design, testing, and scale-up of medical device class iib ... angelica...design, testing, and...

116
Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical industry Angélica Esteves Lopes da Graça Thesis to obtain the Master of Science Degree in Pharmaceutical Engineering Pharmaceutical Engineering Supervisors: Prof. Dr. Joana Marques Marto and Prof. Dr. Jorge Humberto Gomes Leitão Examination Committee Chairperson: Prof. Dr. José Monteiro Cardoso de Menezes Supervisor: Prof. Dr. Joana Marques Marto Members of the Committee: Dr. Diogo Miguel De Sousa Manata and Prof. Dr. Lídia Maria Diogo Gonçalves May 2018

Upload: others

Post on 02-Mar-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

Design, testing, and scale-up of medical device class IIb:

risk analysis as a crucial tool for pharmaceutical industry

Angélica Esteves Lopes da Graça

Thesis to obtain the Master of Science Degree in Pharmaceutical Engineering

Pharmaceutical Engineering

Supervisors: Prof. Dr. Joana Marques Marto and Prof. Dr. Jorge Humberto Gomes

Leitão

Examination Committee

Chairperson: Prof. Dr. José Monteiro Cardoso de Menezes

Supervisor: Prof. Dr. Joana Marques Marto

Members of the Committee: Dr. Diogo Miguel De Sousa Manata and Prof. Dr. Lídia

Maria Diogo Gonçalves

May 2018

Page 2: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

i

Design, testing, and scale-up of medical device class IIb:

the risk analysis as a crucial tool for pharmaceutical

industry

Angélica Esteves Lopes da Graça

Thesis to obtain the Master of Science Degree in Pharmaceutical Engineering

Pharmaceutical Engineering

Supervisors: Prof. Dr. Joana Marques Marto and Prof. Dr. Jorge Humberto Gomes

Leitão

Examination Committee

Chairperson: Prof. Dr. José Monteiro Cardoso de Menezes

Supervisor: Prof. Dr. Joana Marques Marto

Members of the Committee: Dr. Diogo Miguel De Sousa Manata and Prof. Dr. Lídia

Maria Diogo Gonçalves

May 2018

Page 3: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

ii

Abstract Dry eye disease (DED) treatment usually consists in topical administration of eye drop solutions. This

research project was developed to create an effective and safe eye drop solution, containing Hyaluronic

Acid (HA). It was developed two medical devices (MD), an eye drop solution with 0.15% of HA (w/v) and

another with 0.30% of HA (w/v). Pre-formulation studies were performed to select suitable excipients.

After the selection the final formulation and a suitable manufacturing process, it was design a scale-up

study, producing a pilot batch manufacturing process. The pilot batch manufacturing process was

submitted to a Risk Analysis.

Two methods were used: FMEA and the ISO 14971:2007 Qualitative Analysis. After the detention of

possible failure modes, prioritization and corrective actions were applied. FMEA classified 14%

unacceptable risks and it was able to implement corrective actions, while the ISO 14971:2007 method

detected only 11% and it was not capable to implement corrective actions.

A Validation Plan was also performed to study the effectiveness of the manufacturing process. The

process controls performed showed that the results were consistent and respected the defined

specification of each parameter.

In the Product Characterization, measurements of viscosity and mucoadhesive studies were performed.

The viscosity measurements showed that HA 0.30% presented higher viscosity and the viscosity

increased when the mucin interacted with HA, suggesting the existence of interactions. In vitro tests

with ARPE-19 cells demonstrated that both products are promising MD for DED treatment since cell’s

morphology is preserved and the cell viability increased after the product administration.

Key-Words: Dry eye Disease; Hyaluronic Acid; Medical Devices; Risk Analysis; Mucoadhesion.

Page 4: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

iii

Resumo Analítico O tratamento para o olho seco consiste na administração de soluções de conforto. Foi desenvolvido

um projeto para criar soluções eficazes e seguras contendo ácido hialurónico (AH). Decidiu-se investir

em dois produtos, um com concentração de 15% e outro com 30%. Estudos de pré-formulação foram

elaborados para definir os excipientes. Após a conclusão da formulação e do processo de fabrico foi

elaborado um estudo de scale-up onde se desenvolveu o processo de fabrico dos lotes piloto.

Submeteu-se o processo de fabrico a uma Análise de Risco onde se utilizaram duas ferramentas: FMEA

e Análise Qualitativa de acordo com a ISO 14971:2007. Após a detenção dos modos de falha foi

aplicado uma priorização e ações corretivas nas mesmas. A FMEA classificou 14% dos riscos como

inaceitáveis e implementou ações corretivas, a ISO 14971:2007 classificou 11% como inaceitáveis,

contudo não foi capaz de executar ações corretivas.

O objetivo da Validação de Processo foi estudar a eficácia do processo de fabrico. Os controlos de

processo demonstraram consistência nos resultados e conforme com os valores estabelecidos para

cada parâmetro.

Na Caracterização do Produto realizou-se um estudo de viscosidade e de muscoadesividade. Mediu-

se a viscosidade através de viscosímetros, texturómetro e potencial zeta concluindo que o AH 30%

apresenta maior viscosidade e que esta aumenta quando a mucina é adicionada, sugerindo a existência

de interações. Testes in vitro utilizando células ARPE-19 demonstraram que ambos os produtos são

promissores uma vez a morfologia celular é mantida e a viabilidade celular aumenta após a

administração do produto.

Palavras-Chave: Olho Seco; Ácido Hialurónico; Dispositivo medico; Análise de Risco;

Mucoadesividade.

Page 5: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

iv

Agradecimentos Durante a realização desta dissertação muitos desafios foram enfrentados, tanto a nível profissional

como pessoal. Apesar do percurso não ter sido fácil e mais longo e árduo que o esperado, as

dificuldades foram ultrapassadas, na qual chega a hora de expressar os meus sentimentos a todos que

me ajudaram neste caminho e o tornaram mais simples:

À Professora Doutora Joana Marto, minha orientadora principal, pelos conselhos, pela ajuda, pela

constante preocupação e sobretudo pela amizade desenvolvida ao longo deste percurso. Considero-

me uma sortuda por ter tido uma orientadora que não só esteve sempre disponível para me guiar

durante a realização desta dissertação, como esteve sempre disponível para mim;

Ao Professor Doutor Jorge Leitão, pela disponibilidade e pelos conselhos que me facultou;

À Doutora Sara Raposo por me ter dado a oportunidade de elaborar este projeto e pelo convite de

pertencer à equipa Laboratório Edol, Produtos Farmacêuticos S.A.;

Aos Professores Doutor José Cardoso Menezes e Doutor António Almeida como coordenadores do

Mestrado e pela disponibilidade ao longo desta jornada;

Ao Laboratório de controlo microbiológico – LCM, mais precisamente à Dra. Alexandra Nogueira Silva

e à Técnica Paula Machado, pela oportunidade de entrar no mundo da indústria farmacêutica e pelos

conhecimentos desenvolvidos em microbiologia;

À Dra. Rita Carneiro pelos ensinamentos sobre a produção do produto, nomeadamente desde o seu

fabrico até ao enchimento, reforçando desta forma os meus conhecimentos;

Ao Laboratório Edol, Produtos Farmacêuticos S.A. por toda a disponibilização de todos os recursos

necessários à elaboração deste trabalho, com um especial agradecimento aos colaboradores do

departamento de Controlo de Qualidade;

À equipa “Análise de Risco” na qual fez parte o Professor Rui Loureiro e o Dr. Diogo Manata, pelos

ensinamentos aprofundados de análise de risco, pela disponibilidade, pelas ideias e conselhos em

como elaborar um dos mais complexos capítulos desta dissertação;

Às Professoras Doutora Lídia Gonçalves e Doutora Helena Margarida Ribeiro por me terem recebido

nos seus laboratórios, pela simpatia e ajuda que me forneceram;

A todos os membros do “Salão Nobre” do edifício D da Faculdade de Farmácia pela hospitalidade, pela

simpatia e pelo convívio durante a realização da parte laboratorial;

Às meninas da residência universitária Filipe Folque por todo o apoio;

Às minhas irmãs Maria Graça e Iolanda Tomás e aos meus amigos Nuno Costa e Miguel Antunes pelo

incentivo, força e ajuda;

Aos restantes familiares, mais precisamente pais e avós, pelo orgulho que sentem por mim e por o

demonstrarem.

Page 6: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

v

Table of Contents

Abstract..................................................................................................................................................... ii

Resumo Analítico .................................................................................................................................... iii

Agradecimentos ....................................................................................................................................... iv

List of Figures ...........................................................................................................................................x

List of Tables ........................................................................................................................................... xi

Abbreviations .......................................................................................................................................... xii

Aims and Organization of the Dissertation .............................................................................................. 1

Chapter One – Literature Overview ......................................................................................................... 3

1. Theoretical Introduction ................................................................................................................... 3

1.1. Anatomy of the Eye ................................................................................................................. 3

1.2. Structure of the tear film .......................................................................................................... 4

1.2.1. The Lipid Layer ................................................................................................................ 5

1.2.2. The Aqueous Layer ......................................................................................................... 5

1.2.3. The Mucous Layer ........................................................................................................... 6

1.3. Dry Eye Disease ...................................................................................................................... 6

1.3.1. Treatment of Dry Eye Disease ........................................................................................ 8

1.3.1.1. Pharmacological Treatment ..................................................................................... 8

1.3.1.2. Food supplements ................................................................................................... 8

1.3.1.3. Medical Devices ....................................................................................................... 9

1.3.1.3.1. Ophthalmic Comfort Solutions ............................................................................ 10

1.4. Ophthalmic Comfort Solution Characterisation ..................................................................... 11

1.4.1. Mucoadhesion and mucoadhesive properties ............................................................... 11

1.4.1.1. Polymers ................................................................................................................ 12

1.4.1.1.1. Hyaluronic Acid ................................................................................................... 13

1.4.1.2. Factors Important to Mucoadhesion ...................................................................... 13

1.4.1.3. Techniques Used for the Assessment of Mucoadhesion ...................................... 14

1.4.1.4. Mucoadhesion Theories of Polymer Attachment ................................................... 15

1.4.1.4.1. Wetting Theory ................................................................................................... 15

1.4.1.4.2. Diffusion Theory ................................................................................................. 16

1.4.1.4.3. The Mechanical Interlocking Theory .................................................................. 16

1.4.1.4.4. The Electronic Theory ........................................................................................ 16

1.4.1.4.5. The Adsorption Theory ....................................................................................... 16

1.4.2. Evaluation of the Product Efficacy ................................................................................. 16

1.4.2.1. In vitro models ....................................................................................................... 16

1.4.2.1.1. 3D In Vitro Models of the Eye and Ocular Diseases .......................................... 17

1.5. Risk Analysis in Medical Devices .......................................................................................... 19

1.5.1. General Process ............................................................................................................ 20

1.5.2. Initiate Quality Risk Management Process .................................................................... 20

Page 7: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

vi

1.5.3. Risk Assessment ........................................................................................................... 20

1.5.4. Risk Control ................................................................................................................... 21

1.5.5. Risk Communication ...................................................................................................... 21

1.5.6. Risk Review ................................................................................................................... 21

1.5.7. Risk Management Tools ................................................................................................ 21

1.5.8. The Importance of Applying Risk Analysis in Medical Devices ..................................... 22

Chapter Two – Previous Work .............................................................................................................. 23

2. Previous Work - Introduction ......................................................................................................... 23

2.1. Materials and Methods .......................................................................................................... 24

2.1.1. Materials ........................................................................................................................ 24

2.1.2. Methods ......................................................................................................................... 24

2.1.2.1. Appearance ........................................................................................................... 24

2.1.2.2. Determination of the pH values ............................................................................. 25

2.1.2.3. Determination of the Osmolality values ................................................................. 25

2.1.2.4. Determination of the Viscosity values .................................................................... 25

2.1.2.5. Efficacy of Antimicrobial Preservation ................................................................... 25

2.1.2.5.1. Strains and Preparation of Inoculum .................................................................. 25

2.1.2.5.2. Determination of the preservative efficacy of the formulation ............................ 25

2.1.2.6. Stability Testing ..................................................................................................... 26

2.1.2.7. Manufacturing Process .......................................................................................... 26

2.2. Pre-Formulation Studies ........................................................................................................ 26

2.2.1. Formulation Design ....................................................................................................... 26

2.2.2. Required specification ................................................................................................... 26

2.2.3. Selection of Materials .................................................................................................... 27

2.2.3.1. Polymer Selection .................................................................................................. 27

2.2.3.2. Preservative Selection ........................................................................................... 27

2.2.3.3. Buffer Selection ..................................................................................................... 28

2.2.4. Components of the Formulation ................................................................................ 28

2.2.5. Development of the Laboratory Batches ....................................................................... 32

2.2.6. Efficacy of Antimicrobial Preservation ........................................................................... 32

2.3. Final Formulation ................................................................................................................... 32

2.3.1. Polymer .......................................................................................................................... 32

2.3.1.1. Comfort Agents ...................................................................................................... 32

2.3.1.2. Hyaluronic Acid ...................................................................................................... 33

2.3.2. Excipients ...................................................................................................................... 34

2.3.2.1. Highly Purified Water ............................................................................................. 34

2.3.2.2. Potassium Chloride, Magnesium Chloride.6H2O and Calcium Chloride.6H2O ..... 34

2.3.2.3. Boric Acid and Sodium Tetraborate ....................................................................... 35

2.3.2.4. Suttocide (N-hydroxymethylglycinate 50%) and EDTA ......................................... 35

2.4. Manufacturing Process of the Final Formulation ................................................................... 36

Page 8: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

vii

2.5. Stability Testing ..................................................................................................................... 36

2.6. Scale-Up ................................................................................................................................ 36

2.7. Conclusion ............................................................................................................................. 38

Chapter Three – Risk Analysis .............................................................................................................. 39

3. Risk Analysis - Introduction ........................................................................................................... 39

3.1. Materials and Methods .......................................................................................................... 40

3.1.1. Materials ........................................................................................................................ 40

3.1.2. Methods ......................................................................................................................... 41

3.1.2.1. Process Description ............................................................................................... 41

3.1.2.2. Identification of Critical Points ............................................................................... 41

3.1.2.3. Failure Cause and Effect of each Failure Mode .................................................... 41

3.1.2.4. Severity Rating Assignment .................................................................................. 42

3.1.2.5. Occurrence Rating Assignment ............................................................................. 42

3.1.2.6. Detection Rating Assignment ................................................................................ 42

3.1.2.7. Risk Priority Number Calculation ........................................................................... 42

3.1.2.8. Failure Modes Prioritization ................................................................................... 42

3.1.2.9. ISO 14971:2007 Qualitative Analysis .................................................................... 42

3.1.2.10. Corrective Actions .................................................................................................. 43

