and Thymoorgan in Germany.

Hikma continued to expand, not just in Europe, but also in Egyptian market with Alkan Pharma and

Jordan with acquisition of Arab Pharmaceutical Manufacturing Company.

Page 21: Optimization of a Manufacturing Process of Parenteral

21

After all the investments, 2005 was a historic mark and the Group listed on the London Stock

Exchange raising gross proceeds of US$124 million.

The current decade has seen growth through acquisitions of penicillin manufacturing plant in Al Dar Al

Arabia in Algeria.

Hikma saw a future on the injectables business market and bought Baxter’s, Bedford Laboratories and

Ben Venue Laboratories in the United States also as Roxane Laboratories in Columbus who helped

create a good position in the non-injectables market.

Nowadays, Hikma produces over 650 products and is present in 50 countries with 29 manufacturing

plants and seven different R&D centers.

In Portugal, the Group have three different facilities for parenteral drugs: Production of powders,

liquids and injectables and one recently implemented for the production of oncological products. Being

Sintra site known as the “center” of the injectable expertise for the company.

1.2. Regulatory Authorities

The pharmaceutical industry, through current Good Manufacturing Practices (cGMPʹs), maintains high

quality assurance standards with regard to the development, manufacture and control of medicines.

GMP also ensures that all products are manufactured only by authorized manufacturers whose

activities are regularly inspected by the competent authorities using quality risk management

principles.

Furthermore, Regulatory Agencies are responsible for controlling all activities concerning the

development, manufacture and marketing of pharmaceutical products. Given the fact that Hikma

Pharmaceuticals' largest target market is the United States, it is important to cite the Food and Drugs

Administration (FDA) regulatory agency. For this company, not only FDA is important but also the

agencies with national and European jurisdiction, INFARMED I.P. and European Medicines Agency

(EMA).

All European Community pharmaceutical manufacturers require manufacturing authorizations whether

the products are sold inside or outside the Community.

1.2.1. Documentation

Hikma is committed to high standards of ethical conduct and is required by law to create and maintain

a documentation system that involves all specifications, formulations, processing and packaging

instructions, procedures and records of the various manufacturing operations performed. Documents

must be clear, error free and up to date (INFARMED).

Page 22: Optimization of a Manufacturing Process of Parenteral

22

2. Lyophilisation

Lyophilisation, also known as freeze-drying, is a very soft drying process widely used in

pharmaceutical industry to produce various pharmaceutical products including: parenteral products,

vaccines, proteins, antibiotics, serums and biotechnological products (Hottot, Vessot, and Andrieu

2007) (Abdelwahed et al. 2006).

Lyophilisation can be defined as the drying process of a frozen product in which the majority of water

and other solvents is directly removed by sublimation (converted directly into water vapor) without

passing through the liquid state leaving a dry porous mass of approximately the same size and shape

as the original frozen mass (Deluca and Lachman 1965; Nireesha et al. 2013).

Many pharmaceutical products as parenteral and biopharmaceutical products are thermal sensitive

(thermolabile) or unstable in solution form for prolonged storage periods, but that are stable in the dry

state (S. M. Patel, Doen, and Pikal 2010). Lyophilisation process is used in this products that might

lose their quality or activity in conventional evaporative drying, removing the solvent to promoting long-

term stability and increases the shelf life of the pharmaceutical drugs (Taylor et al. 2007; Nireesha et

al. 2013). Freeze-dried formulations not only have the advantage of better stability, but also provide

easy handling (shipping and storage) (Franks 1998; Heller, Carpenter, and Randolph 1997).

Freeze-Drying cycle can be divided into three steps: freezing (solidification), primary drying (ice

sublimation) and secondary drying (desorption of unfrozen water).

Figure 1. Steps involved in lyophilisation (Lyophilisation cycle).

Controlled lyophilisation keeps the product temperature low enough during the process to avoid

changes in the dried product appearance and characteristics (Ellab Validation Solutions 2018).

Page 23: Optimization of a Manufacturing Process of Parenteral

23

2.1. Desired Characteristics of lyophilised

products:

• Intact cake;

• Uniform color;

• Sterile;

• Sufficient strength;

• Rapid dissolution (sufficiently porous);

• Chemically stable;

• Free of particles;

• Dried.

At the end of the lyophilisation a good appearance of dried cake with high quality and a porous

structure allowing a fast reconstitution after rehydration is desired.

Freezing stage

In the past, process optimization was mainly focused on the primary and secondary drying step.

However, with improved process understanding, it was recognized that the freezing step is also

important and start to be highlighted.

Freezing is the first step of lyophilisation. This process consists on converting the liquid solution into

the solid state in which water is converted to ice form by applying low temperatures. As the freezing

process continues, more water contained in the liquid freezes and the concentration of the remaining

liquid increases (called the freeze concentrate) (Tang and Pikal 2004).

The freezing phase is a critical step because it fixes the ice crystals structure, shape and dimensions.

The type of ice crystal structure affects the pores specific area, a key factor for the drying process and

rehydration time and consequently the sublimation time which is the longest time of the whole

lyophilisation process. The ice crystal sizes depend also on the vial size and type and also on the

filling height, larger fill volume takes longer to fully freeze (Hottot, Vessot, and Andrieu 2007;

Abdelwahed et al. 2006).

Some products are simple crystalline material, although the majority of products that are lyophilized

are amorphous and form glassy states when frozen. Frozen products can be categorized as crystalline

or amorphous glass in structure: amorphous solids are rigid structures but they lack a well-defined

shape, they are non-crystalline.

Page 24: Optimization of a Manufacturing Process of Parenteral

24

Crystalline products have a well-defined eutectic freezing and melting point that is its collapse

temperature. Amorphous products have a corresponding glass transition temperature and they more

difficult to freeze dry. The collapse temperature of amorphous products is typically a few degrees

warmer than its glass transition temperature.

The determination of the critical collapse temperature of a product is an important step in establishing

and optimizing a freeze-drying process. This critical temperature determines the maximum

temperature that the product can withstand during primary drying without it melting or collapsing.

To optimize the lyophilisation cycle, optimal ice crystal sizes must be searched to minimize the

operating costs related to the whole duration of these drying steps. Usually, rapid cooling results in a

product with small ice crystals and more difficult to freeze-dry while slower cooling results in large ice

crystals.

The cooling rate and the supercooling degree are factors who control the freezing process and are

correlated with the morphology of the frozen material affecting the drying process. The supercooling

degree is defined as the difference between the equilibrium freezing point and the temperature at

which ice crystals first form. The supercooling degree is important because determines the number of

ice crystal formed (Constantino and Pikal 2004).

Freezing is a key step because microstructure established during this process usually represents the

microstructure of the dried product (Nireesha et al. 2013) and also has shown impact in intra-vial and

inter-vial uniformity as primary and secondary drying performance (Kasper, Winter, and Friess 2013).

Normally, vial breakage can be prevented by using slow freezing.

The cooling rate

The frozen liquid should be kept at the set temperature for sufficient time to transform all the

suspension into solid. Different ramped cooling on the shelves give different supercooling effects. One

practical approach to improve a lyophilised product consists in minimizing the specific surface area of

ice by growing large ice crystals; this can be possible by controlling the rate of super cooling. Normally,

higher supercooling rate results in larger ice specific surface are forming smaller ice crystals. A high

cooling rate induces higher nucleation of the ice crystal making compact ice crystal while a moderate

cooling rate is better for lyophilised products (Tang and Pikal 2004; Hottot, Vessot, and Andrieu 2007).

Primary drying

After all water and solutes have been converted into a frozen matrix, conditions must be established to

start the primary drying stage. In this process, the bulk water can be removed from the frozen product

via sublimation. Typically, the primary drying stage consumes the largest cycle time of lyophilisation

Page 25: Optimization of a Manufacturing Process of Parenteral

25

and the optimization of this step has a large economic impact (Tang and Pikal 2004; Adelaide et al.

2004).

Once the product is frozen, primary drying starts whenever the chamber pressure is reduced by

vacuum pump evacuation and the shelf temperature is raised to supply the heat removed by ice

sublimation. Heat is transferred by thermal conduction from the shelf through the vial and finally gets

into the product. Sublimation requires heat energy to drive the phase change process from solid to gas

and it is important to maximize the surface contact of the vial with the shelf (Abdelwahed et al. 2006).

During this process, the chamber pressure is well below the vapor pressure of ice and ice is migrated

from the product to the condenser and crystallization onto the cold plates in the condenser (Tang and

Pikal 2004). The rate of sublimation ice depends on the difference in vapor pressure of the product

compared to the vapor pressure of the collector because the molecules are transferred from the high-

pressure sample to the lower pressure area. The concentration gradient between dried face and

condenser is the driving force for removal of the bulk water. In the condenser, a refrigerated surface at

a temperature below the product removes the vaporized solvent evolved by the drying material from

the vacuum chamber by converting it back as ice (Deluca and Lachman 1965).

The vapor pressure increases with an increase in temperature and are directly correlated. For this

reason, is necessary that the product temperature to be warmer than the collector temperature.

However, it is difficult to find an equilibrium because the temperature must be also low enough to

prevent any melting of the frozen mass maintaining the integrity and sufficiently high to maximize the

drying process and complete in a reasonable cycle time (Nireesha et al. 2013; Gan et al. 2005) .

Primary drying is a slow process in which product must be kept below its critical collapse temperature,

as the product degradation may occur, to produce acceptable drug product (S. M. Patel, Doen, and

Pikal 2010).

In the end, solute must form the rigid structure to support its weight after ice removal.

Secondary drying

The last stage of lyophilisation is the secondary drying. Secondary drying involves the removal of

adsorbed water from the cake which did not separate out as ice during the freezing step therefore did

not sublimate off.

At the end of primary drying, when all of the free ice crystals have been sublimed, the product will

appear to be dried. However, the moisture content can still have a percentage of water molecules

attached to the product. Usually, residual water is present in enough quantities to cause rapid

decomposition of the product when is stored at room temperature. Accordingly, is necessary to do a

secondary drying (Tang and Pikal 2004).

Page 26: Optimization of a Manufacturing Process of Parenteral

26

When the secondary drying starts, the product and shelf temperature will increase and must be kept at

high temperature for a period sufficient to allow the isothermal desorption. Normally, for an

optimization of the process is better to run high shelf temperature for a short time than a low

temperature for a long period. Contrary to primary drying which use low shelf temperature and high

vacuum, secondary drying raises the shelf temperature (higher than ambient) and reduces the

chamber pressure to the lowest attainable level.

The residual water in the product remaining at this stage is stronger, requiring more energy to be

removed.

When lyophilisation is over, the moistures contents should be less than 5%, resulting in an elegant

porously dried cake (S. M. Patel, Doen, and Pikal 2010).

Figure 2. Lyophilisation process cycle.

The Principle of Freeze-Drying

At the triple point liquid, solid and gas coexist in equilibrium. When the water is heated below the triple

point, then it will change from solid state to vapor state directly. Sublimation can take place at

pressures and temperature below triple point to enable conversion of ice into vapor, without entering

the liquid phase.

The eutectic temperature is the temperature at which a product start to melt into a liquid. During

lyophilisation, a product must be held below the eutectic temperature for sublimation to occur from the

solid state. The first step for lyophilisation is freezing the product bellow the eutectic temperature.

Then in primary drying phase the temperature raises near to the collapse temperature with a safety

margin and is subjected to a reduced pressure turning the water content into vapor leaving solid and

dried components of the original liquid (Nail et al., 2002).

Page 27: Optimization of a Manufacturing Process of Parenteral

27

Figure 3. Phase Diagram of Water.

2.2. Advantages and disadvantages of

lyophilisation process

Advantages:

• Ensures sample stability in a dry state;

• Removal of water without excessive heating of the product

• Rapid and easy dissolution of reconstituted product (rapid reconstitution time);

• Increases shelf life;

• Easily for storage (can be stored at room temperature) and shipping (reduces weight and volume of samples);

• Good for O2 and air sensitive drugs;

• Product is process in the liquid form (simplifies aseptic handling);

• Does not shrink samples;

• Purity of samples

• Elegant cake appearance

Disadvantages:

• Increased handling and processing time;

• Cost (expensive unit operation) and complexity of equipment;

• Issues associated with sterilization and sterility assurance of the dryer chamber and aseptic loading of vials into the chamber (Drug Discovery and Development);

Page 28: Optimization of a Manufacturing Process of Parenteral

28

Figure 4. Design of a lyophilizer

The lyophilisation process occurs in a lyophilizer or freeze-dryer (figure 4). The main components are:

• Chamber;

• Condenser;

• Vacuum pump.