3.1.2.11. Risk Review ........................................................................................................... 43

3.2. Results ................................................................................................................................... 43

3.2.1. Ishikawa diagrams ......................................................................................................... 43

3.2.2. FMEA of Hyaluronic Acid Pilot Batch Manufacturing Process ...................................... 43

3.2.3. Failure Modes Prioritization and Corrective Actions...................................................... 48

3.2.4. Risk Review ................................................................................................................... 49

3.3. Discussion ............................................................................................................................. 50

3.4. Conclusion ............................................................................................................................. 52

Chapter Four – Pilot Scale Batches Process Validation ....................................................................... 53

4. Pilot Scale Batches Process Validation - Introduction .................................................................. 53

4.1. Materials and Methods .......................................................................................................... 53

4.1.1. Materials ........................................................................................................................ 53

4.1.2. Methods ......................................................................................................................... 53

4.1.2.1. Production of three batches of Hyaluronic Acid 0.15% and HA 0.30% ................. 53

4.1.2.2. Appearance ........................................................................................................... 53

4.1.2.3. Determination of the pH values ............................................................................. 54

4.1.2.4. Determination of the Osmolality values ................................................................. 54

4.1.2.5. Determination of the Viscosity values .................................................................... 54

4.1.2.6. Determination of the Density values ...................................................................... 54

4.1.2.7. Bioburden .............................................................................................................. 54

4.1.2.8. Sodium Hyaluronate Assay ................................................................................... 54

4.1.2.9. Sterility Test ........................................................................................................... 54

Page 9: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

viii

4.2. Critical Steps to Control ......................................................................................................... 55

4.2.1. Acceptance Criteria ....................................................................................................... 56

4.2.2. Validation Plan ............................................................................................................... 56

4.2.2.1. Preparation of HA 0.15% and HA 0.30% Eye Drops Solution .............................. 56

4.2.2.2. In Process Control and Holding Time Validation of HA 0.15% and HA 0.30% eye

drops 56

4.2.2.3. Control of the Sterilizing Filtration .......................................................................... 56

4.2.2.4. Filling Control ......................................................................................................... 56

4.2.2.5. Finished Product Control ....................................................................................... 57

4.3. Results ................................................................................................................................... 58

4.4. Discussion ............................................................................................................................. 59

4.5. Conclusion ............................................................................................................................. 60

Chapter Five – Product Characterisation .............................................................................................. 61

5. Product Characterization - Introduction ......................................................................................... 61

5.1. Material and Methods ............................................................................................................ 61

5.1.1. Material .......................................................................................................................... 61

5.1.2. Methods ......................................................................................................................... 61

5.1.2.1. Viscosity Measurements ........................................................................................ 61

5.1.2.1.1. Brookfield viscometer ......................................................................................... 61

5.1.2.2. Mucoadhesion studies ........................................................................................... 61

5.1.2.2.1. Ostwald viscometer ............................................................................................ 62

5.1.2.2.2. Rotational Rheometer......................................................................................... 62

5.1.2.2.3. Zeta Potential ..................................................................................................... 63

5.1.2.3. In Vitro Assay ......................................................................................................... 63

5.1.2.3.1. Cell Culture Condition......................................................................................... 63

5.1.2.3.2. Cell Viability of HA 0.15% and HA 0.30% .......................................................... 63

5.1.2.3.3. 2D model - Evaluation of Cell Morphology and Cell Viability After Dehydration 63

5.1.2.3.4. 3D model - Dry Eye Model and Cell Viability...................................................... 64

5.1.2.4. Statistical Data Analysis ........................................................................................ 64

5.2. Results ................................................................................................................................... 64

5.2.1. Viscosity Measurements of HA 0.15% and HA 0.30% .................................................. 64

5.2.2. Mucoadhesive Studies .................................................................................................. 65

5.2.2.1. Viscosity Measurements ........................................................................................ 65

5.2.2.2. Rheology Measurements ....................................................................................... 66

5.2.2.2.1. Tackiness Testing ............................................................................................... 66

5.2.2.2.2. Oscillation Frequency Sweep ............................................................................. 68

5.2.2.2.4. Zeta Potential ..................................................................................................... 70

5.2.2.3. In Vitro Assay ......................................................................................................... 70

5.2.2.3.1. Cell Viability of HA 0.15% and HA 0.30% .......................................................... 70

5.2.2.3.2. 2D model - Evaluation of Cell Morphology and Cell Viability After Dehydration 71

Page 10: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

ix

5.2.2.3.3. 3D model - Dry Eye Model and Cell Viability...................................................... 72

5.3. Discussion ............................................................................................................................. 73

5.4. Conclusion ............................................................................................................................. 77

Chapter Six – Conclusion and Future Work .......................................................................................... 79

6. Concluding Remarks and Future Work ......................................................................................... 79

6.1. Concluding Remarks ............................................................................................................. 79

6.2. Future Work ........................................................................................................................... 80

References ............................................................................................................................................ 81

Appendix ................................................................................................................................................ 89

Page 11: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

x

List of Figures

Figure 1- Five main layers of the cornea's structure [4]. ................................................................... 3

Figure 2 - The eye anatomy diagrammatic illustration [5]. ................................................................ 4

Figure 3- Tear film multilayer composition [6]. ................................................................................... 5

Figure 4- Major etiological causes of dry eye. Adapted from [19]. ................................................... 7

Figure 5- Schematic representation of a polymer moving along the eye surface film during a

blink [47]. .............................................................................................................................................. 12

Figure 6- Contact angle between a droplet and solid surface [52]. ................................................ 15

Figure 7- Schematic representation of the in vitro cell culture model. Adapted from [65]. ......... 18

Figure 8 - Quality Risk Management Process overview (adapted from ICH Q9 and ISO 31000)

[68,71]. ................................................................................................................................................... 20

Figure 9 - Chemical repeat unit of HA [96]. ....................................................................................... 33

Figure 10 - Sodium hyaluronate chemical structure [100]. ............................................................. 34

Figure 11- Flow chart of the pilot batch manufacturing process of HA 0.15% and HA 0.30%. ... 37

Figure 12- Pareto chart of every failure mode of the manufacturing process of HA 0.15% and

0.30%. .................................................................................................................................................... 46

Figure 13- Results from the filling control test for HA 0.15% and HA 0.30%. ............................... 59

Figure 14 – Typical flow curve of shear stress as function of shear rate for HA 0.15% and HA

0.30% eye drops solution. .................................................................................................................. 65

Figure 15- Viscosity determination for HA 0.15% and HA 0.30% in absence and presence of

Mucin 5% (w/w) (mean SD, n=3). ..................................................................................................... 66

Figure 16- Three samples of pig eye used in the frequency sweep assay (A) and one of the

samples attached to the probe (B). ................................................................................................... 67

Figure 17- Frequency sweep with shear moduli as function of frequency of HA 0.15%, Mucin

and Mucin + HA 0.15% at room temperature. ................................................................................... 68

Figure 18- Frequency sweep with shear moduli as function of frequency of HA 0.30%, Mucin

and Mucin + HA 0.30% at room temperature. ................................................................................... 69

Figure 19- Time Sweep Test for HA 0.15% and HA 0.30% with and without mucin...................... 69

Figure 20- Determination of zeta potential for HA 0.15%, HA 0.30%, Mucin and both products

with mucin (Mean ± SD, n=3). ............................................................................................................. 70

Figure 21- Results of cell viability on ARPE-19 cell lines testing CR 0.30%, CR 0.15%, HA 0.30%

and HA 0.15% ate various concentrations (mean ± SD, n=8). ........................................................ 71

Figure 22-Cell viability ARPE-19 cell line after dehydration treatment exposed for 24h to HA

0.15% and HA 0.30% eye drops solution and commercial formulation CR 0.30% (mean ± SD,

n=9). ...................................................................................................................................................... 73

Page 12: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

xi

List of Tables

Table 1- Overview of medical devices directives, categories, classes/list and examples [33][35].

............................................................................................................................................................... 10

Table 2- In vitro methods to evaluate mucoadhesion in ophthalmic solutions. ........................... 14

Table 3- Properties of selected cell culture models of ocular tissues and respective

applications. Adapted from [65]. ........................................................................................................ 18

Table 4- General case of stability testing types and respective conditions. ................................ 24

Table 5- Criteria for evaluation of antimicrobial activity in terms of log10 reduction in the

number of viable micro-organisms for eye preparations [78]......................................................... 26

Table 6- Proposed specification for the final formulations (HA 0.15% and HA 0.30%). ............... 27

Table 7- Polymer and excipients selected for the developed formulations. ................................. 29

Table 8- Main equipment used during the manufacturing process of HA 0.15% and HA 0.30%

eye drop solution. ................................................................................................................................ 41

Table 9- Qualitative Severity and Probability Levels. ...................................................................... 42

Table 10- Severity, Occurrence and Detection table with respective risk classification. ............ 44

Table 11- Weighting Factor and respective specification according to the years of work

experience and academic level. ......................................................................................................... 44

Table 12- RPN matrix. .......................................................................................................................... 47

Table 13- ISO 14971:2007 risk matrix [70]. ........................................................................................ 47

Table 14- Unacceptable risks obtained through FMEA and current and corrective action. ........ 48

Table 15- Unacceptable risks obtained through ISO 14971 qualitative analyses and current and

corrective action. ................................................................................................................................. 49

Table 16- RPN calculation after the implementation of the corrective actions. ............................ 50

Table 17 - Steps, Process Controls and Acceptance Criteria considered for the validation plan

of Sodium Hyaluronate 0.15% and 0.30% eye drops solution, 8 mL. ............................................ 55

Table 18- Results obtained in the IPC and FPC analysis with respective specifications of HA

0.15% and HA 0.30%. ........................................................................................................................... 58

Table 19- Results obtained in the Bubble Point and Bonfiglioli test for HA 0.15% and HA 0.30%.

............................................................................................................................................................... 59

Table 20- Normal force and area under force time curve results for HA 0.15% and HA 0.30% and

their interactions with Mucin and with Pig Eye. ............................................................................... 67

Table 21- Optical microscope images of ARPE-19 after dehydration in no protective conditions

(Dry Eye), after dehydration preceded by treatment with HA formulations and cells not

submitted to dehydration (Medium). ................................................................................................. 72

Page 13: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

xii

Abbreviations Active Implantable Medical Devices (AIMD)

Active Medical Devices (AMD)

American Type Culture Collection (ATCC)

Aqueous Tear Deficiency (ATD)

Cetyltrimethylammonium broide (CTAB)

CFU (Colony-Forming Unit)

Colony Forming Units (CFU)

Commercial Reference (CR)

Detectability (DET)

Dry Eye Disease (DED)

Dulbecco's Modified Eagle Medium (DMEM)

Ethylenediamine tetraacetic acid (EDTA)

European Pharmacopeia (EP)

Evaporative dry eye (EDE)

Failure Mode Effect Analysis (FMEA)

FDA (Food and Drugs Agency)

Fourier-transform infrared spectroscopy (FTIR)

Heating Ventilation and Air Conditioning

(HVAC)

High Efficiency Particulate Arrestance (HEPA)

Hyaluronic acid (HA)

Hydroxyethyl cellulose (HEC)

In Process Control/Finished Product Control

(IPC/FPC)

In vitro Diagnostics (IVD)

Infrared (IR)

International Organization for Standardization

(ISO)

Kg (Kilogram)

LVER (Linear-Viscoelastic Region)

Medical Devices (MD)

mOsm (milliOsmol)

MW (Molecular Weight)

Negative Control (NC)

Occurrence (OCC)

Out-of-Specification (OOS)

Pa (Pascal)

PB (Pilot Batch)

Polyhexamethylene biguanide hydrochloride

(PHMB)

Polyvinyl pyrrolidone (PVP)

Polyvinylidene Difluoride (PVDF)

Positive Control (PC)

Psi (pound force per square inch)

Quality Control (QC)

R&D (Research and Development)

Risk Priority Number (RPN)

rpm (Rotation per Minute)

Sabouraud-dextrose agar (SDA)

Severity (SEV)

SFM (Serum-Free Media)

Sodium carboxymethyl cellulose (CMC)

Sodium dodecyl sulfate (SDS)

Sodium hydroxymethyl glycinate (SHMG)

Standard Derivation (SD)

The International Conference on

Harmonisation (ICH)

Tryptic soy broth (TSB)

Trypticase soy agar (TSA)

Ultraviolet–visible (UV/Vis)

V (Volt)

World Health Organization (WHO)

Page 14: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

1

Aims and Organization of the Dissertation

Dry eye disease (DED) is one of the most common eye diseases in current times being the most frequent

diagnosed disease in ophthalmology. In order to attenuate some of the symptoms cause by this

pathology tear lubricants containing polymers in their formulation are the most common form of

treatment. For that matter the pharmaceutical company Laboratório Edol, S.A. created in 2015 a project

consisting in the development of a safe and effective eye drop comfort solution leading to this

dissertation.

There are several formulations in the marked of tear lubricant with different types of polymers, each with

their own characteristics in terms if viscosity, duration time, mechanism and mucoadhesion properties.

One polymer which has been used with success in treating patients with severe DED is Hyaluronic Acid

(HA), a natural polymer with similar properties to the mucin in terms of viscoelastic and biophysical

properties. HA eye drops available in the marked have a concentration ranging 0.10% and 0.30% of

HA. Thus, it was decided to develop two products with different concentration: one with HA 0.15% (w/v)

for less severe cases such as slight discomfort and, other with HA 0.30% (w/v) indicated for more severe

cases.

The purpose of this dissertation was to develop two medical devices (MD) suitable and efficient to treat

DED, the identification of potential risks that may be involved in the manufacturing process of the pilot

batches and if the manufacturing process was effective through a Process Validation Plan.

To determine the most suitable formulation for eye drops an intensive bibliographic search was

performed, corresponding to Chapter One – Literature Overview. Since the eye is a very sensitive

organ a formulation that mimics the eye environment was the strategy to prevent ocular damage. This

means the pH, osmolality, electrolyte composition, buffer and preservative chosen must respect certain

specifications. Once the final formulation was developed and the manufacturing process was defined,

the next step was the scale-up of final formulations. This study was performed by Edol’s collaborators

and corresponds to Chapter Two – Previous Work.

In order to evaluate possible risks that may compromise the quality of both products a Risk Analysis

was performed to the pilot batch manufacturing process, corresponding to Chapter Three – Risk

Analysis. Two methods were used, one is a general tool (Failure Mode Effect Analysis - FMEA) and

the other is specific to MD (Qualitative Analysis according to the ISO 14971:2007). The aim of this

chapter was to understand the possible risks associated with the manufacturing process and to compare

each risk analysis method in order to understand and decide which is more sensitive and more efficient

in correcting the detected risks.

HA 0.15% (w/v) and 0.30% (w/v) eye drops solution are new products, thus the pilot batch manufacturing

process must be submitted to a Process Validation to demonstrate to prove the process operates

effectively and to confirm that the resulting product obeys the product requirements. Process controls

were applied to both products to verify if both passed the required specifications and to perceive the

Page 15: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

2

principal differences between them. This corresponds to Chapter Four - Pilot Batches Process

Validation.

The final chapter, Chapter Five – Product Characterization, had the purpose to evaluate the structural

and mucoadhesion properties of HA 0.15% (w/v) and 0.30% (w/v), eye drops solution and the efficacy

of both products by an in vitro Dry Eye model using ARPE-19 cell line. The structural and mucoadhesion

behaviour was evaluated by different viscosity and rheological measurement using mucine and pig eye.

A cell viability test as well a study of cell morphology was also performed to study cell behaviour when

the products were applied.

Page 16: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

3

Chapter One – Literature Overview

1. Theoretical Introduction

1.1. Anatomy of the Eye

The human eye is the essential sense organ that allow us to see whose anatomy is quite complex. The

light is refracted by the eye itself and produces a focused image stimulating the nervous system, giving

the ability to see [1].

The eye consists in a fibrovascular, approximately, globular structure with 24 mm of diameter and with

a mass of about 7.5 g, corresponding less than 0.05% of the total body weight, with compact ocular

tissue whose thickness corresponds to several cell layers [2].

The structure of the eye is divided in the anterior segment and the posterior segment. The anterior

segment comprises the smaller anterior chamber, between the cornea and iris, is primarily responsible

for collecting and focusing light, and the larger posterior chamber, between the iris and the lens, is

responsible for detecting light. The anterior segment of the eye includes the lens, lachrymal system, iris,

aqueous humour, ciliary body, pupil, conjunctiva and the cornea. The posterior segment itself includes

the retina, choroid, sclera, macula, fovea, optic nerve and the vitreous humour [2].

Three concentric adjoining tissue layers comprise the eye. This organ is encircled with a collagenous

layer corresponding to the outermost layer that provides mechanical strength. The epithelial membrane

located in the interior portion is called the cornea, a clear transparent tissue with the purpose of focusing

the light to the retina. The cornea’s structure has both lipophilic and hydrophilic properties and five

distinct layers: epithelium, Bowman’s membrane, stroma, Descemet’s membrane and endothelium

(Figure 1). The epithelium is a lipophilic layer offering approximately 90% resistance to hydrophilic drugs

and 10% to hydrophilic preparations. Underneath the epithelium is the Bowman’s membrane, a

transitional acellular structure composed of protein fibres called collagen, approximately 8 to 14 µm in

thickness. The next layer is the hydrophilic stroma, a gelatinous structure composed by 80% of water,

comprising collagen, mucopolysaccharides and proteins. This layer corresponds to 90% of the cornea’s

total thickness. The Descemet’s membrane is located afterwards, with 6 µm of thickness, supporting the

endothelium and it is responsible to regulate stromal hydration [3].

Figure 1- Five main layers of the cornea's structure [4].

Page 17: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

4

The posterior portion is represented by an opaque collagenous layer named sclera, which maintains the

shape of the eye and gives the attachment to the extrinsic muscle of the eye. The uvea is located in the

middle layer, it is a pigment layer that comprises the iris and the ciliary body in the anterior portion and

the vascular choroid in the posterior portion. The iris is the coloured part of the eye and controls the

amount of light entering the eye, serving as a biological aperture due to the presence of the pupil. The

ciliary body or muscle is a ring-shaped muscle responsible for the shape of the lens by contracting and

relaxing. It secretes aqueous humour providing nutrients to the avascular tissues in the anterior segment

and it also maintains the intraocular pressure. The choroid is a vast network of capillaries that supplies

the retina with nutrients and absorbs unused radiation. The retina corresponds to the innermost layer,

which detects and transduces light signal to the brain by the passage of light through the cornea,

aqueous humour, pupil, lens, the hyaloid and the vitreous humour, before reaching the retina. The retina

contains photosensitive elements called rods and cones, which convert the light detected into nerve

impulses and are sent onto the brain along the optic nerve. The lens is in between the anterior and

posterior segments, responsible for further refracting the light entering the eye. It is a flexible unit

consisted of layers of tissue enclosed in a tough capsule. The optic nerve contains approximately one

million fibres transmitting information from the rod and cone cells of the retina. The location where the

optic nerve leaves the retina is positioned the papilla. The anterior segment of the eye is filled with a

jelly-like substance called aqueous humour and the posterior segment is filled the gelatinous vitreous

humour. This segments are divided by a diagram called hyaloid [1,2]. All tissues and fluids are illustrated

in the Figure 2.

Figure 2 - The eye anatomy diagrammatic illustration [5].

1.2. Structure of the tear film

The tear film is a three-layered structure with the purpose to provide protection and lubrication to the

eye, reduces the risk of eye infection and keeps the surface of the eye smooth and clear. It also protects

the front of the eye from the environment and allows the eyelids to slide comfortably over the front

surface. This liquid comprises an anterior lipid layer, an aqueous layer and a deep mucin layer,

represented in Figure 3 [6][7].

Page 18: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

5

Figure 3- Tear film multilayer composition [6].

When the eye is closed, the tear fluid is contained within a single compartment, the conjunctival sac,

and when the eye opens this fluid is re-distributed between three compartments: the sac, the pre-ocular

film and the menisci [7].

The tear film is only stable for a short period of time, thus after 20 to 40 seconds the human eye has an

urge to blink, due to an unpleasant sensation. In the short time between two blinks occurs the rupture

of the tear film due to dispersions forces and concentration gradients on the mucous layer, forming dry

spots on the cornea, creating irritation to the corneal nerve endings, which induces blinking. After

blinking and during the eyelid opening, a new tear film is created and spreads over the eye surface.

Dispersion forces, interfacial tension and viscous resistance of the mucous layer are the main factors

that define the time of rupture of the mucus and the breakup time of the tear film [8].

1.2.1. The Lipid Layer

The lipid layer, or oily layer, is an essential component to provide a smooth optical surface for the cornea,

enchasing the stability and the spreading of the tear film. This layer prevent the contamination of the

tear film by sebum, sealing the apposed lid margins during sleep, and most importantly retard water

evaporation from the surface of the open eye. The lipid layer remains stable over a series of blinks and

it is composed by meibomian lipids which are formed in the meibomian glands, a tubule-acinar holocrine

gland that discharged the entire content in the secretion process. These lipids are composed by non-

polar lipids such as cholesterol and wax esters and polar lipids such as (O-acyl-)-ω-hydroxy fatty acids

and phospholipids, which interact with the aqueous layer [7,9,10].

1.2.2. The Aqueous Layer

The aqueous layer is responsible for the formation of the tear bulk. This layer provides oxygen and

nutrients to the underlying avascular corneal tissue, flushing away epithelial debris, toxins and foreign

bodies. It is secreted from the lacrimal glands along with specific variety of proteins, electrolytes and

water. The second source of electrolytes and water in the tears is the conjunctival epithelium. This layer

also possesses anti-bodies that are critical for corneal wound repair and protects against infections

[6,10].

Page 19: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

6

1.2.3. The Mucous Layer

The layer of the tear film in contact with the conjunctiva is mucus, which is also responsible for the

formation of the tear bulk, for enabling the tears to wet the eye by adhering to its front surface and for

the removal of unwanted fatty compounds from the corneal surface. Mucus resides at the surface of the

cornea and it is composed predominantly by sugar-rich glycosylated proteins, called mucin. The mucin

is produced by corneal and conjunctival epithelial cells, specialised goblet cells, and subsurface vesicles

found below the conjunctival cells. It is also composed by proteins, lipids, electrolytes, enzymes,

mucopolysacchrides and water. These transmembrane mucins, when anchored into the epithelial tissue

help stabilise the tear film. The gel-like structure of these mucins provide an easily wettable surface by

lower the surface tension of the tears, which helps the rapid spreading of fluid after blinks [7,8,10–12].

This spontaneous and rapid spreading is also possible due to the interaction of conjunctival mucin with

water and lipids, which contributes to the stabilization of the tear film [13].

There are up to 20 mucin genes identified in humans which produce MUC1, MUC2, MUC3A, MUC3B,

MUC4, MUC5AC, MUC5B, MUC6, MUC7, MUC8, MUC12, MUC13, MUC15, MUC16, MUC17, MUC19,

MUC 20, and MUC21. These proteins are situated in the wet-surfaced epithelia of the body such as the

ocular surface, entire airway and gastrointestinal tract. In the ocular surface occurs the expression of

MUC1, MUC4, MUC15, MUC16 and MUC20, being the last one the most expressed mucin in the human

conjunctiva. In the lacrimal gland MUC1, MUC4 and MUC16, also present in the cornea and the

conjunctiva, are present in the acinar cell membranes but only MUC4 and MUC16 are likewise present

in soluble form. All three mucins are located in the tears in soluble form [14].

1.3. Dry Eye Disease

DED, also known as keratoconjunctivitis sicca, is a pathology whose origin comes from several factors,

resulting in symptoms of discomfort, visual disturbance, tear film instability with potential damage to the

ocular surface, increased osmolality of the tear and inflammation. This disease can be divided into two

different types, aqueous tear deficiency (ATD) and excessive tear evaporation or evaporative dry eye

(EDE). DED is a commonly reported clinical problem and the most frequently diagnosed disease in

ophthalmology [11,15,16].

DED ranges from 5% to more than 30% of the population, estimated that about 3.23 million women and

1.68 million men with 50 years and older have dry eye, meaning that this disease affects mostly older

people and women, especially those who suffer from arthritis and allergies. It is also highly prevalent in

contact lens wearers [11,15,17].

Once again, DED can be divided into two main groups, the ATD and the EDE. The ATD can be

subdivided into Non-Sjögren’s syndrome and Sjögren’s syndrome, which is a generalised inflammatory

autoimmune disease associated with lacrimal and salivary gland lymphocytic infiltration. The EDE can

be divided into meibomian gland disease and exposure-related dry eye. Therefore there are four main

groups of diseases that can produce severe DED: the Sjögren’s syndrome, chronic progressive

conjunctival cicatrisation syndromes, specific ocular diseases and non-ocular diseases [9,17] (Figure

4).

Page 20: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

7

Patients with Sjögren’s syndrome usually have dry eyes and dry mouth, but they also have or may not

have an associated rheumatological disease dependent on whether it is primary Sjögren’s syndrome or

secondary Sjögren’s syndrome, respectively [17].

The chronic progressive conjunctival cicatrisation syndromes includes several diseases such as the

Stevens-Johnson syndrome, trachoma, ocular pemphigoid, drug induced pseudopemphigoid, graft

versus host disease, chemical burns and conjunctival cicatrisation that occurs after severe membranous

conjunctivitis [17,18].

The third group is dedicated to those who have signs of DED because of specific ocular diseases, such

as dacryoadenitis, congenital absence of the lacrimal gland, Riley-Day syndrome, cholinergic blockade

due to drugs, chronic blepharoconjunctivitis, sinile atrophy of the lacrimal gland and after refractive eye

surgery [17].

The final group is devoted to those diseases that present DED symptoms but in reality have adequate

tear production, for instance trigeminal nerve paralyses with loss of corneal sensation, facial sensory

nerve paralysis, exposure keratitis and vitamin A deficiency, resulting in xerophthalmia, condition in

which the eye fails to produce tears [17].

Apart from the diseases mentioned above, other factors can cause an alteration to the evaporation rate,

including ambient conditions, hormonal regulation, blink rate, area of palpebral aperture, action of toxic

topical agents such as preservatives and complication in the tear film compartments. Contact lenses

can disrupt the stability of the tear film, since they increase the evaporation rate, causing a rupture in

the tear film about twice as fast as on the surface of the cornea. This lack of stability cause by DED is

due to the rise of the surface tension and osmolality values [9,10,19].

Figure 4- Major etiological causes of dry eye. Adapted from [19].

Page 21: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

8

1.3.1. Treatment of Dry Eye Disease

1.3.1.1. Pharmacological Treatment

The pharmacological treatment of DED is focused on treating inflammation and tear restoration, since

DED is a symptom of various illnesses, resulting in inflammation of the cornea and conjunctiva. A widely

used approach to treat inflammation problems is the use of anti-inflammatory drugs such as topical

corticosteroid eye drops. Steroids are another treatment used in DED, however, it presents many side

effects as microbiological contamination, elevated intraocular pressure and cataract formation, therefore

it is only recommended treatments no longer than one or two weeks. Therefore, the use of non-steroidal

anti-inflammatory drugs increased due to their non-severe side effects in the treatment of DED, eye

discomfort decreases due to its analgesic effect and inflammation reduction. Cyclosporine eye drops,

the first one of the new generation immunomodulatory drugs, has an anti-inflammatory effect in the

lacrimal gland, resulting in an increase in tear production and conjunctival goblet density. Other

treatments consists in the application of antibiotics including oral doxycycline, azithromycin and

tetracycline to treat meibomian gland dysfunction [20–22].

1.3.1.2. Food supplements

According to the European Commission, food supplements are concentrated sources of nutrients (or

other substances) with a nutritional or physiological effect. Such supplements can be marketed as

tablets, capsules or even liquids in measured doses [23]. To alleviate symptoms associated with DED,

supplements available in the market are composed by fatty acids, vitamins and antioxidants such as:

Omega 3 and 6: there has been a great amount of interest generated in the area of using

essential fatty acids. Oral supplements containing omega-3 essential fatty acids, such as alpha

linoleic, docosahexaenoic acid and eicosapentaenoic acid, and omega-6 essential acids, such

as linoleic acid and gamma-linoleic acid, have shown to attenuate symptoms of dry eye by

treating chronic eye inflammation and Sjögren syndrome, due to their anti-inflammatory effect

[24];

Vitamin A: eye drops containing vitamin A have shown to be as efficient as prescription eye

drops. However, it is only indicated for patients with vitamin A deficiency since excessive

vitamin intake may trigger stomach and nerve side effects as well blurred vision [25];

Vitamin D: Patients with vitamin D deficiency present symptoms of dry eye and impaired tear

function. The administration of this vitamin is associate with the enhancement of tear film

stability and the reduction of ocular surface inflammation [26];

Lutein and Zeaxanthin: these antioxidants carotenoid pigments are presented in a high

concentration in the retina, more precisely the macula, working as a filter protecting the macula

from blue light and protect cells against oxidative stress by structurally bound antioxidants.

Studies have shown that lutein and zeaxanthin might reduce the risk of various eye diseases,

especially age-related macular degeneration, influencing cell viability and function, which may

reduce dry eye sensation [27].