A lyophilizer consists of a chamber that contains product shelves capable of cooling and heating the

vials. A vacuum pump, a condenser, a refrigeration unit and associated controls are connected to the

vacuum chamber.

The most common application of lyophilisation is the manufacturing of parenteral products that are

administered after a simple reconstitution step (Shukla 2011).

Page 29: Optimization of a Manufacturing Process of Parenteral

29

3. Process Validation

Effective process validation contributes to assuring drug quality (FDA, 2011). The basic principle of

process validation “is the collection and evaluation of data, from the process design stage through

commercial production, which establishes scientific evidence that a process is capable of consistently

and uniformly producing a product according to the specifications and quality characteristics” (FDA,

2011).

Moreover, process validation assures that product quality is performed, within established parameters,

consistently from batch-to-batch and unit-to-unit (WHO 1996). The purpose of validation is to prove the

effectiveness in a manufacturing process to ensure varied inputs with high quality outputs (V. K. K. B.

Patel and Pethe 2010; ICH Definition).

A well-established PV results in a reduction of product losses, incidence of deviations, quality cost,

down times and the risks of failing in GMP. Additionally, increases the safety levels, the data to provide

knowledge for a robust process, greater rationalization of the activities developed, simpler

maintenance of equipment and easier scale-up from development work (Nandhakumar 2013).

3.1. Different Methods of Validation

Currently, validation is conducted according to different approaches: based on experimental data or

historical data.

Based on experimental data we have prospective validation and concurrent validation. The first one

happens before process commissioning or the product's market reach, while the latter is executed

during production (Comissão Europeia 2001). Retrospective validation is centered in historical data

and used for established products with stable processes.

Prospective Validation

Prospective validation is adopted when new drug products are introduced, a product is made under a

revised manufacturing process or before the process is launch in commercial use. A risk analysis of

the production process is made during the development stage and is examined into individual steps to

determine if they might lead to critical conditions that may affect the quality of the product. This

evaluation is supported by data from experimentation and theoretical considerations.

Three different experiments have to be designed and entirely reported in a protocol. Equipment,

production environment and analytical assay methods need to be validated and critical situations

identified. To have a PV well designed, an extensive sampling and testing should be done at different

steps of the manufacturing process.

Page 30: Optimization of a Manufacturing Process of Parenteral

30

In case unstable conditions are discovered the risk must be evaluated and the potential causes are

considered. Inadequate manufacturing processes are modified and improved until validation proves

that they are acceptable and finally master batch records can be made attending all the critical

conditions and parameters determined (Ahir et al. 2014; Nandhakumar 2013).

Concurrent Validation

This validation occurs during normal production runs and involves in-process monitoring. Concurrent

validation supports documentation to prove that the production process in under control. The decision

to perform this validation needs to be correctly justified, well-reported and approved by authorized

labourers.

Retrospective Validation

is based on historical data, documents that provide evidence for a structure's/system's correct

operation and control. This type of validation is established for products with stable processes and

already in distribution. If the operating procedures, the composition of the product or the equipment

are changed or reviewed this type of validation shouldn't be applied (Ahir et al. 2014; Nandhakumar

2013).

The required protocol with the results of the data review should include batch processing and

packaging records, maintenance logbooks, process control charts, records of personnel changes,

finished product data, storage stability results and process capability studies (WHO 1996).

Retrospective validation is no long encouraged and prospective validation is opted for. In fact, the

impossibility to use retrospective validation in manufacturing of sterile products has a negative impact

which leads to preferences in other types of validation.

Revalidation

Revalidation, used after a change with impact on product quality, serves to demonstrate that any

intentional or unintentional change in a process manufacturing or in the process environment does not

adversely perturb process parameters and quality characteristics. Documentation requests are the

same as for the initial validation of the process.

After a change, it is important to implement the alterations affecting the standard procedure to confirm

they maintain validated and high quality. Whoever facilities, equipment and manufacturing techniques

including cleaning where no significant modifications have been made, needs a periodically review to

confirm they remain valid. Each alteration must be carefully analysed by validation department to

determine if it is sufficient to opt for a revalidation and in that case, in which scope (Ahir et al. 2014;

Nandhakumar 2013).

Some situations that require revalidation are:

• Changes in raw materials (that can affect the process or the product quality);

Page 31: Optimization of a Manufacturing Process of Parenteral

31

• Changes in the manufacturing process- different batch size;

• Changes in the equipment;

• Changes in the facilities;

• Changes in the provider of the active raw material manufacturer;

• Changes in packaging material.

In cases of replacement of the equipment for exactly the same machine normally is not require

revalidation except the new equipment need to be qualified for the use.

A well justified document is necessary to support the decision of not perform revalidation studies and

must be done by authorized persons.

3.2. Process Validation Phases

Validation in general requires a meticulous preparation and careful planning involving various activities

during the lifecycle of the product and process. PV is divided in three phases: process design,

process qualification and continued process verification.

Figure 5. The different phases of Process Validation.

Process Design

PV is described as the collection and evaluation of data. In this phase, is exclusively focused on

qualification efforts and the manufacturing process is drawn based on knowledge gained during R&D

and scale-up activities.

During the research and development phase, important aspects as formulation, dosage forms, stability

and storage conditions, process capability, equipment qualification, pilot batch, scale-up and transfer

technology studies contribute to the process design (Ahir et al. 2014; Nandhakumar 2013).

Process control strategy takes place in this phase.

Page 32: Optimization of a Manufacturing Process of Parenteral

32

Process Qualification

Process Qualification is where process design is evaluated to check if the process is capable to

reproduce commercial manufacturing batches. There are two different aspects of process qualification

to be considered:

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

In this stage, activities are performed to demonstrate that all equipment and utility systems are

suitable for their intended use and to verify if are capable to perform properly. The first step of these

activities is the selection of the equipment and utilities, the construction materials, the operating

principles and the performance characteristics.

The second step is the Installation Qualification (IQ). The IQ exists to verify if all equipment is built and

correctly installed in compliance with the design specifications, if have all capacity and functions

requested and if it is correctly calibrated and connected.

Operational Qualification (OQ) follows IQ and is used to confirm that utilities and equipment operate in

accordance with the specifications and operating ranges.

• Process Performance Qualification (PPQ).

In this stage, GMP procedures should be followed. It is essential evaluate if the process design is able

to reproduce commercial manufacturing batches. After properly tested, even under “worst-case”

conditions, with satisfactory results and all established limits of Critical Process Parameters are

validated it is possible to start the commercial distribution (Ahir et al. 2014; Nandhakumar 2013).

Continued Process Verification

On-going verification it is required during production routine to confirm that the process remains in a

stable and solid control.

Data collected during continued process validation is important to optimize the process or to improve

the quality of the drug. When corrective actions are made due to problems are detected during this

phase, a document describing the changed plan must be written with a rationale for the adjustment, a

good strategy implementation of the change plan and it has to be approved by the quality department.

To assure continued process validation a frequent review of all process related documents including

failures, deviations, change control procedures and process modifications as also validation audit

reports are studied (Ahir et al. 2014; Nandhakumar 2013).

Page 33: Optimization of a Manufacturing Process of Parenteral

33

3.3. Qualifications Steps

For a good work in a pharmaceutical industry it is imperative for the qualification to keep up with

validation. Validation is the “act of prove effectiveness” in a process while qualification is used to

evaluate and qualify an equipment. A process is considered validated when the equipment and

process are able without put in risk the safety and quality of the product.

For this reason, validation and qualification must work synchronized and in harmony (WHO 2006,

Sharma 2013).

Design Qualification (DQ)

In new facilities or equipments, the first section of validation is the design qualification. The design

must be established in compliance with GMP and well reported (Saudi Food & Drug Authority 2010).

Installation Qualification (IQ)

The main goal of this type of qualification is to check if all processing equipments are in accordance

with the installation specifications (manuals and engineering drawings). IQ is executed on new

facilities, systems and equipments and must include the examination of equipment design, the

maintenance adjustments, the equipment calibration, the services and instrumentation checked, the

working instructions, the verification of materials construction, the description of equipment, the

principle of operation and equipment requirements.

In short, IQ is to demonstrate that the installation was successfully completed.

Operational Qualification (OQ)

Operation qualification follows a previously authorized protocol. In this document, OQ protocol should

identify the studies to be undertaken on the critical operating conditions, the acceptance criteria to be

met, the sequence of those studies and the measuring equipment to be used. OQ should take in

account the circumstances, covering upper and lower processing and the upper and lower limits

mentioned as “worst-case” scenario including operation controls and alarms. OQ should include tests

that challenge the equipment and it’s functions and also incorporates the stoppages, interventions, the

start-up and all activities expected during the routine production.

Page 34: Optimization of a Manufacturing Process of Parenteral

34

The main goal of OQ qualification is to prove satisfactory operation over the normal operation routine

procedure and consequently acknowledge the basis for training the operators (Ahir et al. 2014;

Nandhakumar 2013).

Performance Qualification (PQ)

After a successful IQ and OQ, the final step (performance qualification) should be complete to certify

that the equipment is working reproducibly and safety within a certain set of parameters for the critical

variables.

Tests using production materials and tests using a set of conditions to simulate worst-cases, cleaning,

calibration and preventive maintenance need to be reported.

In order to produce PQ batches (normally corresponding to the first commercial-scale batches of the

drug product) a control strategy for the manufacturing process should have been established and

previously well-designed.. It is important to define the performance criteria, how much and what data

must be collect and when data must be collected

Usually, a PQ protocol has a higher level of sampling, additional testing of process manufacturing than

during routine commercial production. The increased level of examination and sampling is used to

establish appropriate levels and frequency for the routine sampling and monitoring, taking in

consideration the process complexity, the size of the batch, the previously experience with the product

and process.

After a successful PQ comes the commercial distribution of the drug product. All data gathered from

commercial-scale batches and also from laboratory and pilot-scale studies is used to support the

decision of beginning commercial distribution (Ahir et al. 2014; Nandhakumar 2013; WHO 1996).

Page 35: Optimization of a Manufacturing Process of Parenteral

35

4. Process Validation, Quality System

and Quality Risk Management (QRM)

Before a batch is commercialized, the manufacturer must demonstrate that the commercial

manufacturing process is capable of consistently produce acceptable quality products within

commercial manufacturing conditions - meeting those attributes relating to identity, strength, quality,

purity, and potency (CQAs). The assurance should be obtained from objective information and data

from laboratory, pilot, and/or commercial scale studies. (Guidance for Industry – Process Validation:

General Principles and Practices, January 2011).

Process validation is part of technology transfer and is used to demonstrate that the manufacturing

process developed by R&D and transferred to industrial scale, operated within established

parameters, can consistently yield a product meeting its specified quality attributes. It is important

however to recognize that process validation is not an isolated event.

The purpose of process validation is to certify a set of varied inputs cannot affect the regular

production and lead to consistent and high-quality yields. PV is an ongoing process throughout routine

production that must be adapted by pharmaceutical companies as a guaranteed quality and safety

manufacturing control.

Quality risk management is a systematic process that identifies and prioritizes, evaluates and controls

potential risks to quality. It facilitates continual improvement (assessment, control, communication and

review of risks) of process performance and product quality throughout the product lifecycle.

Risks identification is a key step for the safe design of a manufacturing process; after the threats are

pointed out it is important to evaluate their consequences, as well as their causes.

4.1. Risk Assessment

The main goal of risk assessment is to minimize the process risk based on the identification of the

potential risk sources. The risk assessment has to be part of a quality risk management system and

ask the following questions:

- What can fail?

- What is the probability that the failure will occur?

- What are the consequences if this process goes wrong?

The first phase of risk assessment is the risk identification and consists in answering the question:

“What can cause the problems?”. The goal is to make a list with all potential risks and can be

extensive as possible. Since identifying all potential failures can be an enormous task when

Page 36: Optimization of a Manufacturing Process of Parenteral

36

considering an entire manufacturing process, the process map generated during the initiation phase of

the risk management process is a tool to do logical and focused steps during the risk assessment.

After the risk identification starts the risk analysis and involves a risk assessment tool , which have a

qualitative or quantitative estimation of the consequence and the ability to detect the failure.

Next the risk analysis, a risk evaluation is required to measure if the risk is in an acceptable level of

risk (the action threshold) or not. At this point, the risk assessment phase ends. The risk management

process continues through the steps of risk control, risk review and communication. This implies that

during lifecycle of the product, the risk assessment is reviewed. This review includes adjustments to

regulatory requirements or to additional information related to the new process.