Page 22: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

9

1.3.1.3. Medical Devices

There are several approaches to treat DED, until now most treatments do not treat the cause of the

disease, but there are symptomatic treatments. This includes medical devices such as tear supplements

called artificial tears, which are artificial lubricants with hypotonic or isotonic buffers containing

electrolytes, surfactants and many types of viscosity agents. Another treatment option is tear retention

devices, which plug the lacrimal puncta preventing tear drainage. Other option is the use of implants to

permanently obstruct the lacrimal puncta, also known as punctal plug. Moisture chamber spectacles

have been supported to alleviate ocular symptoms associated with dry eye. The mechanism is by

increasing the periocular humidity and it can cause a growth of the lipid layer tear film thickness.

Because of this humidity, spectacle wears have a long inter-link interval [20,21,28].

According to the WHO, the definition of MD is an instrument, implement, machine, contrivance, implant,

in vitro reagent, or other similar or related article, including a component part, or accessory which is

intended for use in the diagnosis of diseases or other condition, or in the cure, mitigation, monitoring,

treatment, or prevention of disease, in man or animal. This products are intended to affect the structure

or any function of the body of man or other animals, and which does not achieve any of its primary

intended purposes through chemical action within or on the body of man or other animals and which is

not dependent upon being metabolized for the achievement of any of its primary intended purposes [29].

In other words, all kinds of diverse products from a simple bandage to a complex pacemaker, with no

pharmacological, metabolic or immunological activity, with the purpose of improving quality of life. Their

goal is to achieve the intended effect by other means beyond the ones given by drug consumption [30].

The European Commission defines it in a similar form, adding that MDs help improve the quality of life

of those with disabilities [31].

MDs are intended to be used by patients and consumers, which mean all products, must be submitted

to a safety assessment and performance evaluation before going to the market. Health professionals

also take advantage of the devices by giving the best information and advices on who to make the best

usage of the product [30].

There is a total of five types of MD according to the INFARMED [32]:

Active Medical Devices (AMD), dependent on an energy source non-generated by the

human body;

Active Implantable Medical Devices (AIMD), totally or partially introduced, surgically or

medically, into the human body and which is intended to remain after the procedure;

In vitro Diagnostics (IVD), devices used for examining sample material from the human body,

including excreted material, for providing information to ensure a correct patient diagnosis;

MD made by measure, manufactured specifically according to a written prescription for a

specific patient;

MD Systems and Sets for Intervention, packaged and dispensed together, which are placed

on the market with only one commercial name.

In Europe, registration of MD is subject to harmonised European directives, consisting of three

directives: Directive 90/385/EEC regarding active implantable medical devices (AIMDD 90/385/EC),

Directive 93/42/EEC regarding medical devices (MDD 93/42/EC) and the Directive 98/79/EC regarding

Page 23: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

10

in vitro diagnostic medical devices (IVDD 98/79/EC) [33,34]. Classes and examples of each Directive

are presented in Table 1.

Table 1- Overview of medical devices directives, categories, classes/list and examples [33][35].

EU Directive Device

category Class/List Examples

MDD 93/42/EC MD

Class I (Low Risk) Bandage, wrists, compression socks,

wheelchairs, crutches

Class I Sterile (Low Risk) Examination gloves, sterile dressings

Class I Measurement (Low Risk)

Thermometer, syringes graduates without needle, blood pressure meter

Class IIa (Medium Risk) Syringes with needles, needles, lancets, surgical

gloves, resonance equipment

Class IIb (Low Risk) Blood bags, incubators, ophthalmic comfort

solutions, dressing material

Class III (High Risk) Cardiac valves, stents, hip prostheses, IUDs,

breast implants

IVDD 98/79/EC

High-risk IVD

List A - High Risk Material for detention of HIV 1 and 2, HTLV I and

II, Hepatitis B, C and D

List B - Moderate Risk

Material for detention of Rubella and toxoplasmosis, Phenylketonuria,

Cytomegalovirus infection, Chlamydia, blood groups,

Self-tests Pregnancy tests, ovulation tests, blood glucose

measurement equipment

Low-risk IVD

Other Urine or faeces collection flasks, aseptic urine

collection flask

AIMDD 90/385/EC

AIMD N/A Pacemakers, defibrillators, cochlear implants

1.3.1.3.1. Ophthalmic Comfort Solutions

Tear lubricants are the most common form of the treatment for DED and they are class IIb MD. There

are many lubricants formulation on the market that differ by their mechanism of action. It is not

completely understood the beneficial mechanism of tear lubricants, but it is related to volume

replenishment, tear film stabilization, preservation of the smooth refracting surface, reduction of tear

osmolality and reduction of the friction between the eyelids and the cornea. The mechanism depends

on the formulation used, for example, the hydrogel hydroxypropyl guar exists as monomers in a borate-

containing solution. Once installed in the patient’s eye, the contact between the patient’s tear and the

hydrogel causes the hydrogel to cross-link with the borate creating a more viscous and elastic matrix.

Another strategy is to replace or increase tear components to maintain tear stability, by producing for

example lipids [36,37].

Overall, eye drops differ in terms of composition, viscosity, duration of action, presence and type of

preservatives, osmolality and pH, being the main ingredient polymers. Polymers used in artificial tear

include hydroxypropyl methylcellulose (HPMC), carboxy methylcellulose, polyvinyl alcohol, carbopol,

polyvinylpyrrolidone, polyethylene glycol, dextran, HA and/or carbomers. These molecules may

increase the viscosity of eye drops, enhancing mucoadhesion. Increased viscosity increases the

retention time, although it also causes unwanted visual disturbance. In some cases higher viscosity eye

Page 24: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

11

drops causes precipitation of crystals on the eyelids and lashes. For that reason it is recommended

daytime use of eye drop with low-viscosity and for highly viscous preparations use before sleep [36].

The preservative use in ophthalmic preparation can prevent contamination. Preservatives used in these

formulations include benzalkonium chloride, chlorobutanol, sodium perborate, sodium chlorite,

polyquaternium-1, cetrimonium chloride, EDTA, sodium hydroxymethyl glycinate and

polyhexamethylene biguanide hydrochloride. However, benzalkonium chloride and chlorobutanol

preservatives can cause symptoms similar to DED as they can damage the corneal epithelium, the other

preservatives mentioned may be less harmful to the ocular surface [36,38,39].

One of the characteristics of DED is increased osmolality due to corneal and conjunctival epithelial

damage. Thus, lowering tear osmolality using hypo-osmolar solutions may be a suitable strategy to treat

DED [36,40].

The variability of tear pH may be related to with carbon dioxide saturation or the meibomian lipids in the

tear film, thus the tear pH can range from 6.9 to 7.5 for normal population and patients with DED. The

most common buffer systems used in ophthalmic eye drops are citrate, phosphate, Tris-HCl and borate

buffer. Phosphate has been the buffer of choice for a long time, since it naturally occurs in the eye,

however calcific degeneration of the superficial cornea by calcium hydroxyapatite deposition has been

associated with the use of ophthalmic products containing phosphate. The borate buffer was for that

reason introduced as an ophthalmic buffer, since it was better suited than phosphate and, because of

its antimicrobial activity. There are no cases of complications after the use of citrate and Tris in literature,

though citrate buffer pH ranges between 3.0 and 6.2, which is lower than the tear film pH. Tris buffer

showed effectiveness in treating ocular acid burns, when the time of exposure to acid was short [36,41].

1.4. Ophthalmic Comfort Solution Characterisation

1.4.1. Mucoadhesion and mucoadhesive properties

Mucoadhesion is the adhesion of a material to a mucous membrane or a mucus-covered surface which

is the case of the eye surface. The problem concerning ophthalmic drugs is that due to the efficient

protective mechanisms of the eye, the bioavailability is often low. The act of blinking and lachrymation

remove rapidly foreigner substances, as well drugs from the eye surface. Formulation with

mucoadhesive properties are for that reason a topic of interest in order to enhance drug bioavailability,

prolonging the contact time between the formulation and the corneal epithelium. Due to the

mucoadhesion properties, the frequency to apply the product is decreased, reducing the toxic side

effects and the drug concentration [8,42].

The strategy to increase the residence time of the drug at the ocular surface is by changing the product

characteristics, for instance its viscosity. Although, by increasing the product viscosity the patient vison

may become blurred at the moment of administration, which may compromise the treatment. In organs

with high blood flow and considerable surface area, such as the eye, mucoadhesion drug delivery

systems give rapid absorption and high bioavailability [43].

Mucoadhesive formulations use water soluble polymers as the adhesive component. The mechanism

of adhesion starts with the wetting and swelling of the polymer, allowing an intimate contact with the

tissue. Then the polymer entanglements on the mucins chains forming weak chemical bounds and

Page 25: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

12

allowing the adhesion on the mucosal surface. The bounds between the polymer and the mucous layer

could be physical or mechanical, secondary chemical bounds and covalent bounds. The required

characteristics to a polymer in order to obtain adhesion to the mucous layer are at least one of the

following: sufficient quantities of hydrogen-bounding chemical groups, anionic surface charges, high

molecular weight, high chain flexibility and induction of spreading by surface tensions [44,45].

1.4.1.1. Polymers

Polymers are high candidates to improve the bioavailability of ophthalmic formulations, due to their

mucoadhesive properties. High molecular weight polymers with different function groups, such as

carboxyl, hydroxyl, amino and sulphate, are capable or forming hydrogen bonds with mucous layers,

without crossing biological membranes. The interpenetration of the polymer chains in the mucous layer

cause a strengthening on itself by forming entanglements and secondary bound with the mucin

molecules or due the dehydration of the mucus caused by water movement. This strengthening is

necessary for a strong mucoadhesion. Water-soluble mucoadhesives polymers slowly dissolve in the

tear film, whereas water-insoluble would be retained until the mucin is replaced or until the shear force

of blinking dislodges the mucoadhesive system [8,43,46].

Other vantage for the use of polymers solutions is that they can increase the thickness of the normal

tear film, which is favourable on its stabilization. There are two types of mechanisms responsible for

spreading tears over the ocular surface (Figure 5). The first is by mechanical action, the upper eyelid

pulls water as it is raised, thus the amount of water moved is enhanced by the vertical spreading of

polymer molecules, dragging additional water with them. The second mechanism moves the polymer

molecules from the bulk solution to the surface, providing a surface-pressure gradient which induces

spreading [47]. Nowadays, HA is one of the most used polymer in eye drops formulations due to its

mucoadhesive properties. Examples of other mucoadhesive polymers are cellulose derivatives such as

methylcellulose, acrylates, chitosan, lectins and pectins [8,48].

Figure 5- Schematic representation of a polymer moving along the eye surface film during a blink

[47].

Page 26: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

13

1.4.1.1.1. Hyaluronic Acid

The salt form of HA is sodium hyaluronate and its molecules have physical properties similar to the tear

glycoproteins and can easily cover the corneal epithelium. HA macromolecules are natural and are

presented in the vitreous body of the eye. First, they were used as viscosity agents, but it was found

that these polymers adsorb at the mucin/aqueous interface and extend into the adjacent aqueous layer,

forming a stabilizing thick layer. This polymer exhibits, just like the tear film, a non-Newtonian behaviour,

giving an advantage of high viscosity at rest between blinks, which decreases the dryness sensation of

DED [43,49].

Tear substitutes eye drops solution containing HA have been used with success in treating patients with

severe DED. The similar properties to mucins, such as viscoelasticity and biophysical properties, have

beneficial effects, providing a long-lasting hydration and retention time and obtaining a good lubrication

of the ocular surface. HA presents wound healing properties and promotion of corneal epithelial cell

proliferation, since HA is an important constituent of the extracellular matrix [43]. A study performed by

Johnson et al. was carried out to access the efficacy of two developed eye drops containing 0.10% and

0.30% of HA in the treatment of DED, using thirteen subjects with moderate DED. The eye drops were

administrated and the subjects’ symptoms were measured at 5, 15, 30, 45, 60 min and then hourly until

6 hours after drop installation. The authors concluded that both products reduced symptoms of ocular

irritation and lengthens tear breakup time [50]. Other study performed by Hamano et al. determined the

most effective concentration of HA in prolonging tear film stability. It was concluded that concentration

should be at least 0.10% to delay the breakup of the precorneal tear film.

1.4.1.2. Factors Important to Mucoadhesion

Several factors may affect the polymers bioadhesive properties. Polymer-related factors may be the

molecular weight, since it is necessary to have a high molecular weight in order to obtain good

bioadhesion. The concentration of the molecule also has a massive impact, highly concentrated systems

decreases significantly the adhesive strength, because the coiled molecules become solvent-poor,

making the chains for interpenetration less available. Other reason can be the lack of flexibility of

polymer chains, if the polymer chain decreases the capacity to interpenetrate and entanglement, the

mucous layer also decreases, reducing the bioadhesive strength. Besides molecular weight or chain

length, the conformation of the polymer’s molecule is also important. Exposed adhesively groups are

dependent of the molecule’s conformation, which determents the capacity of bioadhesion in the mucous

layer [44,51].

Factors related to the environment are the pH levels The mucous layer will have a different charge

density depending on the pH due to difference in dissociation of functional groups on the carbohydrate

part of the polypeptide backbone. Thus, the pH of the product is important for the degree of hydration.

The contact time is also important since it determines the extent of swelling and interpenetration of the

polymer chains. Swelling is also other factor and it is dependent of the polymer concentration, ionic

concentration and presence of water. Overhydrated molecules may result in a slippery system, which

decreases adhesion to the mucous layer [44].

Page 27: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

14

1.4.1.3. Techniques Used for the Assessment of Mucoadhesion

The most common and convenient methods to assess the mucoadhesive properties of a potential

formulation is through in vitro tests. There are numerous studies concerning in vitro tests to measure

mucoadhesion, although there is not a standard procedure for such technique. Several approaches

were developed to rank mucoadhesion properties of polymers and formulations to understand their

adhesive behaviour [52].

Methods can be from simple analytical techniques to more sophisticated procedure. One procedure is

tensile strength measurements, also called texture analysis, were the mucoadhesive characterization is

based on the measurement of tensile force to break the interaction between the mucoadhesive platform

and the test substrate. The maximum detachment force and work of adhesion, the area under force time

curve, are used as parameters for comparison of mucoadhesive performance. This method provides

information about possible mucoadhesive strength, though it may be limited due to the dissolution of the

polymer in mucous gel and the absence of biological tissue [42,52,53].

Other methods include rheological techniques, particularly flow and oscillation methods have been

frequently used to study mucoadhesive properties of formulations. Interactions between mucin and

polymer may be measured by flow and oscillation trough viscosity and viscoelasticity properties,

respectively. Rheological methods are the most widely used techniques, providing information

concerning the deformation of the material within a broad range. However, the results obtained depend

on the concentration of the polymer, the mucin type and instrumental factors. Other disadvantage of this

method is that it cannot give information about the weakest region of the mucoadhesive junction since

only the interpenetration layer is simulated with this method [46,52].