4.1.1. Risk Assessment Methodology

The risk assessment is made on the Failure Modes Effect Analysis Method (FMEA). This method is

based on the identification of the potential risk sources – defined as Failure Mode. Each failure mode

is evaluated and classified as result of the Probability of Occurrence [P], Severity of the Potential effect

of the failure [S] and the Detectability [D]. The risk prioritization is calculated as RPN = [P] X [S] X [D].

Frequency of probability [P]

The probability of an event may be rated, but not necessarily limited to, 1, 2, or 3 according to its

likelihood.

• Level 1 (low): The frequency of the event occurring is perceived low;

• Level 2 (Medium): The frequency of the event occurring is perceived to be medium;

• Level 3 (High): The frequency of the event is perceived to be high.

Severity of the potential effect of the failure [S]

This approach requires the team to consider which impact this change/event has on the product

quality. The impact of the consequence may be rated but not limited to1, 2, or 3:

• Level 1 (Low): Expected to have a minor negative impact on the product quality.

• Level 2 (Medium): Expected to have a moderate impact on the product quality.

• Level 3 (High): Expected to have a very high significant negative impact the product quality.

Page 37: Optimization of a Manufacturing Process of Parenteral

37

Probability of detection [D]

The purpose of this stage is 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: reliable detection methods are in place (validated methods of analysis).

• Level 2: there is a reliable detection used but are not designed to properly detect the quality of

the product parameter that is being tested.

• Level 3: there is no reliable detection method available to test the product quality parameter in

question.

Risk Classification [C]

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 [P] and severity [S]. Therefore, the risk

ranges from:

Figure 6. Classification of the risk level.

The Risk Classification (PxS=C) gives us an indication of the impact on product quality or data

integrity:

• Level 1 means that the probability that this failure appearing is high and that the impact on

product quality is high or medium;

• Level 2 means a moderate impact on product quality.

• Level 3 means that we have practically no impact on product quality.

Risk Priority [RPN]

The risk priority is a multiplication of three parameters, thus taking in account the potential failure [C;

Risk Classification] associated with the potential effect and its detectability [D]. The risk priority can

range from:

Page 38: Optimization of a Manufacturing Process of Parenteral

38

Figure 7. Evaluation of the risk priority.

The Risk Priority is used to select the appropriate risk mitigation and to focus the validation effort:

• A high priority RPN means that this function or component is critical and other measures and

controls need to be taken to mitigate the risk.

• A medium priority RPN means that this function or component is potentially critical and other

measures and controls needs to be in place to mitigate the risk.

• A low priority RPN means that this function or component is non-critical and that there are no

actions need to be taken.

Design of freeze-drying process is often approached with trial and error experimental plan or the

protocol used on the first laboratory run is adopted without further attempts at optimization, losing

robustness and efficiency (Tang and Pikal 2004).

All changes that may affect product quality or reproducibility of the process should be formally

requested, documented and accepted. The likely impact of the change of facilities, systems and

equipment on the product should be evaluated, including risk analysis.

Page 39: Optimization of a Manufacturing Process of Parenteral

39

5. Technology Transfer

Technology transfer implicates transfer of product and process knowledge to achieve the

manufacturing process for commercialization and it includes all the activities required since the

pharmaceutical development (R&D) to production, for new products, or from one manufacturing site to

another.

During the technology transfer, many aspects need to be taken into consideration to allow proper

evaluation and validation of the manufacturing process before starting routine production for

commercialization. A proper analysis of material attributes and process parameters, inputs,

manufacturing controls, the process outputs and their quality attributes are essential for successful

technology transfer and an inappropriate understanding of this correlation may lead to an uncontrolled

process. An uncontrolled process can result in extensive product losses, batch rejection, difficulties

with regulatory submissions and lately in a non-approval submission.

Through a technology transfer, all unit operations and their associated equipment should be identified

as well as all the parameters and attributes considered critical. All relevant information about the

product and the process gained during the development phase, should also be properly reviewed and

evaluated.

When a lyophilised product is to be manufactured, an appropriate lyophilisation cycle should be

developed. During R&D the cycle development is done, however adjustments in cycle times and

parameters may occur when transferring from R&D freeze dryer to an industrial lyophiliser.

Lyophilisation involves many parameters (essential to achieve for a proper process control and to

obtain a cake with desired characteristics). These operation parameters include heat and mass

transfer, load condition (partial or full load), the container system, shelf temperature and chamber

pressure that are essential to achieve for a proper process control and to obtain a cake with desired

characteristics. These parameters sometimes differ with the technology transfer. Therefore,

manufacturing of test batches is advisable before production of the PPQ batches (Rogers, Takegami,

and Yin 2001).

5.1. Scale-up

Scale-up is generally defined as the process of increasing the batch size. Scale-up of a process can

also be viewed as a procedure for applying the same process to different outputs (volumes).

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

and engineering. Therefore, for a successful scale-up is necessary the critical material attributes

(CMAs) and critical quality attributes of (CQAs) the products and the critical process parameters

(CPPs) of the equipment used in manufacturing process are well-defined.

Page 40: Optimization of a Manufacturing Process of Parenteral

40

Risk management tools can help controlling the critical steps of the process to achieve a successful

scale-up.

Three different scales exist: the laboratory scale batches, following the pilot batches scale and the

production batches scale.

Laboratory Scale Batches

The traditional process of pharmaceutical product development until it reaches the global market

involves a long journey of many experiments, observations, challenges, and resolutions. These

batches are produced at the research and development (R&D) laboratory stage and they present a

very small size.

During the initial formulation stage, certain parameters are considered to convert the raw materials

into a drug formulation with an aim to maintain the required quality attributes. However, at the initial

level, these investigations are performed at a small scale by using small-output equipment, where the

methods and results observed from these investigations are appropriate to that scale only (Amirkia

and Heinrich, 2015).

The production of R&D batches is 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 2013).

Pilot Batches

Moving from R&D to production scale, it is sometimes essential to have an intermediate batch scale-

Pilot Batches which is defined as the manufacturing of a product by a procedure fully representative of

and simulating that for manufacturing scale.

These batches are helpful to gain knowledge and also make possible the production of enough

product for clinical testing and samples for marketing. However, inserting an intermediate step

between R&D and production scales does not in itself guarantee a smooth transition. A well-defined

process may produce a perfect product in laboratory and pilot scale and then fail quality assurance

tests during production.

Production Scale Batches

The batches are of the normal size which will be produced during the routine marketing of the product.

Data on production scale batches may not always be available prior to granting marketing

authorization. When production scale data are not available it is necessary the evaluation and

Page 41: Optimization of a Manufacturing Process of Parenteral

41

characterization of the critical process parameters at laboratory or pilot scale followed by the

manufacture of submission batches and to complete a validation program on production scale batches

(Levin, 2001).

6. Manufacturing Process of Lyophilised

Products

Parenteral drugs are preparations intended for injection through the external tissue, for example the

skin, so that the active substances are administered using gravity or force directly into a blood vessel,

organ, tissue or lesion. These drugs are available as liquid (solutions, emulsions or suspensions) or

solid products (powder – cephalosporins or lyophilised products). Injectable pharmaceutical products

are prepared by methods properly designed to ensure safety and quality and to meet Pharmacopeial

requirements for sterility, pyrogens, particles and contaminants.

6.1. Manufacturing Process

The manufacture of pharmaceutical products should follow the requirements of the current GMPs. The

adherence to cGMPs assure a proper design, monitoring and control each step of a process and the

correct facilities. The use of GMPs guarantees the quality, safety and purity of drug products by

requiring that is manufactured in a properly way and strictly controlled each operation process.

The manufacture of lyophilised products must occur according the sterile rules in clean areas,

constantly cleaned following a standard, and supplied with air who has passed through HEPA filters.

The sterile pharmaceutical products can by produced by terminal sterilization or aseptic processing.

The aseptic processing method is just acceptable when terminal sterilization is not possible due to the

degradation of the drug product when exposed to terminal sterilization conditions, as is the case with

lyophilised products.

Aseptic Processing

Sterile drug products can be manufactured using two techniques: terminal sterilization or aseptic

processing. Terminal sterilization usually involves heat or irradiation. However, some drugs can be

destroyed by exposure to heat or irradiation and it is required to use aseptic filling.

In aseptic filling the drug product and container/closure are subjected to sterilization methods

separately, as appropriate, and then brought together. Because there is no process to sterilize the

product in its final container, it is crucial the containers be filled and sealed in an extremely high-quality

Page 42: Optimization of a Manufacturing Process of Parenteral

42

environment. Before aseptic assembly into a final product the individual parts of the final product must

be subjected to various sterilization processes (Sandle, 2011).

All the components of the final product are sterilized. The rubber stoppers are subjugated to moist

heat sterilization in an autoclave or acquired irradiated (pre-sterilized), the glass containers pass

through a depyrogenation tunnel – dry heat sterilization- and the liquid product subjected to one or

more sterilization filtrations under Grade A. All these manufacturing processes are individually

validated and continuous controlled.

Figure 8. General Steps Involved in the Manufacture of Lyophilised Products.

The first step of the manufacturing process of lyophilised products is the dissolution of the active

substance ingredient (API) and the excipients in the water for injection (WFI) or another solvent. After

the compounding, the bulk solution must pass through a 0,22 µ bacterial retentive filter to sterilize the

product.

While the bulk solution is prepared, the vials are washed to remove the particles and submitted to dry

heat sterilization passing by through a depyrogenation tunnel. Each individually container is filled with

the sterile bulk solution and partially stoppering the containers under aseptic conditions (Grade A).

Page 43: Optimization of a Manufacturing Process of Parenteral

43

The partially stoppered vials filled will be transported to the lyophilizer and loading into the chamber

under aseptic conditions. After that, the solution first will be freeze by placing on cooled shelves in a

freeze-drying chamber and secondly vacuum will be applied to the chamber heating the shelves in

order to sublimate the water from the frozen state. When all the water is sublimed from the vials a

complete stoppering will be done by hydraulic or screw rod stoppering mechanisms installed in the

lyophilizers. Finally, the stoppered vials will be transported from the freeze-drying chamber to the

capper to seal.

Thus, a compound that is heat-sensitive and not sufficiently stable in aqueous solution can be

formulated into a stable solid form, rapidly soluble and elegant injectable preparation (Deluca and

Lachman 1965).

Figure 9. Lyophilised Manufacturing process and facilities.

6.2. Description of the Line and Facilities

To describe the manufacturing process, it is necessary to do an approach of the lyophilisation

manufacturing facilities for the filling of aseptic sterilized products in the Injectable Division at Hikma I.

The batch is prepared in compounding room using the preparation tanks. During and after the

preparation, samples are taken for in process control (IPC) testing. After the compounding, the product

is transferred from the preparation tank to the transfer tank and is connected to the filling machine

(FLC) through a sterilizing grade, integrity tested, filter.

The product is supplied to the filling system via intermediate filling tank. The filling is controlled via

valve, normally closed, and when the valve opens the intermediate filling tank is filled using the

overpressure in the on-site product tank.

The washer for the vials is coupled with the depyrogenation tunnel, in grade D, and is connected with

a turntable of the FLC, under grade A, were the vials after washed and submitted to dry heat

sterilization, wait to be filled.

Filler and Stopper

Pré-Filter

Autoclave

Washer +

Tunnel

Compounding

Vials

Parts

Stoppers

Frames

Grade C Grade D Grade A

Lyophilization

Grade D

Capper100 % Visual Inspection

Pharmaceutical

Area

Under Grade A

Page 44: Optimization of a Manufacturing Process of Parenteral

44

The intermediate tank is connected to six pumps and consequently to six needles who reach the fill

volume to the vials who are running in a transition belt.

The FLC have an integrated monitoring level to control the IPC. Some containers are removed from

the transport belt with the IPC starwheel and weighed in a platform then are placed back into the filling

station and filled. After the filling station, the same vials are taken back to the gross weighing station

and tared. The measure of the values is to check if the vials filled can properly continuous on the

route, in case the fill volume is not correct the containers are not stoppered and in a station latter they

will be rejected from the transition belt.

Figure 10. General View of the Filling Machine.

For the products sensitive to the oxygen, the gassing system can be used to reduce the residual air

inside of the container by flushing to the interior of the vials a protective gas. The protective gas is

supplied by an infrastructure from an on-site system.

The stopper lock is used to release a stopper to the stopper-placing wheel when a vial is present that

should receive a stopper. The vacuum is used to hold the stoppers in the stopper-placing wheel and

the wheels move synchronic with the vial main transport. The wheels of the stopper station are taken

the container from the main transport and partially setting the stopper on the container and take it back

to the main transport.

After the stopper station, the vials are inspected by two sensors, in order to detect the presence of the

stoppers and to detect if stoppers are too high. The reject station is used to reject bad containers: low

volume, high volume, vials without stopper or not correctly placed.

Page 45: Optimization of a Manufacturing Process of Parenteral

45

The good partially stoppered vials are supplied by an in feed belt to the GLT Vials Tray Loader where

are stacked into rows. Once the tray has received the number of the rows determined by the recipe,

the vials are pushed into a tray and thereafter placed to the Lyo trays loader machine (MOTUS).

Figure 11. General GLT layout.

The lyophilisation trays loader machine (Motus) is used for the loading of trays filled with partially

stoppered vials from GLT to the lyophiliser and after the lyophilisation process, to unload the trays to

the turntable in feed of the capping machine. During freeze drying, the containers are totally stoppered

and when all lyophilisation stages are finished the trays will be unloaded by Motus and placed to the in

feed zone of the capping machine.

Figure 12. General load/unload line description.

Stako system is used to separate the vials from the trays for the turntable in feed of the capping

machine, while is collecting and transferring the trays for storage. The stoppered vials are transported

on a conveyor belt to be capped, under grade A air supply. After all the containers are sealed, they

proceed to the visual inspection for defects, ultimately labelling and final packaging, in the packaging

area.

1.1.1.

Section 1 2 3 4 5

Machine Bosch Vials Tray

Loader

Motus Semi-

Automatic Loading System

Hull

Lyophilizer

Stako Vials/Frames

Separation Station

Genesis Vials

Capping Machine

Page 46: Optimization of a Manufacturing Process of Parenteral

46

7. Related Work: Improvement of

Lyophilised Pharmaceutical Form

Due to confidentiality with Hikma Farmaceutica, S.A., this work will not mention the name of the Active

Substance (SA), or the dosages or the commercial name of the finished product, referring only to the

pharmacotherapeutic group to which it belongs and the pharmaceutical form. The drug studied will be

called Product for Injection Y.

7.1. Description of Product Y 5 g/vial and 10 g/vial

Product Y Lyophilised powder for Injection is a new product developed by Research and Development

Department of Hikma Pharmaceuticals at Jordan where the R&D is performed.

Product Y is for intravenous injection, a parenteral, indicated for treatment of severe infections and it is

a generic transferred to Hikma Farmacêutica manufacturing facilities in Portugal. The product Y, is

almost white, or tan to brown, free flowing powder, odourless and having a bitter taste, hygroscopic

powder, freely soluble in water and insoluble in ether and in chloroform.

The formulation is a simple aqueous-based formula that includes the API as well as an anti-gelling

agent and water for injection (WFI), both disappearing during lyophilisation process. The bulk solution

concentration for both strengths is the same 166.67 mg/ml and the pH should be between 2.5 – 4.5.

Studies indicate that Product Y is sensitive to light, unstable under high humidity, unstable under high

temperature and sensitive to oxygen.

The reconstitution volume is 100 ml for the 5 g/vial presentation and 95 ml for the 10 g/vial

presentation using sterile WFI.

Product Target fill volume Batch size Vial size

Injection Y (5 g/vial) 30.50 mL 40 L 100 mL, neck 20 moulded

Injection Y (10 g/vial) 61 mL 80 L 100 mL, neck 20 moulded

Table 1. Parameters for the production of Product Y.

Page 47: Optimization of a Manufacturing Process of Parenteral

47

During the characterization of the Product Y it was used the Freeze-drying microscopy (FDM) a

technique for identifying critical formulation parameters and is the only method for reliably determining

collapse temperatures. Using the FDM, the sample is firstly frozen until it reaches a temperature of -

40ºC. Then, the vacuum pump is turned on and the drying begins. The temperature is then increased,

slowly to allow to see the differences in behavior of the analyzed product.

Figure 13: Freeze-dried Product Y bulk solution at -4.0 ºC. The dried part is beginning to suffer bigger

breakage, whilst the matrix is still holding its structure.

Figure 14: Freeze-dried Product Y bulk solution at -0.7 ºC. As get closer to 0ºC, solution suffers

complete collapse.

Page 48: Optimization of a Manufacturing Process of Parenteral

48

Figure 15: Freeze-dried Product Y bulk solution at -0.3 ºC. The matrix melts completely.

In crystalline solutions the Critical Temperature for Freeze Drying is the eutectic point and in

amorphous products this is the collapse temperature.

The instability of the frozen matrix begins at approximately -5ºC/-4ºC. Because no crystalline shape is

manifested, and no eutectic behavior present (solid and liquid phases presented simultaneously), the

Product Y is an amorphous structure.

7.2. Problem Reported

In January of 2019 after the approval of FDA, Hikma Farmacêutica started to produce Product Y for

Injection, USP 5 g/vial and 10 g/vial and, for launching the product in the US Market and consequently

in order to validate the scale-up process, three Process Validation (PV) batches of 170 L in 400 L

tanks of Product Y for Injection USP 5 g/vial were manufactured at Liquid & Lyophilised Injectable

Production Department- Line 1 at Hikma Farmacêutica Portugal.

To perform all process validation activities, it was necessary more two PV batches under the scope of

PV protocol. The PV included compounding of the bulk solution, pre-filtration through a 1.2 µm filter,

filtration through sterilizing 0.2 µm filter, filling, lyophilisation, stoppering, capping and 100% visual

inspection. In the end, a side by side comparison of finished product results was performed between

40 L batch and 170 L batches manufactured in Line 1 in order to evaluate the reproducibility of results

or if any manufacturing constraint related to the scale up process is noticed.

The evaluation of the effects of the lyophilisation on the quality of the filled vials, lyophilisation

uniformity and the evaluation of the results of 100% visual inspection of the vials was performed in

Page 49: Optimization of a Manufacturing Process of Parenteral

49

three batches. During capping, in one of the inspection spots in the capper, it was noted that a

significant percentage of cracked vials were rejected (between 13.6% and 8.8% of the unloaded

quantity). After the amount of rejected vials was verify it was necessary to open a variance notice and

the batch was immediately put into retention and therefore it was not possible to perform 100% visual

inspection for the PV batches.

The significant percentage of cracked vials rejected turned the manufacturing process in a non-robust

and irreproducible process becoming invalidated. For that reason, the improvement of Product Y 5

g/vial as well as the identification of the root cause for the broken vials was necessary.

During the investigation it was possible to confirm that the problem of vial breakage was in the

freezing phase. The pattern of the crack (Figure 13) indicates that it is caused by expansion of the

cake during freezing phase due to the fact that freezing occurs rapidly and in one shot shocking.

The fast-freezing makes the system of the matrix to expand suddenly without allowing the crystals, for

enough time, to be shaped according to the vial size creating pressure on the edges and causing that

bottomless breakage of the glass followed by a crack on the wall of the vial (Figure 13).

Large fill volumes are strongly correlated with higher percentage of vial cracks especially with fast

temperature gradient. The strain on the vial's axial direction is significantly higher than the hoop

direction typically resulting in bottom lens of the vial coming off (Jiang, 2007).

Figure 16. Typical Bottomless Breakage of the Product Y 5 g/vial Presentation during Lyophilisation.

To follow this investigation, it was necessary to review all data collected and analyze the life cycle

history of the product.

Page 50: Optimization of a Manufacturing Process of Parenteral

50

7.3. Life Cycle of Product Y for Injection, USP 5

g/vial and 10 g/vial

Figure 17. Life Cycle of Product Y for Injection, USP 5 g/vial and 10 g/vial.

In 2011, after the R&D team prepared the formulation and studied the manufacturing process Hikma

Farmacêutica decided to propose to the authorities a new product: Product Y for Injection USP 5 g/vial

and Product Y for Injection USP 10 g/vial. To implement this product in the installations was defined

activities intended to evaluate the manufacturing process for this product for US Market.

A Risk Analysis and Process Validation studies were made to provide evidence that a defined

manufacturing procedure will consistently yield product meeting its predetermined specifications and

quality characteristics (Appendix 1).

Based on Process Validation, one submission batch of 40L was manufactured for Product Y for

Injection, USP 5 g/vial and one batch of 80L for Product Y for Injection, USP 10 g/vial. The submission

batches of Product Y for Injection USP 5 g/vial (40 L) and Product Y for Injection USP 10 g/vial (80 L)

were prepared on 23rd October 2011 and 24th October 2011 respectively at Hikma Farmacêutica

Portugal.

During the primary drying stage in submission batch for Product Y for Injection USP 10 g/vial (80 L),

the product was visually inspected through the sight glass located in both sides of the freeze dryer and

signals of breakage were observed. However, as the cycle was continued and, after the unloading of

the lyophiliser, around 90% of the obtained vials were broken.

In order to completely overcome the problem of vials breakage, R&D department in Hikma

Pharmaceuticals-Jordan started to investigate and to improve the problem appeared in the submission

batch. This research included different optimized cycles (additional trials) performed at R&D level and

industrial scale. After all changes, when R&D create a robust cycle with no vial breakage a new

submission batch of Product Y for Injection, USP 10 g/vial was applied.

The reason behind vial breakage was attributed to the fast-freezing rate applied during freezing phase

on the lyophilisation cycle. To support that, two 20 L batches (Trial #1 and Trial #2) were produced by

applying new optimized lyophilisation cycles and it was decided to proceed for the scale-up to 80 L for

Page 51: Optimization of a Manufacturing Process of Parenteral

51

the second submission batch for Product Y for Injection USP 10 g/vial. Though, after unloading 7% of

vial breakage was still verified.

In order to completely overcome the problem of vials breakage, additional tests were performed by

R&D Jordan and a new modified lyophilisation cycles were applied in 20 L trial batch (Trial #3 and Trial

#4).

First submission batch

During the first submission batch it was verified around 90% of the vials were broken. The applied

cycle was following:

Phase No. Process-Phase Phase Time (h: m) Set Temp. (ºC) Vacuum (µbar)

1. Loading (Precooled shelves) --:-- +5

OFF

2. Freezing

06:00 -50

04:00 -50

3. Primary Drying (Sublimation)

06:00 -20

770

38:00 -20

06:00 -10

04:00 -10

04:00 0

23:00 0

03:00 +10

04:00 +10

02:00 +15

02:00 +15

4. Secondary Drying (Desorption)

02:00 +20

520

03:00 +20

03:00 +25

08:00 +25

02:00 +35

Page 52: Optimization of a Manufacturing Process of Parenteral

52

12:00 +35

Total Cycle Time 132:00 hours

Table 2. First submission lyophilisation cycle parameters.

The main reason for vial breakage was attributed to the fast-freezing rate applied – ramp down to -50

ºC in 6 hours - a factor that is especially important to take in consideration when applied on high

concentrated solutions and large fill volumes, as the case of Product Y 10 g/vial. Following this

investigation, a trial batch with slower freezing rate was applied, Trial #1.

Trial #1

The main goal was to elongate the freezing time from 6 hours to 12 hours. The first 28 hours of the

cycle applied on Trial #1 were considered sufficient to evaluate the performance of the cycle in terms

of containers breakage.

Phase No. Process-Phase Phase Time (h: m) Set Temp. (ºC) Vacuum (µbar)

1. Loading (Precooled shelves) --:-- +5

OFF

2. Freezing

12:00 -50

04:00 -50

3. Primary Drying (Sublimation)

06:00 -20

770

06:00 -20

Total Cycle Time 28 hours

Table 3. First R&D trial lyophilisation cycle parameters.

The Trial #1, was achieved successfully on the R&D-Lyophilizer, the containers did not show any

evidence of breakage. Later on, this cycle was applied at Hikma Farmacêutica Portugal and signs of

cracks on the vials surface started to appear after 28 hours of the cycle. To have a better insight, it

was decided to proceeded up to 69 hours and after the unloading, 167 out of 257 vials were broken,

representing 66% of the total batch. Despite the elongation time could slightly decrease the

percentage of broken vials, a new development trial was made to overcome the problem.

Trial #2

The following modifications were introduced on Trial #2:

Page 53: Optimization of a Manufacturing Process of Parenteral

53

• Adding a hold step for 1h at -10 ºC, the super-cooling temperature, in order to help arrange

the crystals;

• Adding a hold step for 3h at -20 ºC (Product Y freezing point);

• The terminal freezing point was modified to -45 ºC instead -5 ºC;

• Reducing the holding time at -45 ºC from 4h to 3h;

• Decreasing the ramp-up (from -45 ºC to -20 ºC) time from 6h to 3h to enhance the drying of

the cake.

The 3rd and 4th step were introduced to reduce the time at the freezing stage which will minimize the

pressure exerted on the wall of the vials. All the modifications were due to prevent sudden expansion

due to initial crystallization and reduce the time of the product at frozen state which would minimize

the physical stress exerted by the frozen structure on the vial.