Table 2 shows some examples given by authors to study the mucoadhesion capacity of polymers using

different methods.

Table 2- In vitro methods to evaluate mucoadhesion in ophthalmic solutions.

In vitro

methods Excipients (Polymer) Mucoadhesion results Conclusions Reference

Tensile Strength

Carbopol C971 and C974

4% (w/v) in water based

gels

Higher maximum force

in C974 (6.21 ± 2.75 N >

4.60 ± 1.71 N)

Higher elastic

component

and highly

crosslinked polymer

[54]

Tensile Strength

Polycarbophil 3% (w/w)

and poly(methylvinylether-

co-maleic anhydride 3%

(w/w) (Gantrez S97) in

vaginal gels

Gantrez S97 showed

higher mucoadhesive

strength (0.50 ± 0.04 N

< 0.37 ± 0.03 N)

Increased diffusion,

interpenetration and

entanglement with

mucin

[55]

Viscosity

Measurements

Sodium Hyaluronate

HA1100, HA800, HA500

and HA250 at several

concentrations (data not

shown)

Higher concentration

increased the viscosity

Strength of

formulation/mucin

interaction increases

with HA

concentration

[56]

Page 28: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

15

Table 2- In vitro methods to evaluate mucoadhesion in ophthalmic solutions (Continuation).

In vitro

methods Excipients (Polymer) Mucoadhesion results Conclusions Reference

Viscosity

Measurements

Chitosan and Alginate

(concentration data not

shown)

Increase of viscosity in

both cases when

interacted with mucin

Electrostatic

interactions,

hydrogen bounding,

hydrophobic

interactions and

entanglement with

mucin

[57]

Oscillatory

Measurements

Sodium Hyaluronate

HA500 0.67% (w/w)

G’ > G’’ at high

frequencies (10 Hz)

Physical

entanglements with

mucin

[56]

1.4.1.4. Mucoadhesion Theories of Polymer Attachment

A definition of the process of mucoadhesion does not exist, since this is a complex process. Numerous

theories have been proposed in order to explain this phenomenon: the wetting theory, the diffusion

theory, the mechanical theory, the electronic theory, the adsorption theory, the cohesive theory and the

facture theory. In Table 2 is present some examples of published studies who used some of the below

theories to discuss their results.

1.4.1.4.1. Wetting Theory

The wetting theory discusses the bounding between the formulation and the surface tissue through

intermolecular interaction and interfacial tension. It is usually applied for liquid or low mucoadhesive

systems, measuring the ability of the system to spread across the biological substrate. The spreading

indicates that there are interactions and can be measured by the liquid-solid contact angle. If the contact

angle of liquids on surface tissue is lower than 90°, a greater affinity for the liquid to the layer surface is

achieve and the liquid tends to spread out to a large area, due to adhesive forces. If the contact angle

is greater than 90° the wetting of the surface is unfavourable, because of the cohesive forces within the

liquid molecules, maintaining the shape of the droplet and minimizing its contact area to the solid surface

[44,45,52] (Figure 6).

Figure 6- Contact angle between a droplet and solid surface [52].

Page 29: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

16

The interfacial tension between the solid and the gas is represented by γSG, the interfacial tension

between solid and liquid by γLG and θ the contact angle between solid and liquid interface. The interfacial

tension may be calculated by Young’s equation:

𝛾𝑆𝐺 = 𝛾𝑆𝐿 + 𝛾𝐿𝐺 cos 𝜃 (1)

1.4.1.4.2. Diffusion Theory

The diffusion theory supports the concept that interpenetration and entanglement of bioadhesive

polymer chains and mucous chains produce semi-permanent adhesive bounds, the deeper the

penetration the stronger the bound. This penetration is dependent on the concentration gradient and

diffusion coefficient. The bound strength for a given polymer is achieved when the depth of penetration

is approximately equal to the end-to-end distance of the polymer chains. It is important that the polymer

and the mucous have a similar chemical structure, the more structurally similar a polymer is to its target,

the greater the mucoadhesive bond will be [44,45]. A study performed by Reinhart and Peppas reported

that the diffusion coefficient depended on the polymer’s molecular weight and that it decreased with the

increase of cross-linking density [58].

1.4.1.4.3. The Mechanical Interlocking Theory

This theory only considers the adhesion between the liquid and a rough surface or a surface rich in

pores forming an interlocked structure, which rise to adhesion. The adhesion between the

mucoadhesive system and the surface occurs within a diverse biological environment [44,52].

1.4.1.4.4. The Electronic Theory

The bioadhesive material and the target biological material have a different electronic structure, when

the contact occurs, it results in a transfer of electrons amongst the surface forming an electronic double

layer at the bioadhesive-biological material interface. This theory suggests that the electronic forces are

critical in generating bound adhesions [44,45,52].

1.4.1.4.5. The Adsorption Theory

The various surface interactions that results in adhesion is due to the presence of intermolecular forces,

including primary bounds and secondary bounds formation. These forces include hydrogen bonding and

Van der Waals interactions. Although these forces are individually weak, the number of interactions can

produce an intensive adhesive strength. This is the most accepted theory [44,45,52].

1.4.2. Evaluation of the Product Efficacy

1.4.2.1. In vitro models

Cell-based assays are nowadays a widely used method in drug development process providing a

simple, fast and cost-effective tool in order to avoid large-scale animal testing. The conventional 2D

culture system has helped the scientific community to study the cellular physiology and their behaviour.

This system consists in a monolayer of cells cultured on a flat and rigid substrates, in this conditions the

extracellular matrix components, cell-to-cell and cell-to-matrix interaction important to cellular

Page 30: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

17

differentiation and proliferation and cellular functions in vivo are lost. This happens because 2D cell

cultured does not consider the natural 3D environment of cells in vivo, meaning that the results may

provide misleading and non-predictive data for in vivo responses [59,60].

3D cultured cells provides a behaviour closer to the complex in vivo conditions, giving a major advantage

over the 2D approach that the gap between cell cultured systems and the cellular physiology is

decreased. The 3D approach replicates or mimics the extracellular matrix making it a good near-to-in

vivo system, other advantages of this approach for evaluation drug candidates include oxygen and

nutrient gradients, increased cell-to-cell and cell-to-extracellular matrix interactions, non-uniform

exposure of cells within the 3D structure to the test molecule and varying cell proliferation zones [59–

62].

The reason for the near-to-in vivo behaviour in 3D cultures is the use of matrices and scaffolds. The

type of matrix or scaffold used depends on the type of morphological and physiological behaviour of the

cells as well the nature of the study. Commonly used scaffolds are agarose, collagen, fibronectin,

gelatin, laminin and vitronectin, mimicking the native extracellular matrix by porosity, fibrous,

permeability and mechanical stability. This micro environment enhances the biophysical and

biochemical interactions of the adhered cells, providing more realistic and predictive data for in vivo

studies and cost effective screening platform for drug development and testing [60,63].

1.4.2.1.1. 3D In Vitro Models of the Eye and Ocular Diseases

Ocular diseases have been investigated trough animal and cell cultured models to understand the

molecular mechanism which causes those diseases and to study potential drug candidates. The human

eye presents unique and complex features whose animal models just cannot mimic. Cell culture are

therefore an advantage, because they are experimentally controlled systems, making the results more

reproducible than those obtained from animal models [64].

To determine a drug’s efficacy in ocular diseases, in vitro 3D cultured cell studies have been widely

used, providing useful data comparable to in vivo studies. In drug discovery research, a 2D cell model

may lead to a selection of a candidate drug that cannot reach its target in vivo, making the 3D model

more efficient since it creates a microenvironment more appropriate for demonstrating long-term effects

of the drug. It only exists to date reginal parts of ocular in vitro models, like corneal, conjunctival and

retinal models, but not an in vitro ocular equivalent as an organ. Meaning that the 3D model must be

built in order to simulate the conditions and the specific region of the disease or the environment in study

(Figure 7) [64,65].

Various types of cells can be used in this model, cell-based lines systems form animals or human

resources, the second type with the advantage of providing reliable results by avoiding species-related

problems. These cells can be specified primary or immortalized cells depending of the location of

study and the application (

Table 3).

In order to test the efficacy of eye drops to treat the DED, some authors altered cell culture conditions

to mimic the conditions given by the disease. Salzillo et al. optimized hyaluronan-based eye drop

formulation using dehydrated porcine corneal epithelial cells by removing the medium and the multiwells

Page 31: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

18

were incubated at 37 ºC and 5% CO2, without the lid for about 20 minutes [56]. Other study performed

by Meloni et al. experimentally induced in vitro dry eye on human corneal epithelium (HCE) model by

placing HCE tissues under controlled environmental conditions to mimic dryness, <40% relative humidity

at 40 ºC ± 5 ºC temperature and 5% CO2 [66].

Figure 7- Schematic representation of the in vitro cell culture model. Adapted from [65].

Table 3- Properties of selected cell culture models of ocular tissues and respective applications.

Adapted from [65].

Ocular Tissue Cell culture model Applications

Corneal epithelium Primary rabbit cells: cultured onto fibronectin/collagen/laminin coated membrane using SFM for 7-8 days

Permeability and transport studies

Immortalized human cells: HCE-T cell line, cultured on collagen-coated membranes using SFM for

6 days

Cell biology, toxicity, ocular irritancy, gene/drug delivery

Corneal endothelium Immortalized human corneal endothelial cells: IHCEn cell line, cultivated onto lyophilized human amniotic membrane

Positive expression of Na+/K+ ATPase

Conjunctival epithelium Primary rabbit cells: cultured on collagen-coated membrane using SFM for 8-10 days

Permeability and transport studies

Primary bovine cells: cultured on collagen-coated membrane, 10% serum medium for 9-11 days

Cytotoxicity screening, cytokeratin expression

Immortalized rat cells: CJ4.1A and CJ4.3C cell lines, cultured in 10% serum medium for 4 days

Investigation of antigen translocation across a mucosal barrier

Retinal pigment epithelium (RPE)

Primary isolated bovine cells: co-culture with endothelial cells for 14 days

Effect of endothelial cells on barrier function of the RPE

Primary isolated rat cells: cultured onto laminin coated filters using SFM for 5-7 days

Influence of serum on tight junction formation

Immortalized human cells: ARPE-19 cell line, cultured onto collagen-coated membrane, 10% serum medium for 9-11 days

Characterization of ARPE-19 as a human RPE cell line forming polarized epithelial monolayers

Retinal capillary

endothelium

Primary isolated bovine retinal capillary endothelial cells: cultured onto polycarbonate filters (coated with gelatin, laminin, fibronectin, and collagen)

Establishment of retinal capillary endothelial cell model

Immortalized rat retinal capillary endothelial cells: TRiBRB cell line

Functional expression of cell membrane transporters

Page 32: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

19

1.5. Risk Analysis in Medical Devices

In the pharmaceutical industry quality systems approach was hurled by de FDA with the publication of

“Pharmaceutical cGMPs for the 21st Century—A Risk Based Approach”. The aim of this publication was

to transform regulatory approaches into a science-based and risk-based approaches with an integrated

quality systems orientation. Since that time, the use of quality systems has been a massive success in

this industry [67].

It is important to understand that the risk involving the quality of the medical product is just one

component of all the possible risks associated with the product manufacture, since the components are

also at risk. The product quality should be maintained throughout the product lifecycle, which means

that all the attributes that constitute the product quality must remain consistent with those used in clinical

studies. Risk management is therefore a powerful tool to ensure high quality of the product to the patient

by providing proactive means to identify and control potential quality issues during development and

manufacturing. By applying risk management, all kind of advantages are possible such as, make better

decision if a quality problem arises, provide regulators with grater assurance of a company’s ability to

deal with potential risks and can beneficially affect the extent and level of direct regulatory oversight

[68].

The word “risk” is defined as the combination of the probability of occurrence of harm and its severity. It

is difficult to create a standard application of risk management since every case has its own

particularities, each case might perceive different potential harms, different probabilities of occurrence

and their level of severities. In the pharmaceutical industry the main concern in the quality of the product

is its safety when administrated to the patient, all the components must be within the acceptable range

in order to guarantee patients health [68–70].

In all activities of a company there is a risk involved, they manage this risk through its analysis and the

identification, when it is detected a necessary alteration. The implementation and maintenance of risk

management allows [68]:

Increase the likelihood of achieving the pretended goals;

Encourage a proactive management;

Be aware of the necessity of identifying and manage potentials risks;

Identifies opportunities and threats;

Comply with applicable legal and regulatory obligations and international standards;

Improve mandatory and voluntary reporting;

Improve governance;

Increase confidence and credibility of the organization;

Establish a reliable basis for decision-making and planning;

Use resources in the treatment of the risk effectively;

Reinforce safety and health;

Reduces losses;

Improves organizational learning and resilience.

Quality risk management has principles, namely, the evaluation of the risk should be based on scientific

knowledge and ultimately link to the protection of the patient. However, the level of effort, formality and

Page 33: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

20

documentation of the quality risk management process should be commensurate with the level of risk.

The company should elaborate and implement strategies to improve their risk management [68,69].

ISO 14971:2017 is an ISO standard specific for the application of risk management to MD to identify

hazards associated with MD, including in vitro diagnostic (IVD) MD, to estimate and evaluate the

associated risks, to control these risks, and to monitor the effectiveness of the controls during the

product life cycle [70].

1.5.1. General Process

Quality risk management process works as a map to help in the assessment, control, communication

and review of risks that may cause damage to the quality of the drug product (Figure 8). This process

should be included in the culture and practices of the organization and it must be built to fit perfectly in

the procedure that needs to be controlled [68,69,71].

1.5.2. Initiate Quality Risk Management Process

In order to define a problem or a potential risk, an assumption should be made of the potential risk,

collecting its background information and identify all necessary resources. These are the steps to initiate

a risk management process. Quality risk management should include systematic processes with the

propose of facilitate and improve science-based decision regarding the risk [68].

1.5.3. Risk Assessment

Risk assessment consists of identifying and analysing potential hazards, as well as identifying risks

associated with those hazards. This step of the Quality Risk Management Process should be performed

by a multidisciplinary team composed by engineering, quality assurance, validation and manufacturing

Figure 8 - Quality Risk Management Process overview (adapted from ICH Q9 and ISO 31000)

[68,71].

Page 34: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

21

experts, resulting in a risk question which is the main focus in the risk assessment. The brainstorming

should involve the resolution of three main questions: What can go wrong, how likely is it to go wrong,

and how severe are the consequences? By answering these questions risk identification, risk analysis

and risk evaluation, which are key characteristics of a risk assessment process, are completed [67].