Phase No. Process-Phase Phase Time (h: m) Set Temp. (ºC) Vacuum (µbar)

1. Loading (Precooled shelves) --:-- +5

OFF

2. Freezing

02:00 -10

01:00 -10

01:00 -20

03:00 -20

03:00 -45

03:00 -45

3. Primary Drying (Sublimation)

03:00 -20 770

38:00 -20

520 06:00 -10

04:00 -10

04:00 0

420

23:00 0

03:00 +10

04:00 +10

02:00 +15

02:00 +15

4. Secondary Drying (Desorption) 02:00 +20 310

Page 54: Optimization of a Manufacturing Process of Parenteral

54

03:00 +20

03:00 +25

08:00 +25

02:00 +35

12:00 +35

Total Cycle Time 131:00 hours

Table 4. Second R&D trial lyophilisation cycle parameters.

After vials unloading, two vials out of the 213 vials were found broken representing less than 1% of the

total batch indicating that the applied cycle modifications were successful in improving vial breakage

during freezing.

Regarding the R&D-Lyophiliser successfully results, an 80 L submission batch was produced with the

following modifications:

• Holding step for 1h at -12 ºC, the super-cooling temperature;

• Holding step for 4h at -20 ºC, the Product Y freezing point;

• Increasing the ramp-down (from -20 ºC to -45 ºC) time from 3h to 3h and half.

When the lyophilisation cycle was finished it was noticed 7% of vials breakage, and compared with the

first submission batch in which 90% of broken vials were verified, the new cycle did not completely

solve the problem of vials breakage.

In addition, the reconstitution time was longer suggesting that the lyophilised cake was not totally dried

forming a lump upon the addition of reconstitution water complicating the dissolution.

Trial #3

The following modifications were purpose on Trial #3:

• Slow ramp down to -18 ºC in 5h;

• Adding a hold step at -18 ºC for 6h;

• Slow ramp down to -45 ºC in 5h because is expected with a slower freezing rate, large

crystals;

• Holding for 4 hours at -45 ºC;

• Addition of annealing step by increasing the temperature to -18 ºC in 2 hours, holding for 4

hours at -18 ºC and then lowering the temperature down to -40 ºC in 3 hours. This would help

rearranging the crystals and obtaining a more homogeneous frozen structure;

Page 55: Optimization of a Manufacturing Process of Parenteral

55

• Start the primary drying at -10 ºC instead of -20 ºC to help in better and faster primary drying;

• The vacuum applied in primary drying was increased to 100 µbar to enhance the water vapour

sublimation from this large volume cake.

After unloading, 55 % of the vials were found broken showing a non-effective cycle, which did not

solve the minor percentage of 1% of vial breakage (trial #2) and the 7% in the subsequent 80 L

submission batch. More particularly, the addition of the annealing step was not positively.

Trial #4

The modifications involved in the development of this cycle were:

• Removing the annealing step during product freezing stage;

• Addition of a hold step for 6h at -20 ºC, the super-cooling temperature at which the crystals

formation and arrangement start;

• Modification of the terminal freezing point to be -45 ºC instead of -40 ºC to ensure that the

product is reaching the freezing point;

• Reducing the Holding time at -45 ºC from 4h to 3h to reduce the time at the frozen state which

will reduce the pressure exerted on the wall of the vial;

• In the sublimation phase, addition of a holding step for 6h at -30 ºC prior to further increase in

temperature to ensure that the cake is completely sublimed before giving further energy.

Page 56: Optimization of a Manufacturing Process of Parenteral

56

Phase No. Process-Phase Phase Time (h: m) Set Temp. (ºC) Vacuum (µbar)

1. Loading (Precooled shelves) --:-- +5

OFF

2. Freezing

04:00 -20

06:00 -20

05:00 -45

03:00 -45

3. Primary Drying (Sublimation)

00:30 -35

100

03:00 -35

03:00 -30

06:00 -30

05:00 -10

18:00 -10

04:00 0

27:00 0

03:00 +10

05:00 +10

4. Secondary Drying (Desorption)

00:30 +10

300

03:00 +20

04:00 +20

01:00 +25

12:00 +25

01:00 +35

19:00 +35

Total Cycle Time 133:00 hours

Table 5. Fourth R&D trial lyophilisation cycle parameters.

In September 2012 a batch with the modifications was manufactured. After vials unloading and 100%

visual inspection, no vials were found broken. This indicates that the applied lyophilisation cycle was

Page 57: Optimization of a Manufacturing Process of Parenteral

57

successful by solve the problem of vials breakage. Thereafter, the reconstitution time was evaluated

and it was out the specifications.

Forthwith, changes in the optimized cycle (having as reference the cycle applied in trial #4) were

successful in enhancing the mentioned product characteristics: crystallization, cake dryness, water

content and reconstitution time. The changes applied are not anticipated to cause vial breakage since

the phenomenon is directly affected by freezing stage and primary drying. Moreover, this cycle was

challenged by loading 100 mL filled vials along with 60 mL ones and no vials breakage was observed

by the end of the cycle.

Based on the results obtained from all the trials, scale-up and submission batches, it was

recommended to proceed with the preparation of the submission batch of Product Y 10 g/vial applying

the following lyophilisation cycle:

Phase No. Process-Phase Phase Time (h: m) Set Temp. (ºC) Vacuum (µbar)

1. Loading (Precooled shelves) --:-- +5

OFF

2. Freezing

06:00 -20

08:00 -20

06:00 -45

03:00 -45

3. Primary Drying (Sublimation)

00:30 -35

100

03:00 -35

03:00 -30

06:00 -30

05:00 -10

18:00 -10

04:00 0

35:00 0

4. Secondary Drying (Desorption)

04:00 +25 100

00:30 +25

50 16:00 +25

01:00 +35

Page 58: Optimization of a Manufacturing Process of Parenteral

58

19:00 +35

Total Cycle Time 138:00 hours

Table 6. Final Lyophilisation cycle parameters to be applied for FDA submission.

After all data collection, Hikma Farmacêutica wrote, in 2012, a Data Compilation Report for the

submission of Product Y for Injection, USP 5 g/vial and Product Y for Injection, USP 10 g/vial where

the critical process steps in the manufacturing process were evaluated according to the Risk

Assessment (Appendix 2).

The compounding of the bulk solution formula, bioburden reduction filtration through a 1.2 µm filter,

final filtration through a 0.2 µm filter, filling, lyophilisation and inspection were accepted in October,

2018 when the FDA gives the approval for the manufacturing process for Product Y for Injection, USP

5 g/vial and 10 g/vial.

7.4. Practical Case

After the review of all data collected about Product Y and an exhaustive investigation of the life cycle it

was possible to confirm that the problem was in the freezing phase similar what happened in the past

with the 10 g/vial form. In the freezing phase, it happens quickly the icing of the lyophilised cake and

this step causes an expansion in the cake volume.

In order to improve and solve the cause of this type of breakage during lyophilisation attributed to the

freezing phase it was proposed to adjust the recipe (LYC018) of Product Y 5 g/vial to become similar

to the freezing step of the Product Y 10 g/vial, already improved in the past, in which the freezing

pattern is gradual rather than shocking the matrix sudden and with very low temperature.

To adapt the recipe of Product Y 10 g/vial in the recipe of Product Y 5 g/vial it was necessary to

perform a Risk Assessment where all the risks were evaluated.

Page 59: Optimization of a Manufacturing Process of Parenteral

59

Approved 10 g/vial lyo cycle Approved 5 g/vial lyo cycle

Time

(hr:min)

Temp

(°C)

Vacuum

(µbar)

Time

(hr:min) Temp (°C)

Vacuum

(µbar)

Freezing

6:00 -20

-

8:00 -20

6:00 -45 5:00 -50

-

3:00 -45 4:00 -50

Table 7. Approved freezing phase parameters.

In terms of the lyophilisation cycle, Product Y, USP 10 g/vial recipe presentation is considered the

worst-case, having the double of the fill volume (and the same container than in Product Y 5 g/vial)

and higher stress and impact on the Primary Package, namely the vial itself. However, the 10 g/vial

presentation shows not suffer from breakage as the 5 g/vial.

Upon reviewing the Lyophilisation Cycle for both strengths; it was found out that the 10 g/vial has a

gradual freezing pattern in the cycle, while the 5 g/vial lacked this, but rather it was a sudden and

much fast freezing without any intervals.

It was extrapolated that by applying the same freezing rates and pattern to the 5 g/vial this vial

breakage will be eliminated, and the frozen matrix will behave similarly to the 10 g/vial (as both share

exactly the same formulation).

Since both formulations share the same bulk solution, container closure system – both are filled in a

100mL molded vials- and they only differ in the fill volume it was concluded during the Risk Analysis

that the adaption of the freezing recipe of Product Y 10 g/vial in the recipe of Product 5 g/vial has no

impact in the quality of the product and manufacturing process.

The modifications performed in the freezing step intend to decrease the impact of lyophilisation on the

rejects quantity. No changes were made to the rest of the recipe, primary and secondary drying, for

Product Y for Injection, USP 5 g/vial.

Page 60: Optimization of a Manufacturing Process of Parenteral

60

Approved 10 g/vial lyo cycle Approved 5 g/vial lyo cycle Improved 5 g/vial lyo cycle

Time

(hr:min)

Temp

(°C)

Vacuum

(µbar)

Time

(hr:min)

Temp

(°C)

Vacuum

(µbar)

Time

(hr:min)

Temp

(°C)

Vacuum

(µbar)

Loading - +5 - - +5 - - +5 -

Freezing

6:00 -20

-

6:00 -20

-

8:00 -20 8:00 -20

6:00 -45 5:00 -50

-

6:00 -45

3:00 -45 4:00 -50 3:00 -45

Primary

Drying

00:30 -35

100

2:00 -20

770

2:00 -20

770

3:00 -35 12:00 -20 12:00 -20

3:00 -30 4:00 -10 4:00 -10

6:00 -30 4:00 -10 4:00 -10

5:00 -10 3:00 0 3:00 0

18:00 -10 19:00 0 19:00 0

4:00 0 3:00 +10 3:00 +10

35:00 0 2:00 +10 2:00 +10

2:00 +15 2:00 +15

2:00 +15 2:00 +15

Secondary

Drying

4:00 +25 100 2:00 +20

520

2:00 +20

520

00:30 +25

50

3:00 +20 3:00 +20

16:00 +25 2:00 +25 2:00 +25

1:00 +35 3:00 +25 3:00 +25

19:00 +35 2:00 +35 2:00 +35

8:00 +35 8:00 +35

Table 8. Lyophilisation cycle parameters comparison for Product Y 5 g/vial (Current and Improved

Freezing Phase) and 10 g/vial.

Every time an ANDA holder needs to do a modification in a manufacturing process already approved it

is necessary to report to the authorities. Depending on the type of change, the applicant must notify

FDA about the change in a supplement or by inclusion of the information in the annual report to the

application.

There are two types of moderate change:

Page 61: Optimization of a Manufacturing Process of Parenteral

61

- One type of moderate change requires the submission of a supplement to FDA at least 30 days

before the distribution of the drug product made using the change “Changes Being Effected in 30

Days” ("CBE-30").

- CBE-0 is a moderate change for which distribution of the product can occur when FDA receives the

supplement.

To do the modification in the lyophilization recipe it was necessary to submit a "Changes Being

Effected" ("CBE-0") for FDA.

After all the changes were made, an evaluation of 100% inspection was done and compared with

another PV batch of Product Y 5 g/vial that was lyophilised using the approved freeze-drying cycle

before the improvement.

APPROVED LYO CYCLE AS

PER LYC018

IMPROVED FREEZING

PHASE CYCLE

Total vials 4779 4980

Total of broken/cracked vials rejected 652 36

% of total of broken/cracked vials rejected 13.6 0.7

Table 9. Side-by-side Inspection results of batch of Product Y for Injection USP 5 g/vial.

From the evaluation of the analytical data presented from the manufactured batches of Product Y for

Injection USP 5 g/vial, no negative impact is observed on the product quality after the freeze-drying

phase cycle improvement. As it can be observed from Table 9, it was noticed a decrease of cracked

vials from 13.6% to 0.7% in the capping loading process.

7.5. Line Transfer and Scale-up for Product 5 g/vial

– a Process Validation

After the successful improvements, it was further suggested a Line Transfer from Line 1 to Line 9 and

a scale-up from 170 L to 310L batch size of Product Y for Injection USP 5 g/vial for US Market. For

that, it was necessary to perform a Process Validation Protocol to define the activities and sampling

requirements intended to evaluate and validate the scale-up and the Line Transfer.

In order to validate the scale-up process, three Process Validation (PV) batches of 310 L of Product Y

for Injection USP 5 g/vial were manufactured at Line 9 in Hikma Farmacêutica S.A. One PV batch was

Page 62: Optimization of a Manufacturing Process of Parenteral

62

for submission purpose and, upon approval of the process two additional PV batches were

manufactured, in order to complete the process validation activities.

After the filling the PV batches, the vials were submitted to 100% inspection for particles and defects.

All activities required for full qualification of facilities, equipment and systems to be used on the

process, were previously made and approved. All batches were tested using validated analytical

methods and, raw-materials and primary package components were analysed and released before

production.

Hikma Farmacêutica, S.A validation program includes the validation of processes used in

manufacturing pharmaceutical products. In the case of Product Y for Injection USP 5 g/vial, this

includes compounding of the bulk solution, pre-filtration through a 1.2µm filter, filtration through

sterilizing 0.2µm filter, filling, lyophilisation, stoppering, capping and 100% visual inspection. The

filtered bulk solution was filled into a 100 mL vial presentation.

The critical process steps in the manufacture of Product Y for Injection USP 5 g/vial were evaluated

based on Risk Management Tools (FMECA) as per the following rational:

7.5.1. Process Evaluation Activities and Results

The following activities were performed during the production of all validation batches in order to

define certain process details and to guarantee that the manufacturing process is suitable for the

production of Product Y 5 g/vial at Line 9 with a batch size of 310 L.

Evaluation of quality of the compounded bulk solution

The range of mixing speeds and processing durations used were evaluated during compounding to

assure that complete dissolution occurs under all approved compounding conditions, dissolution

should be complete, and the solution should be uniform. Different speeds were used in the three

batches based on previous experience with similar products compounded using the same tanks.

After the final mixing step was complete, samples from the top and bottom of the compounding vessel

were taken for physiochemical analysis and from the bottom for microbiological analysis.

It was observed a high dissolution time during the compounding, studies are being performed by R&D

department in order to optimize the compounding step and the compounding parameters needs to be

re-evaluated on the next commercial batches.

Evaluation of effects of elapsed compounding (Bulk holding time)

Page 63: Optimization of a Manufacturing Process of Parenteral

63

Although this holding time was previously established as 24h in the preparation tank, this holding time

was re-evaluated at cold conditions (8ºC – 12ºC) until 48h after transference process.

The bulk solution of one batch presentation was evaluated for the maximum bulk holding time in the

preparation tank. Samples were taken from the bottom of the preparation tank at 0h, 18h, 24h and 48h

after the completion of transference activities, for IPC and HPLC and samples for microbiological

testing were collected at 0h, 24h and 48h.

Based on results obtained from one batch, it can be concluded that a bulk holding time of 48 hours

(with transference activities) has no negative impact on the product quality and the product from both

presentations can stay up to 48h in the transference tank from the end of compounding until the end of

filling.

Evaluation of the effects of initial set up (Dead Volume) and line stoppages

An initial quantify of filled units is typically discarded as part of initial line set up. The purpose of this

study was to establish the minimum amount of product that needs to be discarded prior to start filling.

For this reason, samples were collected from each batch before staring the filling, and the system was

purged.

The results obtained on second batch, Product Y (%), pH and related substances are within

specifications and aligned with the finished product results. Therefore, it can be concluded that no

impact is observed on product quality after purging the system with 300 mL and no further actions are

required.

To establish acceptable line stoppage duration and determine the minimum quantity of units to be

discarded prior to re-start filling, one line stoppage of a minimum of two hours was incorporated into

the filling. Samples were collected for analysis and it was observed that the first vials from stage#1

were already within specifications and aligned with finished product results. Therefore, it can be

concluded that line stoppages up to 2h have no impact on the quality of the product, and no further

actions are required.

Evaluation of the effects of filtration and filling on the quality of the

compounded solution

To evaluate the filtration and the filling uniformity, filled units were collected from the beginning, middle

and end of the filling process for testing and every hour for visible particles. The beginning of the

process is defined as a point in which the filtered bulk solution is no longer discarded and line flush

and set up activities have been completed. Sampling activities are designed to validate the entire

filling process.

Page 64: Optimization of a Manufacturing Process of Parenteral

64

From the manufacturing process and in-process controls it can be concluded that all samples met in-

process physiochemical criteria. The filling machine is capable of filling 30.5 mL in 100 mL vials within

the specified parameters for the 310L batch size and the filling process does not affect the product

quality. Additionally, results of beginning, middle and end obtained after lyophilisation are within the

acceptance criteria hence assuring the quality of the filled solution.

Evaluation of the filling machine speed

The maximum and minimum filling machine speed were challenged in order to set a filling speed

range. Filled units were collected at the minimum and maximum filling process speed from the filler

and from the evaluation performed, it can be concluded that the filling machine is capable of filling

30.5mL in 100 mL vials sizes within specification, with a filling machine speed between 40 vials/min

and 70 vials/min.

Evaluation of the holding time between end of API addition and beginning of

Lyophilisation cycle

This holding time is covered by bulk holding time studies in the transference tank performed and, the

holding time between the end of API addition and the beginning of lyophilisation cycle, (freezing

phase) for Product Y for Injection USP, 5 g/vial was 26h 35min for one batch. However, the holding

time between the end of API addition and the beginning of lyophilisation cycle (freezing phase)

already evaluated and considered validated was 24h, consequently the holding time was exceeded

in 02h 35min. Based on the investigation performed, it can be concluded that the present deviation

has no negative impact on the quality of the product of this batch, since from finished product

analytical and microbiologic results are within specification and no out of trend was detected.

Meanwhile, bulk holding time studies performed after transference activities was re-evaluated on

another different batch. This holding time covers the maximum time that the product withstands in

the transference tank (from end of compounding until the end of filling) without affecting the product

quality. The maximum time qualified as part of bulk holding time study is also applicable to determine

the maximum time between end of API addition and the beginning of the lyophilisation cycle as a

worst-case, covering and controlling the total time which the product can stay in the liquid state.

Evaluation of the quality of the Finished Product

Capped vials from the lyophilised product were also taken from the beginning, middle and end of the

process validation batches for physiochemical and microbiological testing as per finished product

criteria. With the results analysis of all batches, it can be concluded that the manufacturing process for

Page 65: Optimization of a Manufacturing Process of Parenteral

65

the 310L batch size of Product Y for Injection USP, 5 g/vial is capable to produce consistently a

product complying with defined specifications.

Evaluation of the effects of Lyophilisation on the quality of the filled vials and

lyophilisation uniformity

The process validation batches were lyophilised as per lyophilisation cycle in SOP#LYC018.

Lyophilizer from Line 9 was considered the worst case from a loading perspective because the loading

and unloading at Line 9 is automatic while in Line 1 is manual despite reduces aseptic handling. Both

lines have similar lyophilizers however the controls and also the condensers capacity are different.

To evaluate the quality and uniformity of lyophilisation, vials were collected from the freeze-dryer for

analysis (appearance of the cake, reconstitution time and water content) from the indicated positions

presented on Figure 15 and properly labelled accordingly to correlate the results with the position in

the lyophilizer.

Notes:

a) Each batch will fill approximately

10 shelves per lyophilizer.

b) Samples from three shelves per

lyophilizer should be

collected/tested.

c) If the last shelf is not fully loaded

collect samples from the previous

one.

d) All vials must be capped

(manually or using the capper).

Figure 18- Loading positions for PV batches and samples collection.

Based on the results evaluated from samples taken from several lyophilizer positions, it can be

observed that all results are within specifications and aligned between them, which means that there is

uniformity within lyophilizer. Additionally, the water content results show that the lyophilisation cycle is

efficient, properly drying the product. Moreover, the water content results for finished product obtained

are well below the acceptance criteria and aligned when compared with the results of the previous

manufactured batches of Product Y for Injection USP, 5 g/vial with 170L batch size in Line 1.

Middle shelf

First shelf

Last Shelf

Page 66: Optimization of a Manufacturing Process of Parenteral

66

Despite the lyophilizer from Line 1 and Line 9 are different as also the controls and the capacity it was

possible to confirm with the results from this evaluation that there is uniformity within lyophilizers.

Evaluation of 100% inspection

Lyophilised vials from the process validation batches were submitted to 100% visual inspection for

defects and particles after being capped. The 100% inspection is executed using the automatic

inspection machine. From the results, it can be concluded that the percentage of total number of vials

rejected with defects is between 0.1% and 0.6% of the total number of vials submitted to capping and

100% inspection showing all results are conforming.

Successful process validation studies provide evidence that a defined manufacturing procedure will

consistently yield product meeting its pre-determined specifications and quality characteristics. Each

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

From the manufacturing process and in-process controls (beginning, middle and end) and from

finished product testing and side by side comparison (Attachment 4), it can be concluded that the

process is capable of producing a product within specifications with scale up from 170L to 310L and

line transfer from Line 1 to Line 9.

After the lyophilisation during the PV activities for the Line Transfer and scale-up it was analyzed the

number of broken vials (Table 11).

B# 1 B# 2 B# 3 B# 4

% of total of broken/cracked

vials rejected 0,04% 0.04% 0,08% 0,2%

Table 11. Percentage of cracked vials reject after Scale-up and Line Transfer.

After scale-up and Line Transfer activities it was possible to verify that all changes done before in the

lyophilisation cycle were successful and without negative impact in the number of broken vials.

Additionally, the quality of the final product was kept. In short, the results shown that we are facing a

robust and validated process who is reproducible in Line 9 with a batch size of 310 L and possible to

launch in the market with high quality.

Page 67: Optimization of a Manufacturing Process of Parenteral

67

Figure 19. Life cycle of Product Y for Injection after Launching.

7.6. Launching

Fill Volume Price

Product Y 5 g/vial 10.52 €/vial

Product Y 10 g/vial 20.45 €/vial

Table 12. Launching for Product Y for Injection USP.

The Product Y 10 g/vial is valued as 20.45 € per vial and the presentation of Product Y 5 g/vial as

10.52 €/vial.

After the improvement made in Product Y 5 g/vial it was possible to reduce the percentage of

breakage vials from 13.6% to 0.7% which means a decrease in 12,9% of breakage vials per batch.

The theoretical value per batch is as described in Table 12.

Line Minimal Quantity (units) Maximum Quantity (units)

1 5 573 5 573

9 10 163 10 163

Table 13. Approved Batch Sizes for Product Y for Injection, USP 5 g/vial.

After all the modifications, it was measured the percentage of good vials per theoretical batch that was

possible to rise up and calculated the value possible to monetize per batch: 7 563€ for Line 1 and

Launching(Product Y)

5 g/vial

10 g/vial

No problemsdetected

Validatedprocess

2019

High BreakageRate

US Market

Problem: FastFreezing

New LyophilizationRecipe adapted

CBE 0

Line Transferand Scale-up

Line

1

Line

9CBE 30

Page 68: Optimization of a Manufacturing Process of Parenteral

68

13 792€ for Line 9. In one year if we produce 25 batches in Line 1 and 25 batches in Line 9 it is

possible to increase for 533 875€ per year.

For the future, one Line will be dedicated for Product Y and if in half-year (26 weeks) it is produced

Product Y 5 g/vial two times per week in Line 9, for example, with this modification it will be possible to

improve in 717 184€.

The values represented above do not represent a significant impact for the company. However, with

the improvement of Product Y 5 g/vial it was possible to validate the manufacturing process and

consequently launch the product for the US Market.

8. Conclusion

Before a drug product reach the market and start to be routinely manufactured, it passes through

several steps, from R&D to normal production. During pharmaceutical development, the product and

the manufacturing process are designed to manufacture a product, which is intended to be use in

accordance with regulatory authorities’ requirements and specifications.

When a product and his process are developed, several activities need to be evaluated to ensure that

the process can be reproducibly and consistently to obtain a product with good attributes and desired

quality.

Technology transfer implicates transfer of product and process knowledge to achieve the

manufacturing process for commercialization and it includes all the activities required since the

pharmaceutical development (R&D) to production, for new products, or from one manufacturing site to

another.

Process validation is part of technology transfer and is applied to demonstrate that the manufacturing

process developed, operated within established parameters, can consistently deliver the intended

product. For a successful process validation and consequently technology transfer and scale-up, a

proper correlation between critical material attributes, critical quality attributes of the products and

critical process parameters of the equipment must be well-established. While there are numerous

inputs, outputs and controls correlated it is important to have a systematic tool who understands the

connection between product and process, based on quality risk management, to identify and to

evaluate the process validation activities to be performed during technology transfer.