1.5.4. Risk Control

Once the hazards are identified and described, a plan is developed to reduce and/or accept risks. That

plan consists in determining if the risk level is acceptable, if not what can be done to reduce or eliminate

it, if there is a balance between risks, benefits and resources and if new ricks are introduced by

controlling the initial risk. The purpose of risk control is therefore to reduce the risk to an acceptable

level [67].

1.5.5. Risk Communication

Risk Communication consist in sharing information about risk and risk management between the

decision makers and other interested parties, regulators and industries, either within or outside the

company. The output/result of the quality risk management process should be appropriately

communicated and documented, the information might relate to the existence, nature, form, probability,

severity, acceptability, control, treatment, detectability and other features of risks to quality [67–69].

1.5.6. Risk Review

A mechanism to review or monitor events should be implemented as part as the ongoing quality

management process. Once a quality risk management process has been initiated, that process should

continue to be utilized for events that might impact the original quality risk management decision,

whether these events are planned or unplanned. The frequency of any review should be based upon

the level of risk and it might include reconsideration of risk acceptance decisions [67–69].

1.5.7. Risk Management Tools

Some tools are described to structure Risk Management intended to organize data and facilitate

decision-making, some methods are simple and possible to use with general information, while others

require more information and detail. No tools or set of tools are applicable to every situation, tools must

be studied and chosen according with the specific situation in order to build a specific quality risk

management procedure [68].

Simple techniques used in risk management are basically scheming to facilitate organization and

indicate, in a simple form, the progress of the situation. These includes Flowcharts, Check Sheets,

Process Mapping and Cause and Effect Diagrams, also known as Ishikawa diagram. More complex

tools used to characterize in detail possible risks and to prioritized them include: Failure Mode Effect

Analysis (FMEA), Failure Mode, Effect and Criticality Analysis (FMECA), Fault Tree Analysis (FTA),

Hazard Analysis and Critical Control Points (HACCP), Hazard Operability Analysis (HAZOP),

Preliminary Hazard Analysis (PHA) and Risk Ranking and Filtering [68].

Page 35: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

22

The ISO 14971:2007 offer two tools to evaluate and to classify risks identified in a process, through

Qualitative analysis and through Semi-quantitative analysis.

1.5.8. The Importance of Applying Risk Analysis in Medical Devices

When manufacturing a MD it must guarantee it is safe and effective for human use. Risk analysis

involves the identification, understand, control and prevent failures that can result in hazards in the

manufacturing process of a MD. The benefits of conducting risk analysis during a MD manufacturing

process can be significant and can be used to balance some or all of the cost of implementing risk-

mitigating measures. By implementing risk analysis in a manufacturing process of a MD a higher

probability of producing a consistent product with high quality can be a possibility [67,68,70].

Page 36: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

23

Chapter Two – Previous Work

2. Previous Work - Introduction

The International Conference on Harmonization (ICH) Q8 (R2) defines pharmaceutical development as

designing a quality product and its manufacturing process who gives a consistent delivery of the

intended performance of the product. Pharmaceutical development studies and manufacturing

experience provide important information in understanding and defining a design space, a

multidimensional combination and interaction of input variables and process parameters that

demonstrated the assurance of quality of the product [72]. The ISO 13485:2016 explains in detail the

development of a MD, which in our case is an eye drop [69].

Once a promising compound for development is defined the selection of the excipients and their

assembly is decided next, which can be a complex procedure, because the choice may not be

straightforward. A robust and reproducible product manufacturing process must result from this study

[73].

Pre-formulation testing is the first step in the rational development of dosage forms. In the formulation

of artificial tear substitutes, lubrication and nutrition with extended ocular surface time needs to be

demonstrated, meaning that some key elements need to be consider [74]:

Selection of active ingredients;

Decision on salt composition and osmolality;

Selection of viscosity agents;

Choice of pH/buffering agents/ buffering capacity;

Inclusion of other ingredients;

Exclusion or inclusion of preservatives;

Avoidance of toxicity to the ocular surface.

Osmolality and pH value parameters should be within range of normal tears in order to allow full recovery

of epithelial barrier function. The residence time of tear substitutes and its stability are two of the most

important characteristics that an eye drop should have, being viscosity, surface tension and

mucoadhesive ability to the ocular surface, important aspects to maintain the tear film intact. Eye drops

are administrated many times per day and in some cases before sleep, which means drug related

toxicity is disallow because it may induce ocular surface damage [74].

The drug substance has a shelf life and optimal storage conditions. This information is provided by

stability studies, whose purpose is to provide evidence on how the quality of a drug substance or drug

product varies with time under the influence of a variety of environmental factors for instance

temperature, humidity and light. It also establishes a re-test period for the drug substance or a shelf life

for the drug product and recommended storage conditions. Stability studies data should be provided on

at least three batches of the drug product, all tree with the same formulation and packed in the same

container closure system as proposed for marketing department [75].

The testing should include physical, chemical, biological and microbiological attributes such as

preservative content and challenge test, since these attributes of the drug are susceptible to change

during storage and are likely to influence the quality, safety and efficacy of the product. This susceptibility

Page 37: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

24

to change is due to storage conditions such as temperature, light, air and humidity, as well as the

package components. These are the conditions under which the product shall be submitted in order to

establish a suitable shelf life [75,76].

The combination of small volume and aqueous medium for many ophthalmic products can result in

particular challenges, when applied stability test conditions [77]. The test conditions defined in the

guideline ICH QIA (R2) are represented in Table 4 [75].

Table 4- General case of stability testing types and respective conditions.

Study Storage condition Minimum time period covered by data at

submission

Long term 25 ± 2 °C / 60 ± 5% RH or

30 ± 2 °C / 65 ± 5% RH 12 months

Intermediate 30 ± 2 °C / 65 ± 5% RH 6 months

Accelerated 40 ± 2 °C / 75 ± 5% RH 6 months

*RH – relative humidity

A minimum of 12 months duration should be covered for the long term testing, using at least three

batches at the time of submission. The duration of the stability test should cover the proposed shelf life.

Results from these studies will form an integral part of the information provided to regulatory authorities

[75].

In this chapter it is described the necessary steps for the development and the respective characteristics

of two MD pilot batches. These MD are eye drops containing in their formulation HA, one with

concentration of 0.15% (w/v) (HA 0.15%) and the other 0.30% (w/v) (HA 0.30%) intended for the

treatment of DED.

2.1. Materials and Methods

2.1.1. Materials

High MW and low MW Sodium hyaluronate were a kind gift from Inquiaroma (Espanha). PHMB 20%

(Cosmocil) was a gift from DS Produtos Quimicos (Portugal) and N-hydroxymethylglycinate 50%

(Suttocide) from Ashland (EUA). Potassium chloride, sodium chloride, sodium tetraborate and EDTA

were purchased form VWR International (Portugal), calcium chloride.6H2O was purchased from José

Manuel Gomes Santos, magnesium chloride.6H2O was purchased from Sigma Aldrich Quimica and

acid boric was purchased from LaborSpirit (Portugal).

2.1.2. Methods

2.1.2.1. Appearance

The macroscopic appearance of each formulation was visually analysed and used as first stability

indicator.

Page 38: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

25

2.1.2.2. Determination of the pH values

The pH values were measured using a pH meter with an incorporated temperature sensor (827 pH Lab,

Metrohm, Switzerland) by inserting the electrode in the sample and read directly at a temperature

between 20°C and 25 °C.

2.1.2.3. Determination of the Osmolality values

The osmolality values were measured using a osmometer (OSMOMAT 030, Gonotec, Germany) by

collecting 50 μL of the sample using a micropipette and place it in a vial, the results were read directly

in the osmometer.

2.1.2.4. Determination of the Viscosity values

The viscosity values were measured using a viscometer (BROOKFIELD DV-I +, Brookfield, USA) by

placing a 16 mL of the sample on the UL/Y adaptor, using needle nº 0, at 6 and 12 rpm for HA 0.15%

and 0.30% and 0.6 rpm for HA 0.30%.

2.1.2.5. Efficacy of Antimicrobial Preservation

2.1.2.5.1. Strains and Preparation of Inoculum

The bacterial and fungal strains tested were obtained from the American Type Culture Collection

(ATCC): Staphylococcus aureus ATCC 6538, Pseudomonas aeruginosa ATCC 9027, Candida albicans

ATCC 10231 and Aspergillus brasiliensis ATCC 16404. Bacterial strains were grown in Tryptic Soy agar

(TSA) at 30-35°C for 18-24h and C. albicans was grown in Sabouraud-Dextrose agar (SDA) agar at 20-

25°C for 24-48h. For Aspergillus brasiliensis the inoculation was performed in SDA and incubated at 20-

25°C until good sporulation was obtained. The stock solution was obtained in a test tube with 10 mL of

suspension medium adjusting the inoculum to 108 spores/mL. A suspension was prepared for each

microorganism with 108 CFU/mL by comparing with the MacFarland scale (Remel) in suspension fluid

medium (APT) and used immediately. Series of decimal dilutions were performed by transferring 1 mL

of the stock solution to 9 mL of suspension fluid medium, until final dilution of 10-8 is reached obtained.

The number of CFU presented in each dilutions was determined by spreading 100 µL of the last counting

dilutions (10-5 to 10-7) in TSA plates for bacteria and SDA plates for fungi. The plates were incubated

until possible count.

2.1.2.5.2. Determination of the preservative efficacy of the formulation

The antimicrobial effectiveness test was executed by inoculating the two eye-drops formulations, HA

0.15% and HA 0.30%, with the microbial suspension to obtain the initial concentration of 105-106

CFU/mL of each organism. The inoculated product was stored at 20-25°C protected from light and the

samples were withdrawn from the containers at specific time intervals. A sample of 1 mL of the

inoculated product was removed at 0h, 6h, 24h, 7 days, 14 days and 28 days taking into account the

neutralization method. The number of viable micro-organisms was determined by plate count in

duplicate and negatives controls of the media and the sample in study was conduct at all times.

Page 39: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

26

Table 5- Criteria for evaluation of antimicrobial activity in terms of log10 reduction in the number

of viable micro-organisms for eye preparations [78].

NR: No recovery NI: No increase in number of viable micro-organisms compared to the previous reading

2.1.2.6. Stability Testing

Three batches of 500 mL were produce of HA 0.15% and HA 0.30% and were stored during 6 months

at room temperature (25 ± 2 °C / 60 ± 5 % RH), intermediate conditions (30 ± 2 °C / 65 ± 5 % RH) and

under accelerated conditions (40 ± 2°C / 75 ± 5 % RH).

2.1.2.7. Manufacturing Process

The manufacturing process starts with the addiction of the polymer, sodium hyaluronate, in highly

purified water. After complete dissolution of the polymer, the electrolytes potassium chloride,

magnesium chloride hexahydrated and calcium chloride hexahydrated are added and stirred until

complete dissolution. Next the sodium chloride and the buffering agents boric acid and sodium

tetraborate are added and stirred until complete dissolution. The manufacturing process finishes with

the addition of the preservative, stirred until homogenization. The pH is adjusted with hydrochloric acid

1M or sodium hydroxide 10M, if the pH level is not between 7.0-7.6, and the osmolality is adjusted with

sodium chloride if the value is outside the range of 280-320 mOsm/Kg.

2.2. Pre-Formulation Studies

2.2.1. Formulation Design

In 2015 it was developed an eye drop solution with an electrolyte composition similar to the lacrimal

fluid, with the purpose of developing an effective and safe eye drop solution for dry eye treatment. The

human eye is a very sensitive organ, which means that any type of compound that differs from its natural

environment may cause serious damages. Therefore the formulation of the saline solution must respect

all the requirements necessary to the eye comfort [79].

2.2.2. Required specification

To avoid eye irritation and provide ocular lubrication and comfort, there as some required specifications.

These specifications are the pH value, osmolality, sterility and appearance. The electrolyte composition

must be similar to the lacrimal fluid, which is mainly composed with Na+, K+, Cl- and HCO-. The tear fluid

normal pH is 7.4, consequently the formulation for an eye solution should be ideally between pH 7.0

and 7.6. The buffer should have low buffering capacity in order to allow the tears to regain the pH level

more rapidly. The osmolality of the tear film ranges between 294 and 310 mOsm/Kg [80] due to the

number of ions dissolved in the aqueous layer of the tear film. Thus, the formulation must have a similar

Log10 reduction

6 h 24 h 7 d 14 d 28 d

Bacteria A 2 3 - - NR

B - 1 3 - NI

Fungi A - - 2 - NI

B - - - 1 NI

Page 40: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

27

osmolality value to avoid an osmotic pressure differential. The specifications for the final formulation are

represented in Table 6 [79,81].

Table 6- Proposed specification for the final formulations (HA 0.15% and HA 0.30%).

Tests Specifications

Appearance Limpid, clear and odourless solution

pH 7.0 – 7.6 at 20 - 25° C

Sodium Hyaluronate Assay 90-110 %

Viscosity Under study

Osmolality 280 – 320 mOsm/Kg

Sterility Absence of growth

2.2.3. Selection of Materials

2.2.3.1. Polymer Selection

An extensive study was carried out by Charles J. White et al. for the selection of most effective polymer

to use as a comfort agent. For the selection of comfort agents, the evaluation of water retention, apparent

flow viscosity, zero-shear viscosity, intrinsic viscosity, surface tension and comfort agent index were

performed. It was concluded that polysaccharides comfort agents are more effective than acrylic comfort

agents and that the HA was the most effective comfort agent at low molecular weight (MW) and

concentration, and should be given priority when selecting comfort agents [82].

Eye drops containing HA available in the market have a concentration ranging 0.10% to 0.30%. Topical

ophthalmic solutions should exhibit viscosity to prevent drainage from the ocular surface and to prolong

residence time. However, solutions with very high viscosity may cause blurred vision and problems in

the sterile filtration system of the manufacturing process. Precorneal tear film stability was significantly

increased in patients with dry eye when treated with 0.10% or 0.30% HA solutions, reducing the

symptoms. It was decided to develop a 0.15% HA solution for daily use and a 0.30% HA solution for

more severe cases of dry eye or before sleep application [83].

2.2.3.2. Preservative Selection

The selection of the preservative is an important assignment since microbial contamination may

represent a source of infection for the patient’s eye and modify the properties of the administrated drug.

The addition of these components in the formulation is designed to kill microorganisms or to avoid

microbial growth [39]. Two types of preservatives were tested, sodium hydroxymethyl glycinate (SHMG,

Sutoccide) with disodium ethylenediaminetetraacetic acid (EDTA) and Polyhexamethylene biguanide

hydrochloride (PHMB, Cosmocil) with and without EDTA [39,84].