This includes establishing strong quality management systems, obtaining appropriate quality raw

materials, establishing robust operating procedures, detecting and investigating product quality

deviations and maintaining reliable testing laboratories. It helps to prevent the occurrence of

contaminations, mix-ups, deviations and failures and assures that the drug products manufactured

meet their quality standards.

Page 69: Optimization of a Manufacturing Process of Parenteral

69

Some manufacturing procedures, as the lyophilisation of products, have a panoply of controls and

parameters since R&D until the normal routine production, making it more difficult to perform.

When a lyophilised product is manufactured, an appropriate lyophilisation cycle should be developed.

During R&D the cycle development is done however adjustments in cycle times and parameters may

occur when transferring from R&D freeze dryer to an industrial lyophiliser.

In 2019, Hikma Farmacêutica started to produce a new product already submitted to the authorities

and approved by them: Product Y for Injection USP 5 g/vial and Product Y for Injection USP 10 g/vial.

During the PV activities it was noticed a high percentage of broken vials of Product Y 5 g/vial

becoming a process non-robust and invalidated. For the improvement of manufacturing process of this

product it was necessary to do an investigation through the life cycle of this product.

The root cause of the breakage problem was attributed to the fast-freezing rate applied during freezing

phase on the lyophilisation cycle. The fast-freezing makes the product to expand suddenly without

allowing the crystals, for enough time, to be shaped according to the vial size creating pressure on the

edges and causing the bottomless breakage of the glass.

During the investigation it was observed that the problem with the breakage rate was noticed in the

past with the 10 g/vial form and it was purposed some modifications on the freezing phase.

In order to optimize the lyophilisation cycle of 5 g/vial form and based in a Risk Assessment it was

purposed to adapt the freezing recipe of 10 g/vial in the recipe of 5 g/vial. Changes were made and

the improvement of lyophilisation cycle, namely, the freezing phase was successfully and it was

possible to decrease the percentage of vial breakage from 13.6% to 0.7% of vials per batch becoming

a validated process and therefore open the possibility to launch the product in the US Market.

After the successful improvement, a Line Transfer from Line 1 and Line 9 and a scale-up to 310 L

were proposed and it was necessary to perform a Process Validation where the critical steps were

analysed. The PV results showed no negative impact resulting in an effective Line Transfer and scale-

up.

The development and optimization of a manufacturing process of a Lyophilised product can be

sometimes a “tricky and complex” question. The development phase, of routine production as wells as

the scale-up and technology transfer of lyophilisation processes, remain challenging.

In short, it is also important to have an effective tool to collect data to be able to improve based on

past experiences.

Page 70: Optimization of a Manufacturing Process of Parenteral

70

8.1. Future work

The manufacturing of injectable pharmaceutical product, in particular lyophilized products, is a

complex process. There are many different operations with several parameters and each unit

operation must be properly validated and controlled in order to assure obtaining a sterile drug product

in accordance with the specifications and regulatory authorities.

Nowadays, many pharmaceutical companies dismiss many tests with trial and error process and

consequently time and money expenditure. For the future, mainly at Hikma, it will be interesting if

other applications should be considered as an opportunity to ease and improve the process.

Process Analytical Technology (PAT) is a mechanism to provide a higher degree of process control I

which analyzes, designs and control pharmaceutical manufacturing processes through the

measurement of Critical Process Parameters (CPP) which affect Critical Quality Attributes (CQA). It

allows real-time monitoring and control to adjust the processing conditions so that the output remains

constant, being more efficient in testing while at the same time reducing over-processing, enhancing

consistency and minimizing rejects. PAT can be used in the pharmaceutical industry, both for process

validation activities and routine process control.

Reduction of time in production cycles, avoid waste during the manufacturing process, enables real-

time process control facilitating continuous processing, improves efficiency and variability

management are some of the advantages of PAT which can represent a benefit over other

pharmaceutical companies that do not apply this technology.

Although, the implementation of a PAT system is a large financial investment, which requires skilled

and skilled human resources, process changes, equipment modification and the operation of the

pharmaceutical units.

Hikma Farmaceutica, S.A. does not make use of PAT tools in manufacturing processes. The

processes are carried out using the most traditional methods and the production is done in batches.

However, it would be interesting in future terms, to implement PAT and develop a more in-depth study

of the following aspects:

- Trend analysis of the results and deviations observed during commercial batches and

validation batches to identify the most frequent problems and identifying improvement

opportunities and their potential benefits;

- Determining causes that lead to the current situation and possible opportunity areas;

- Defining the variables that may impact on the process and the extent they do;

- Running experimental trials to control or optimize variables;

- Defining measures that allow us to maintain consistently trough time achieved improvements;

- Examination about which could be the most necessary PAT tools to implement at Hikma

Farmacêutica.

Page 71: Optimization of a Manufacturing Process of Parenteral

71

9. References

Abdelwahed, Wassim, Ghania Degobert, Serge Stainmesse, and Hatem Fessi. 2006. “Freeze-Drying

of Nanoparticles : Formulation , Process and Storage Considerations ☆” 58: 1688–1713.

https://doi.org/10.1016/j.addr.2006.09.017.

Adelaide, Edinara, Rubens Maciel, Eduardo Coselli, and Vasco De Toledo. 2004. “Freeze Drying

Process : Real Time Model and Optimization” 43: 1475–85.

https://doi.org/10.1016/j.cep.2004.01.005.

Ahir, Keyur B, Khushboo D Singh, Sushma P Yadav, Hetal S Patel, and Chetan B Poyahari. 2014.

“Review Article Overview of Validation and Basic Concepts of Process Validation.” Department of

Quality Assurance, SSR College of Pharmacy, Sayli, Silvassa, India 3 (2): 178–90.

Deluca, P., and L. Lachman. 1965. “Lyophilisation of Pharmaceuticals IV.” Journal of Pharmaceutical

Sciences 54 (10): 1411–15. https://doi.org/10.1002/jps.2600541004.

Ellab Validation Solutions. 2018. “The Freeze Drying Theory and Process - Things to Consider,” 1–16.

https://www.ellab.com/Files/Files/White-Papers/The-Freeze-Drying-Theory-and-Process_ellab-

whitepaper.pdf.

European Medicines Agency. Committee for Medicinal Products for Human Use (CHMP) Committee

for Medicinal Products for Veterinary Use (CVMP) Guideline on Process Validation for

Finished Products - Information and Data to Be Provided in Regulatory Submissions. 2016.

Food and Drug AdministrationFood and Drug Administration. Guidance for Industry Process

Validation: General Principles and Practices. 2011.

Franks, Felix. 1998. “Freeze-Drying of Bioproducts: Putting Principles into Practice.” European Journal

of Pharmaceutics and Biopharmaceutics. https://doi.org/10.1016/S0939-6411(98)00004-6.

Gan, K H, R Bruttini, O K Crosser, and A I Liapis. 2005. “Freeze-Drying of Pharmaceuticals in Vials on

Trays : Effects of Drying Chamber Wall Temperature and Tray Side on Lyophilisation

Performance” 48: 1675–87. https://doi.org/10.1016/j.ijheatmasstransfer.2004.12.004.

Ge Jiang, et al. “Mechanistic Studies of Glass Vial Breakage for Frozen Formulations. I. Vial Breakage

Caused by Crystallizable Excipient Mannitol.” PDA Journal of Pharmaceutical Science and

Technology, vol. 61, no. 6, Nov. 2007, pp. 441–451, journal.pda.org/content/61/6/441.

Accessed 31 Oct. 2019.

Heller, Martin C, John F Carpenter, and Theodore W Randolph. 1997. “Manipulation of Lyophilisation-

Induced Phase Separation : Implications For Pharmaceutical Proteins” 7938 (97): 590–96.

Hottot, Aurélie, Séverine Vessot, and Julien Andrieu. 2007. “Freeze Drying of Pharmaceuticals in Vials:

Influence of Freezing Protocol and Sample Configuration on Ice Morphology and Freeze-Dried

Page 72: Optimization of a Manufacturing Process of Parenteral

72

Cake Texture.” Chemical Engineering and Processing: Process Intensification 46 (7): 666–74.

https://doi.org/10.1016/j.cep.2006.09.003.

Kasper, Julia Christina, Gerhard Winter, and Wolfgang Friess. 2013. “European Journal of

Pharmaceutics and Biopharmaceutics Recent Advances and Further Challenges in

Lyophilisation.” EUROPEAN JOURNAL OF PHARMACEUTICS AND BIOPHARMACEUTICS.

https://doi.org/10.1016/j.ejpb.2013.05.019.

Nail, Steven L, Shan Jiang, Suchart Chongprasert, and Shawn A Knopp. 2002. “Fundamentals of

Freeze-Drying.”

Nandhakumar, Dharmamoorthy. 2013. “Available through Online” 5 (1): 2446–64.

Nireesha, G R, L Divya, C Sowmya, N Venkateshan, M Niranjan Babu, and V Lavakumar. 2013.

“Lyophilisation / Freeze Drying - An Review” 3 (4): 87–98.

Patel, Sajal M, Takayuki Doen, and Michael J Pikal. 2010. “Determination of End Point of Primary

Drying in Freeze-Drying Process Control” 11 (1). https://doi.org/10.1208/s12249-009-9362-7.

Patel, Viral Kumar Kishor Bhai, and Anil M. Pethe. 2010. “Process Validation: An Essential Process in

Pharmaceutical Industry.” Pharmaceutical Reviews 8 (4). https://doi.org/10.7439/ijasr.v1i4.1781.

Rogers, Everett M., Shiro Takegami, and Jing Yin. 2001. “Lessons Learned about Technology

Transfer.” Technovation 21 (4): 253–61. https://doi.org/10.1016/S0166-4972(00)00039-0.

Sandle, Tim. “Aseptic Processing - an Overview | ScienceDirect Topics.” Sciencedirect.Com, 2011,

www.sciencedirect.com/topics/agricultural-and-biological-sciences/aseptic-processing.

Accessed 29 Oct. 2019.

Sastri, Process Validation for Medical Device Manufacturers and Their Suppliers Vinny R. “Process

Validation - an Overview | ScienceDirect Topics.” Sciencedirect.Com, 2014,

www.sciencedirect.com/topics/engineering/process-validation. Accessed 29 Oct. 2019.

Sharma, Sumeet, and Gurpreet Singh. “PROCESS VALIDATION IN PHARMACEUTICAL INDUSTRY;

AN OVERVIEW.” Journal of Drug Delivery and Therapeutics, vol. 3, no. 4, 15 July 2013,

jddtonline.info/index.php/jddt/article/view/582, 10.22270/jddt.v3i4.582. Accessed 29 Oct. 2019.

Snyder, Meg. “Lyophilization: The Basics - Drug Discovery and Development.” Drug Discovery and

Development, 7 Mar. 2017, www.drugdiscoverytrends.com/lyophilization-the-basics/.

Accessed 29 Oct. 2019.

Shukla, Soham. 2011. “Freeze Drying Process: A Review.” International Journal of Pharmaceutical

Sciences and Research 2 (12): 3061–68.

Tang, Xiaolin Charlie, and Michael J Pikal. 2004. “Design of Freeze-Drying Processes for

Pharmaceuticals : Practical Advice” 21 (2).

Taylor, Publisher, H Sadikoglu, M Ozdemir, M Seker, H Sadikoglu, M Ozdemir, and M Seker. 2007.

“Drying Technology : An International Journal Freeze-Drying of Pharmaceutical Products :

Research and Development Needs Freeze-Drying of Pharmaceutical Products : Research and

Page 73: Optimization of a Manufacturing Process of Parenteral

73

Development Needs,” no. November 2012: 37–41. https://doi.org/10.1080/07373930600734018.

.World Health Organization. “WHO Expert Committee on Specifications for Pharmaceutical

Preparations - WHO Technical Report Series, No. 863 - Thirty-Fourth Report: Annex 6 - Good

Manufacturing Practices: Guidelines on the Validation of Manufacturing Processes: Introduction.”

Who.Int, 2018, apps.who.int/medicinedocs/en/d/Js5516e/16.html. Accessed 29 Oct. 2019.

York, New, et al. Pharmaceutical Process Scale-Up. 2001.

Page 74: Optimization of a Manufacturing Process of Parenteral

74

10. Appendix

Attachment 1- Risk Assessment and Process Validation Approach of the batch for submission of Product Y for Injection USP 5 g/vial and Product Y for

Injection USP 10 g/vial (First Submission Batch).

Process Risk Assessment

Evaluation (ID) Validation Approach Rationale Necessary validation activities

Evaluation of quality of the

compounded bulk solution From A to E

The two presentations have the same bulk formulation but different batch sizes

(40 L and 80 L) One batch of each size

Evaluation of the effects of elapsed

compounding (Bulk hold time) N/A

The two formulations have the same bulk formulation. The batch size parameter

has no impact for this evaluation One batch of any presentation

Evaluation of the effects of filling on

the quality of the compounded

solution

E The two presentations will be filled in 100 mL vials with different fill volumes One batch of any presentation

Dead Volume and Line Stoppages

(up to 2h) N/A

The two presentations will be filled in 100 mL vials. The fill volume has no impact

for this evaluation

One batch of Product Y for

Injection USP 10 g/vial

Holding time between end of API

addition and beginning of

lyophilisation cycle (loading phase)

N/A

The two presentations will have different holding time between end of API

addition and beginning of Lyophilisation cycle due to the different batch size.

Since the vials will be loaded in cool shelves (5 ºC) it could be considered for the

Holding time study the actual time the vials are loaded

One batch of any presentation

Page 75: Optimization of a Manufacturing Process of Parenteral

75

Evaluation of the effects of

lyophilisation on the quality of the

filled Vials

G

The two presentations will be filled in 100 mL vials with different fill volumes and

different lyophilisation cycles will be used. After lyophilisation the lyophilised FP

specifications are not the same

One batch of any presentation

MICRO Validation N/A

The two presentations have the same bulk formulation. Samples for Sterility and

Endotoxins Validation should be collected for (FP) and samples for Bioburden

and Bacterial Endotoxins should be collected for Bulk solution

One batch of any presentation

Filter Validation N/A The two presentations have the same product formulation One batch of any presentation

Page 76: Optimization of a Manufacturing Process of Parenteral

76

Attachment 2 - Risk Assessment and Data Compilation Approach with the batches used to qualify the Product Y for Injection, USP 5 g/vial and 10 g/vial

Process

Risk

Assessment

Evaluation (ID)

Validation Approach Rationale

Necessary

validation

activities

Batches evaluated

Evaluation of quality of

the compounded bulk

solution

From A to E The two presentations have the same bulk formulation but

different batch sizes (40 L and 80 L)

One batch of each

size

One batch of Product Y for Injection USP 5

g/vial

One batch of Product Y for Injection USP 10

g/vial

Evaluation of the

effects of elapsed

compounding (Bulk

hold time)

N/A The two formulations have the same bulk formulation. The

batch size parameter has no impact for this evaluation

One batch of any

presentation

One batch of Product Y for Injection USP 10

g/vial

Evaluation of the

effects of filling on the

quality of the

compounded solution

E The two presentations will be filled in 100 mL vials with

different fill volumes

One batch of any

presentation

One batch of Product Y for Injection USP 5

g/vial

One batch of Product Y for Injection USP 10

g/vial

Dead Volume and Line

Stoppages (up to 2h) N/A

The two presentations will be filled in 100 mL vials. The fill

volume has no impact for this evaluation

One batch of

Product Y for v USP

10g/vial

One batch of Product Y for Injection USP 10

g/vial

Holding time between

end of API addition and

beginning of

lyophilisation cycle

N/A

The two presentations will have different holding time

between end of API addition and beginning of

lyophilisation cycle due to the different batch size. Since

the vials will be loaded in cool shelves (5 ºC) it could be

considered for the Holding time study the actual time the

One batch of any

presentation

One batch of Product Y for Injection USP 5

g/vial

One batch of Product Y for Injection USP 10

g/vial

Page 77: Optimization of a Manufacturing Process of Parenteral

77

(loading phase) vials are loaded

Evaluation of the

effects of lyophilisation

on the quality of the

filled Vials

G

The two presentations will be filled in 100 mL vials with

different fill volumes and different lyophilisation cycles will

be used. After lyophilisation the lyophilised FP

specifications are not the same

One batch of any

presentation

One batch of Product Y for Injection USP 5

g/vial

One batch of Product Y for Injection USP 10

g/vial

MICRO Validation N/A

The two presentations have the same bulk formulation.

Samples for Sterility and Endotoxins Validation should be

collected for (FP) and samples for Bioburden and

Bacterial Endotoxins should be collected for Bulk solution

One batch of any

presentation

One batch of Product Y for Injection USP 10

g/vial

Filter Validation N/A The two presentations have the same product formulation One batch of any

presentation

One batch of Product Y for Injection USP 5

g/vial

Page 78: Optimization of a Manufacturing Process of Parenteral

78

Attachment 3

Product Y for Injection, USP 5g/vial

Flow Diagram for Batch Preparation- Line 9

Excipient 1

Mix until complete homogenization. Keep the temperature (8 – 12ºC).

Check the pH of the solution (2.5 – 4.5).

Mix slowly and under vigorous agitation

assuring its complete incorporation.

Sparging with filtered nitrogen until dissolved

oxygen < 1 ppm.

Decrease the agitation and agitate until

complete dissolution and Sparging between

Stainless Steel

Sparging with filtered nitrogen until dissolved

oxygen ≤ 0.25 ppm.

Product Y, USP

Product Y, USPP

Adjust volume to 100%.

Mix until complete homogenization.

Sparging with filtered nitrogen until dissolved

Deaereted WFI (8-12ºC)

Deaereted WFI (8 –

Start

Notes: Protect the samples from air

exposure, from light (by wrapping with

aluminium foil) and from heat sources.

End

IPC:

-Description

-pH

-Density

QC:

Deaerate WFI for a

minimum of 20 min

(≤ 20ºC)

Cold WFI

Cold WFI (8 – 12ºC)

20L Stainless Steel

Container

Page 79: Optimization of a Manufacturing Process of Parenteral

79

Grade C

20mm red

aluminium Flip-off

Secondary packaging

Capping (under UFH – Grade A air

supply)

Grade C

Compounding in SS

Tank

Pallets preparation

100% Visual inspection for defects

Freeze Dryers

Grade A

Frames loader

Grade A

Product filtration by

Grade C

VHP

Chamber

Filling and Stoppering

Grade C

Pre-filtration by 1.2

µm filter

Shipping

Final filter integrity

test before and

Grade C Area

Grade A Room

Unclassified - Warehouse

Unclassified - Packaging

Area

Unclassified - Warehouse

Active and Inactive

Raw Materials

100ml vials, neck

20 mm, Type I,

Moulded, Clear 20mm Lyo Stoppers

Filling size

parts

Frames

Grade D

Page 80: Optimization of a Manufacturing Process of Parenteral

Attachment 4 – Scale up and Line Transfer evaluation – side by side

A side by side comparison of finished product results was performed between 170L batches manufactured in Line 1 and 310L batch manufactured in Line 9 of Product Y for Injection USP, 5 g/vial in order to evaluate the reproducibility of

results or if any manufacturing constraint related to the scale up process and the line transfer process is observed.

Table A2.1 – Finished Product Results of batches of Product Y for Injection USP 5 g/vial manufactured in Line 1 with 170L batch size vs Line 9 with 310L batch size

Conclusion: From the evaluation of the analytical data presented from the manufactured batches of Product Y for Injection USP 5 g/vial no negative impact is observed on the product quality after scaling up from 170L to 310L batch

size, transferring the manufacturing process from Line 1 to Line 9.

Batch # 1909010.1 Batch # 1909098.1 Batch # 1909099.1 Batch # 1909099.2 Batch # 1901016.1 Batch # 1901015.1 Batch # 1901014.1 Batch # 1901013.1 Batch # 1901012.1

Mfg. Date: January 2019 Mfg. Date: July 2019 Mfg. Date: July 2019 Mfg. Date: July 2019 Mfg. Date: January 2019 Mfg. Date: January 2019 Mfg. Date: January 2019 Mfg. Date: January 2019 Mfg. Date: January 2019

Reference SOP: FPL031 - Rev. G Exp. Date: January 2021 Exp. Date: July 2021 Exp. Date: July 2021 Exp. Date: July 2021 Exp. Date: January 2021 Exp. Date: January 2021 Exp. Date: January 2021 Exp. Date: January 2021 Exp. Date: January 2021

Tests Standard Specifications Results Results Results Results Results Results Results Results Results

1. Description Off-white to tan colored lyophilized powder Conforms Conforms Conforms Conforms Conforms Conforms Conforms Conforms Conforms

2. Identification

2.1. By IR Conforms Conforms Conforms Conforms Conforms Conforms Conforms Conforms Conforms Conforms

2.2. By HPLC Conforms Conforms Conforms Conforms Conforms Conforms Conforms Conforms Conforms Conforms

3. Water (%) NMT 5.0 2.2 2.9 2.6 2.7 1.8 1.6 1.6 1.7 1.5

4. pH 2.5 to 4.5 3.7 3.2 3.5 3.5 3.3 3.6 3.7 3.7 3.7

5. Uniformity of Content (WV)The Acceptance Value of the first 10

dosage units ≤ L1%0.4% 0.7% 0.9% 1.1% 2.9% 0.7% 0.7% 1.0% 1.7%

6. Bacterial Endotoxins ( USP EU/mg) NMT 0.33 < 0.02 < 0.02 < 0.02 < 0.02 < 0.02 < 0.02 < 0.02 < 0.02 < 0.02

7. Sterility Sterile Conforms Conforms Conforms Conforms Conforms Conforms Conforms Conforms Conforms

8. Particulate Matter

8.1. Sub-Visible particles

Ø ≥ 10 µm NMT 6000 particles/Vial 4053 3460 3673 4280 1640 4660 5300 2900 2860

Ø ≥ 25 µm NMT 600 particles/Vial 73 140 60 93 13 73 93 73 40

8.2. Visible Particles Essentially free from visible particle matter Conforms Conforms Conforms Conforms Conforms Conforms Conforms Conforms Conforms

9. Color and clarity of constituted solution

9.1. Color (Abs) ≤ 0.125 0.020 0.016 0.012 0.011 0.029 0.018 0.017 0.012 0.012

9.2. Clarity (T %) ≥ 90 100 100 100 100 100 100 100 100 100

10. Assay (%) 90.0 – 115.0 99.3 101.7 102 101.9 97.2 99.6 98.6 101.6 102.7

11. Vancomycin B (%) NLT 90.0 94.5 94.2 94.7 94.7 94.6 94.8 94.9 95.0 95.2

12. Related substances (%)

EP Impurity A NMT 4.0 0.47 0.59 0.60 0.60 0.47 0.48 0.56 0.62 0.46

EP Impurity B (B1 + B2) NMT 4.0 2.17 2.7 2.13 2.15 2.41 2.10 2.05 2.01 2.09

EP Impurity C NMT 4.0 0.12 0.14 0.12 0.13 0.13 0.10 0.11 0.11 0.10

EP Impurity D NMT 4.0 0.31 0.33 0.31 0.31 0.35 0.26 0.26 0.28 0.28

Other Individual Unspecified Impurity NMT 3.0 0.70 0.66 0.72 0.76 0.53 0.69 0.65 0.62 0.51

Total Impurities NMT 10.0 5.3 5.6 5.2 5.2 5.3 5.1 4.9 4.9 4.5

13. Heavy Metals (ppm) NMT 30 < 30 < 30 < 30 < 30 < 30 < 30 < 30 < 30 < 30

14. Ethanol Content (ppm) NMT 10000 376 1068 1237 937 1334 1170 1125 1477 1301

15. Reconstitution Time (min) NMT 3 2 1 1 1 1 2 2 2 2

16. Residual Solvents Complies Complies w ith USP <467> Option 1 Complies w ith USP <467> Option 1 Complies w ith USP <467> Option 1 Complies w ith USP <467> Option 1 Complies w ith USP <467> Option 1 Complies w ith USP <467> Option 1 Complies w ith USP <467> Option 1 Complies w ith USP <467> Option 1 Complies w ith USP <467> Option 1

17. Elemental Impurities CompliesComplies w ith USP <232> and ICH

Q3D Option 2b

Complies w ith USP <232> and ICH

Q3D Option 2b

Complies w ith USP <232> and ICH Q3D

Option 2b

Complies w ith USP <232> and ICH

Q3D Option 2b

Complies w ith USP <232> and ICH

Q3D Option 2b

Complies w ith USP <232> and ICH

Q3D Option 2b

Complies w ith USP <232> and ICH

Q3D Option 2b

Complies w ith USP <232> and ICH

Q3D Option 2b

Complies w ith USP <232> and ICH

Q3D Option 2b

18. Injections and Implanted Drug products Complies Complies w ith USP <1> Complies w ith USP <1> Complies w ith USP <1> Complies w ith USP <1> Complies w ith USP <1> Complies w ith USP <1> Complies w ith USP <1> Complies w ith USP <1> Complies w ith USP <1>

Product Name: Vancomycin Hydrochloride for Injection, USP

Label Claim: 5.0 g Vancomycin/ vial

170L batch size - Line 1310L batch size - Line 9