Page 41: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

28

2.2.3.3. Buffer Selection

The limited buffering capacity of the tear fluid is mainly due to the dissolved carbon dioxide and

bicarbonate, thus the buffer selected must have low buffering capacity and significant antimicrobial

activity. A borate buffer was evaluated for its antimicrobial activity in a study by R. Dennis Houlsby et

al., and the results demonstrated that the buffer exhibited significant antimicrobial activity against many

type for strains, with or without carbon source. However, it does not meet by it-self the criteria for

effectiveness required, other agents must be added to meet the specifications [85,86].

2.2.4. Components of the Formulation

The qualitative composition of the final formulations is represented in Table 7, which contains the

comfort agent and the excipients selected. During the pharmaceutical development a variety of

excipients were reviewed and used in the formulations in order to prepare and optimize the eventual

excipient mixture and the product properties. All the below excipients are recognized as safe materials

for human administration, being regarded as non-irritant and nontoxic on the amounts used [87–92].

Page 42: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

29

Table 7- Polymer and excipients selected for the developed formulations.

Main Function Chemical Structure Molecular

Weight Solubility

Ex

cip

ien

ts

Sodium Hyaluronate Comfort Agent

*

H2O 5mg/mL at 25°C

Highly Purified Water

Solvent

18.015 g/mol

-

Potassium Chloride

Establish an electrolyte composition similar to the tear fluid

74.548 g/mol

H2O 35.5g/100mL

Magnesium Chloride Hexahydrate

Establish an electrolyte composition similar to the tear fluid

167.845 g/mol

H2O 1.670 g/L at 20°C

Page 43: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

30

Table 7- Polymer and excipients selected for the developed formulations (Continuation).

Main Function Chemical Structure Molecular

Weight Solubility

Ex

cip

ien

ts

Magnesium chloride.6H2O

Establish an electrolyte composition similar to the

tear fluid

219.068

g/mol

H2O 81.1 g/100

mL at 25°C

Anhydrous Sodium Chloride

Adjust osmolality

58.44 g/mol

H2O 36g/100mL at 25°C

Boric Acid

Buffering agent

61.831 g/mol

H2O 50 mg/mL at 25 °C

Sodium Tetraborate

Buffering agent

201.22 g/mol

H2O 26 g/L at 20 ºC

Page 44: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

31

Table 7- Polymer and excipients selected for the developed formulations (Continuation).

*Confidential information

Main Function Chemical Structure Molecular

Weight Solubility

Ex

cip

ien

ts

EDTA Preservative booster

292.244 g/mol

H2O 1g/mL at 25 °C

Cosmocil (PHMB 20%)

Preservative

185.275

g/mol

426 g/L at 25 °C

Suttocide (N-

hydroxymethylglycinate 50%)

Preservative

127.075

g/mol

H2O 1g/mL at 25 °C

Page 45: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

32

2.2.5. Development of the Laboratory Batches

Initially 12 formulations were developed, 6 formulations regarding 0.15% of the HA and the other 6 with

0.30% of the HA. The formulations differed on the sodium hyaluronate MW, the addition of EDTA and

the type of preservative. After the measurements of pH, osmolality and viscosity values some

formulations suffered some adjustments in order to respect the specific ranges of each measure. It was

concluded only 4 of the initial 12 formulations passed the defined criteria proposed on Table 6 and

selected for the efficacy of antimicrobial preservation test. From the chosen formulations, 3 of them

present 0.30% concentration of HA, 2 with Sutoccide as a preservative and the other with Cosmocil.

The remaining formulation has 0.15% concentration of HA with Sutoccide as preservative.

2.2.6. Efficacy of Antimicrobial Preservation

The selected formulations were subjected to an efficacy of antimicrobial preservation test, in order to

investigate if the chosen preservative is the most adequate. The preservative properties of the product

are adequate if there is a significant fall or increased in the number of microorganisms in the inoculated

product after the times and the temperature recommended. According to the European Pharmacopeia

to ophthalmic preparations, the formulations must respect the criteria of the evaluation of antimicrobial

activity reduction on the number of vial microorganisms specifically described [93].

Once collected and analysed the results, according to Table 5 none of the formulations passed the

required criteria A, but three of them passed the criteria B.

2.3. Final Formulation

Based on the physic-chemical and microbiological results, it was decided that the final formulation has

Suttocide (N-hydroxymethylglycinate 50%) with EDTA as a preservative. Thus, excipients used in the

manufacturing process of HA 0.15% and 0.30% eye drops solution, 8 mL solution are: Sodium

Hyaluronate, Potassium chloride, Magnesium chloride.6H2O, Monosodium Phosphate monohydrate,

Disodium Phosphate dodecahydrate, Sodium Chloride, Calcium chloride.6H2O, Anhydrous Sodium

Chloride, Boric acid, Sodium tetraborate, Suttocide (N-hydroxymethylglycinate 50%), EDTA, Highly

Purified Water, Sodium hydroxide (NaOH 40%) and Hydrochloric acid (HCl 10%). All these excipients

will be further analyzed in the next section of the present report.

2.3.1. Polymer

2.3.1.1. Comfort Agents

Comfort agents are used extensively within topical eye drop formulations promoting comfort through

different mechanisms of action such as retaining tear volume by reducing drainage rates, stabilizing the

tear film, changing tear film surface tension, preventing tear evaporation, and altering tear fluid viscosity.

Frequent application of comfort agent in eye drops can greatly increase the level of comfort perceived

by consumers. Their molecules function in order to relieve ocular discomfort by enhancing

characteristics of the tear film, stabilizing and retaining tear volume and lubricating the ocular surface.

There are two broad classes of comfort agents: polysaccharide comfort agents and acrylic comfort

agents. Polysaccharide comfort agents are typically macromolecules composed of one or more types

Page 46: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

33

of monosaccharide. Polysaccharide comfort agents are typically linear, hydrophilic, and possess high

MW. Substitution along the polymer backbone is common and can affect the overall conformation of the

macromolecule, particularly at high degrees of substitution. These substitutions can be branches, alkyl

groups, functional groups, or even salt complexes. Solution viscosity of polysaccharide comfort agents

is typically high and present a shear-thinning behavior. In general, all polysaccharides have high water

affinity and high rheological-modifying properties. This type of comfort agent was chosen for the study

formulation. Acrylic comfort agents are linear chains composed of carbon–carbon backbones with

regular repeat units, often including at least one functional group. This type of comfort agents can be

used as polyelectrolytes or in the neutral state. This category also includes polyacids, which are less

used than other agents but have slightly increased water retention properties when compared to neutral

acrylic agents [47,82,94].

2.3.1.2. Hyaluronic Acid

Since its discovery, HA, also called hyaluronan, has received great attention as a versatile and highly

functional biopolymer (Figure 9). HA is evolutionarily conserved from simple prokaryotes all the way to

complex eukaryotes, which is an undeniable testament to its biologic relevance. Structurally, HA is not

inert. The native form, high MW, can be broken down into smaller MW fragments in response to

glycosidase activity upregulated by environmental cues, such as pH and reactive oxygen species. The

MW variants have been used in a variety of biomedical applications, eliciting varying biologic responses.

For instance, low MW of HA promotes the production of inflammatory mediators. Similarly, high MW of

HA inhibits production of pro-inflammatory mediators, suggesting differential macrophage activation by

different molecular weight polymers, even dough the same molecule [56]. Clinically, HA has been used

in several applications, including ophthalmology as a drug delivery system and lubricant, in osteoarthritis

for viscosupplementation and as a dermal filler [95].

Each HA molecule consists of as many as 50000 repeats of the simple disaccharide glucuronic acid β

(1→3) N-acetylglucosamine β (1→4). There is one ionisable carboxyl group per disaccharide. Individual

segments of an HA molecule fold into a stiff rod-like conformation because of the β linkages between

the saccharides and the extensive intrachain hydrogen bonding between adjacent sugar residues. Due

to the large number of hydrophilic residues on its surface, HA binds a large amount of water and forms

a viscous hydrated gel. The first isolation of the compound was in 1934 by Meyer and Palmer extracted

from vitreous humour of 100 cattle eyes, but it can also be found in skin, umbilical cord, cartilage and

synovial fluid [96–98].

Figure 9 - Chemical repeat unit of HA [96].

Page 47: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

34

Sodium hyaluronate is the sodium salt of HA (Figure 10) and it is a natural polymer comfort agent, class

of polysaccharides, used in many ophthalmic preparations. Its appearance is a white to beige powder,

highly soluble in water with a melting point of 241-247°C [99].

Figure 10 - Sodium hyaluronate chemical structure [100].

This salt is commonly used as a bioavailability-enhancer in eye drops. In the presence of HA, the

precorneal residence times of pilocarpine, timolol, aceclidine, tropicamide, arecoline, gentamicin, and

tobramycin were pro-longed. In addition to its viscosifying and mucoadhesive properties, HA has other

beneficial effects on the corneal epithelium, including protection against dehydration, reduction of

healing time, reduction of the inflammatory response caused by dehydration, and lubrication of the

ocular surface. Due to this clinical efficacy, HA is largely used in ophthalmology not only as an excipient

but also as the main component of the artificial tear substitutes commonly prescribed for the treatment

of DED [56].

2.3.2. Excipients

2.3.2.1. Highly Purified Water

Highly purified water is used as a vehicle and for dissolving substances, it should be prepared from

drinking water as a minimum-quality feed-water. This type of water is a unique speciation for water found

only in the European Pharmacopoeia. It must meet the same quality standard as water for injections,

including the limit for endotoxins (not more than 0.25 IU of endotoxin per mL), but the water-treatment

process used may be different. Quality standards for this type of water meet the same standards as for

water for injections but he production methods are considered less reliable than distillation and thus it is

considered unacceptable for use as water for injections. Current production methods include, for

example, double-pass reverse osmosis coupled with other suitable techniques such as ultrafiltration and

deionization [101,102]. Edol´s facility uses reverse osmosis and deionization.

2.3.2.2. Potassium Chloride, Magnesium Chloride.6H2O and Calcium Chloride.6H2O

One of the many tears function is to provide the entire ocular surface with a moist environment with the

appropriate electrolyte composition. The ocular surface has a narrow range of pH, osmolality, and ionic

concentrations necessary for optimal function. Small changes in these variables, especially osmolality

and ion concentration lead to ocular surface disease. Tear osmolality is derived from the ionic

composition of tears, which is unique when compared to plasma or other body fluids. Tears contain Na+,

K+, Cl- , HCO-, Ca2+ and Mg2+, and trace levels of other ions. Tears have higher K+ and Cl- concentration

Page 48: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

35

and similar Na+ concentration compared to plasma [103]. The selected salts were chosen to establish

an electrolyte composition similar to the tear fluid.

2.3.2.3. Boric Acid and Sodium Tetraborate

Gram-negative rods represent some of the most common contaminants of commercially produced eye

care products. Many of these products are formulated in a phosphate buffer preserved with

benzalkonium chloride. Others, designed for use with hydrogel contact lenses, are preserved with

thimerosal, chlorhexidine, or sorbic acid. Inexpensive and widely available, these eye care products are

often subjected to widespread consumer abuse. Their preservative systems are weak in comparison to

those of topical products because of the extreme sensitivity of ocular tissues to chemicals in general.

Studies have shown the borate-buffered vehicle appears better suited for use in ophthalmic products

than phosphate-buffered formulations. The lack of proliferation and survival of microorganisms in a

borate-buffered vehicle provides an increased level of safety for ophthalmic products. However, this

preservative system does not, by itself, meet the criteria for effectiveness. Thus, the borate buffer

requires augmentation by other agents to meet the specifications [86].

2.3.2.4. Suttocide (N-hydroxymethylglycinate 50%) and EDTA

Suttocide is a colourless to yellow liquid with characteristic amine odour sold as a 50% aqueous solution.

Highly soluble in water, methanol propylene glycol, glycerine, but insoluble in most organic solvents.

This preservative releases formaldehyde when decomposed in an aqueous solution. One molecule of

formaldehyde is formed by the decomposition of each molecule of sodium hydroxymethylglycinate, as

described in the equation below [92].

(2)

It is a broad-spectrum preservative with good antimicrobial properties, active against Gram-negative,

Gram-Positive bacteria, yeast and molds. It has good anti-bacterial and anti-mold properties, but it is

weak against yeast. Multidose artificial tears containing the mixture of 0.002% sodium hydroxymethyl

glycinate (SHMG) and 0.10% EDTA as a novel preservative have been marketed in Italy recently. Since

these solutions are claimed to have a mild impact on the ocular, an evaluation was made whether

different concentrations of SHMG alone or in combination with EDTA could be an efficient preservative

for different ophthalmic preparations. It was demonstrated that SHMG shows a wide bacteriostatic

activity at concentrations ranging from 0.0025% to 0.0125% and that higher concentrations of SHMG

(0.25% or 0.5%) are needed for the fungistatic activity against C. albicans and A. brasiliensis. The

addition of 0.10% EDTA to SHMG substantially reduces the minimal concentration of SHMG required

for both bacteriostatic/fungistatic and bactericidal/ fungicidal activity in vitro. In fact, in this condition,

0.005% and 0.025% concentrations of SHMG were, respectively, sufficient to inhibit growth or to kill all

tested organisms [84]. The addition of EDTA has broaden even more the preservative antimicrobial

Page 49: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

36

spectrum. These compost is a stabilizing and chelating agent which helps in restricting metal-catalyzed

oxidation of various drugs and has been proven to promote the action of antimicrobial preservatives

[104].

2.4. Manufacturing Process of the Final Formulation

The manufacturing process of the final formulation starts with the introduction of highly purified water in

an appropriate recipient and the addition of sodium hyaluronate stirred until complete dispersion. The

description of this manufacturing process applies to the product HA 0.15% and HA 0.30%. Afterwards

occurs the addition of potassium chloride, magnesium chloride hexahydrated and calcium chloride

hexahydrated and stirred again until complete dissolution. Sodium chloride, boric acid, sodium

tetraborate and EDTA are added and stirred at the same conditions as before or until complete

dissolution. The final step is the addition of the preservative Suttocide (N-hydroxymethylglycinate 50%),

which is stirred, once again, until complete homogenization. The pH and the osmolality levels are

adjusted with NaOH 40% and HCl 10%, and sodium chloride, respectively.

2.5. Stability Testing

Samples of HA 0.15% and HA 0.30% were analysed for macroscopic appearance, pH, osmolality,

viscosity, density and sodium hyaluronate identification before the storage period and on months 1, 3

and 6 of storage. The six batches, three of each product, remained within specifications after 6 months.

2.6. Scale-Up

Once the final formulation and its respective manufacturing process is defined, the scale-up project was

studied. Scale-up is generally defined as the process of increasing the batch size and can also be

viewed as a procedure for applying the same process to different output volumes. However, batch size

enlargement does not always translate into a size increase of the processing volume. In mixing

applications, scale-up is undeniably concerned with increasing the linear dimensions from the laboratory

to the plant size. It is then necessary and very important to understand the type of procedure applied, in

which an increase of the scale may be counterproductive and scale-down is required to improve the

overall quality of the product. The passage between R&D to production scale, it is essential to have an

intermediate batch scale called the pilot scale, which is defined as the manufacturing of product by a

procedure fully representative of the manufacturing scale. Nevertheless, inserting an intermediate step

between R&D and production scales does not in itself guarantee a successful transition. Despite of a

well-defined process that may generate a product according to the specifications in both the laboratory

and the pilot plant, it may fail quality assurance tests in production [105].

In order to avoid the repetition of lengthy and costly tests, it is necessary to gather information during

properly designed development and process optimisation studies, when scaling up from laboratory

through pilot to production scale. Such information provides the basis for justification that scale-up can

be achieved without a consequent loss in quality. Therefore, the aim of the pilot batch scale is to provide

predictive data of the production scale product. It should be notice that the pilot batch size should

correspond to no less than 10% of the production scale batch, at least [106].

Page 50: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

37

A flow chart of HA 0.15% and HA 0.30% solutions pilot batch scale production is represented in Figure

11.

With this manufacturing process, a detailed Risk Analysis study was made in order to identify possible

risks which may compromise the final quality of the product (Chapter Three – Risk Analysis). The final

product resultant from this manufacturing process was subjected to physical-chemical studies to

determine the characteristics that the product possesses (Chapter Four – Pilot Scale Batches Process

Validation).

Figure 11- Flow chart of the pilot batch manufacturing process of HA 0.15% and HA 0.30%.

Page 51: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

38

2.7. Conclusion

In 2015 it was developed a formulation that allowed the determination of suitable excipients to produce

an eye drop solution for dry eye treatment. During the product development a study of the possible

excipients used in eye drops formulation was performed, were three main groups where the primary

focus: the choice of the polymer, the preservative and the buffer. After some research and with the

development of the laboratory batches it was decided that the final formulation contains the polymer HA,

the preservative Suttocide and EDTA and boric acid and sodium tetraborate as a buffer. With the final

formulation decided the scale-up of the product was designed. Along the course of the production a Risk

Analysis was made and with the products resulting from the scale-up production an IPC and FPC study

was performed among with the characterization of the final product. These studies resulted in the

following chapters.

Page 52: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

39

Chapter Three – Risk Analysis 3. Risk Analysis - Introduction

Ophthalmic products, such as eye drops, require well-defined specifications. Since the eye is a sensitive

organ it is necessary that the eye drop’s formulation must present physical-chemical characteristics

similar to the lacrimal fluid and it must be sterile, in order to avoid eye damages and infections [74].

During the manufacturing process the product may be exposed to various factors that may put at risk

its final quality. The manufacturing of sterile products requires special manipulation in order to minimize

risks of microbiological contamination, which depends on the training and skills of the personal involved

[107]. By applying risk analysis in the manufacturing process a better understanding of what may be the

primary causes that may contribute to the production of a non-conform product can be achieved,

allowing the management of potential risks [71].

The creation of risk management tools, especially tools for a specific product, encourages and

demonstrates the importance of using risk analysis in the manufacturing process of a product. ISO

14971:2017 establishes the requirements for risk management to determine the safety of a MD by the

manufacturer during the product life cycle [70].

Failure Mode Effect Analysis (FMEA)

FMEA is one of the most commonly used methods for pharmaceutical risk assessment, it is a step-by-

step approach for identifying all possible failures in design, manufacturing or assembly process, or a

product. It provides for an evaluation of potential failure modes for processes and their likely effect on

outcomes and product performance. Once the specific operation is studied, failure modes are

established, by implementing risk reduction the potential failure is may be eliminated, contained,

reduced or controlled. It is a team-based structure risk assessment method that can assign a numerical

Risk Priority Number (RPN) based on the severity of the risk, consequently this method is dependent

on the expertise of the team members. This method is more applied to processes that do not have

several sub processes. Failures should be prioritized according to how serious their consequences are,

how frequently they occur and how easily they can be detected. Action to eliminate or reduce failures

should begin with those with the highest priority [67,68,108,109].

It consists in building a table with specific columns such as:

Potential Failure Modes Colum: List of potential failures that may occur in a specific situation.

Modes can be broken down into the following categories: Total failure, partial failure, intermittent

failure, over-function and unintended function;

Effects of Failure Column: Description of the possible effects of the failure;

Severity (SEV) Column: The severance of the failure. Severity is a numeric ranking of the

seriousness of the failure. The number shall be assigned using the definition given in the FMEA

Score Sheet. The FMEA Score Sheet is a table containing the ranking criteria for the SEV, OCC

and DET;

Potential Cause of Failure Column: What are the causes or mechanisms of the failure;

Page 53: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

40

Occurrence (OCC) Column: The frequency of the failure. Occurrence is a numeric ranking of

the probability of the cause for the failure occurring, also assign by the FMEA Score Sheet. The

occurrence is evaluated relative to the likelihood of the failure occurring when it is caused;

Control Column: Current system controls on place to pervert the failure mode, there two types

of design controls: Prevention or Detection of the cause/mechanism of failure or the failure

mode. The Prevention control prevents the failure from occurring or reduce the rate of

occurrence. The Detention control leads to a corrective action when the failure occurs.

Detection (DET) Column: The likelihood of the failure detection. Detection is a numeric ranking

of the ability of the design to detect a potential cause/mechanism and subsequent failure mode,

it is also assign by the FMEA Score Sheet.

Risk Priority Number (RPN): Number that results in the multiplication of SEV, OCC and DET

scores, the resulting number helps prioritize risks our actions for problem resolution.

𝐑𝐏𝐍 = 𝐒𝐄𝐕 × 𝐎𝐂𝐂 × 𝐃𝐄𝐓 (3)

Criticality Index (CRIT): Index that helps prioritizes risks or actions when the RPN is equal, given

greater emphasis to the SEV and frequency OCC.

𝐂𝐑𝐈𝐓 = 𝐒𝐄𝐕 × 𝐎𝐂𝐂 (4)

Analysis and Recommended Corrective Actions Column: After the calculation of all RPNs and

CRIT, recommended actions should be taken in order to reduce the overall RPN for failure

modes that era considered unacceptable or intolerable.

ISO 14971:2007 — Application of Risk Management to Medical Devices

ISO 14971:2007 specifies in identifying risks associated with MD, including in vitro diagnostic MD, to

estimate and evaluate the associated risks, to control these risks, and to monitor the effectiveness of

the controls. The requirements given in this norm are applicable to all stages of the life-cycle of a MD

and do not apply to clinical decision making [70].

Regarding Risk Management, this norm describes in detail the general requirements for risk

management process thought out the MD life cycle, which includes risk analysis, evaluation, control and

risk review in the production and post-production phases. It is given guidance in how to use risk concepts

important for managing the risks associated with the product and an example of a qualitative analyses

[70].

3.1. Materials and Methods

3.1.1. Materials

Sodium hyaluronate was a kind gift from Inquiaroma and N-hydroxymethylglycinate 50% (Suttocide)

from Ashland (EUA). Potassium chloride, sodium chloride, sodium tetraborate and EDTA were

purchased form VWR International (Portugal), calcium chloride.6H2O was purchased from José Manuel

Gomes Santos (Portugal), magnesium chloride.6H2O was purchased from Sigma Aldrich Quimica

(Portugal) and acid boric was purchased from LaborSpirit (Portugal).

Page 54: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

41

3.1.2. Methods

The steps to design an FMEA method and a Qualitative Analysis according to the ISO 14971:2007 are

described below.

3.1.2.1. Process Description

A thorough study of the process benefits in identifying the potential risks and failures associated with

the quality of the final product. A flow diagram presented in Chapter Two, section 2.6, was used to

facilitate the identification of the potentials failure modes and to understand the whole process.

The qualitative composition of HA 0.15% and HA 0.30% eye drop solution as well the function of each

excipient are described in Table 7. The equipment used in the manufacturing process are also listed

below in Table 8.

Table 8- Main equipment used during the manufacturing process of HA 0.15% and HA 0.30% eye

drop solution.

Equipment Brand Model

Mixer SEITE-WERKE DB 110 A FW

Filling and encapsulating IMA F57

Sealing test Bonfiglioli PKV 212

HPLC Hitachi LaChrom Elite

pH meter Metrohm 827 pH LAB

Osmometer Gonotec Osmomat 030

Osmomat 3000

Viscometer Brookfield DV-II + PRO

Laminar flow cabinet ADS Laminaire Optimale 12

3.1.2.2. Identification of Critical Points

Six process steps were chosen as critical points of the process since a risk in these steps may

compromise the product quality. For each process step, numerous risks were identified.

3.1.2.3. Failure Cause and Effect of each Failure Mode

Potential causes and effects for each failure mode were described. Each step, except In Process

Control/Finished Product Control (IPC/FPC) and Storage, were subjected to a detailed study by

designing Ishikawa diagrams.

Page 55: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

42

3.1.2.4. Severity Rating Assignment

A Severity rating was given to each effect from 1 to 10, with 10 being the most severe. This classification

was made by Edol’s collaborators from surveys design for each step. The final results correspond to the

mean, standard deviation (SD) and a weighted arithmetic mode of the collaborators' answers.

3.1.2.5. Occurrence Rating Assignment

The failure frequency was determined and rated appropriately from 1 to 10, being 10 the most likely,

after collecting data on the factors responsible for the failure. The classification was prepared the same

way as described previously in section 3.1.2.4 Severity Rating Assignment.

3.1.2.6. Detection Rating Assignment

A list of all controls currently existing in order to prevent each failure from occurring was prepared and

a Detection rating was assigned for each failure from 1 to 10, being 10 being a low likelihood of

Detection. The classification was prepared the same way as described previously in section 3.1.2.4

Severity Rating Assignment.

3.1.2.7. Risk Priority Number Calculation

RPN was calculated by multiplying the Severity rating by the Occurrence rating by the Detection rating.

3.1.2.8. Failure Modes Prioritization

The prioritization of the failure modes identified was dependent on their RPN number. The higher the

RPN number, the higher the risk. To distinguish failures which RPN is equal the CRIT number was

calculated by multiplying the Severity number by the Occurrence number.

3.1.2.9. ISO 14971:2007 Qualitative Analysis

Qualitative Analysis was performed using ISO 14971:2007 Annex D, section D.3.4.1 [70]. An N-by-M

matrix approach was used to describe the probabilities and severities of the risk associated with each

hazardous situation, defining N levels of probability and M levels of severity. To classify each failure

modes in terms of severity and probability levels, the common terms of the table below were applied

(Table 9).

Table 9- Qualitative Severity and Probability Levels.

Level Common Terms Possible description

Severity

Significant Death or loss of function or structure

Moderated Reversible or minor injury

Negligible Will not cause injury or will injure slightly

Probability

High Likely to happen, often, frequent

Medium Can happen, but not frequently

Low Unlikely to happen, rare, remote

Page 56: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

43

3.1.2.10. Corrective Actions

For each failure mode was determined the action to be taken in order to reduce or eliminate the risk.

3.1.2.11. Risk Review

After the application of the corrective action, the classification of the Severity, Occurrence and Detection

rating was performed once more with the purpose of determining if the failures suffered a general

reduction or elimination.

3.2. Results

3.2.1. Ishikawa diagrams

Six steps of the manufacturing process were identified as potential critical points to ensure quality of the

final product. The chosen steps were Weighting, Mixing, Filtration, IPC/FPC, Filling/Packing and

Storage. It was decided to use Ishikawa diagrams to help with the brainstorming and to list potential

causes that may induce errors in each specific step. Four primary causes were used to construct the

diagrams, following the 4M’s: Manpower, Methods, Machinery and Material. An adaptation was made

to Operators, Methods, Equipment and Raw Material. However, two steps were not subjected to the

diagrams, IPC/FPC and Storage, since the four primary causes are not applicable. In the case of

IPC/FPC, the risks identified were based on various possible results from the analysis performed.

Possible risks that may occur on the Storage step were obtained from Patel et al. [107] and through

experience from Edol’s collaborators. The Ishikawa diagrams are presented in Appendix Figure A 1,

Figure A 2, Figure A 3 and Figure A 4.

3.2.2. FMEA of Hyaluronic Acid Pilot Batch Manufacturing Process

After brainstorming possible risks from each process and constructing ishikawa’s diagrams a FMEA

table was created. In order to classify each risk presented, key activities, such as, understanding the

impact of the risk, ranking the significance of risk by scoring 1 to 10 (Table 10) and the calculation the

RPN and CRIT were performed during risk analysis.

Page 57: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

44

Table 10- Severity, Occurrence and Detection table with respective risk classification.

In Table 10 it is also present the classification regarding the ISO 14971:2007 Qualitative Analysis. It was

decided that values between 1 and 3 are considered as Negligible and Low, between 4 and 6 as

Moderate and Medium and between 7 and 10 as Significant and High in the classification of the Severity

and Occurrence aspect, respectively.

For the statistical treatment of the results obtained from the surveys it was decided to do the mean, SD

and a weighted arithmetic mode of the responses. In the weighted mode a score or weighting factor was

given to each collaborator depending on their academics level and years of work experience. In Table

11 it is shown the specification needed for each weighting factor.

Table 11- Weighting Factor and respective specification according to the years of work

experience and academic level.

Weighting Factor Years of Work Experience Academic Level

3 ≥ 3 College Degree

2 < 3 College Degree

≥ 3 Technical Course

1 < 3 Technical Course

Severity Occurrence Detection

Le

ve

l

Effect Criteria:

Severity of effect

ISO

14

97

1

Le

ve

l

Probability of failure

Criteria: Likelihood

of occurren-

ce ISO

49

71

Le

ve

l

Detection

Criteria: Likelihood

of detection

10 Dangerously

High Safety regulatory

consequences

Sig

nific

an

t

10 Almost certain Failure is

almost inevitable

Hig

h

10 Absolute

uncertainty

No design control or

no detection

9 Extremely

High 9 Extremely 9

Very remote

Very remote

detention

8 Very high

High degree of dissatisfaction

of Quality

8 Very high Repeated

failures

8 Remote Remote

detention

7 High 7 High 7 Very low Very low detention

6 Moderate

Mo

de

rate

6 Moderate to

high

Occasional failure

Me

diu

m

6 Low Low

detention

5 Low Slight

Dissatisfaction of Quality

5 Moderate 5 Moderate Moderate detention

4 Very Low 4 Low to

moderate 4

Moderately high

Moderately high

detention

3 Minor Minimum

effect

Neg

ligib

le 3 Low

Relatively few failures

Lo

w

3 High High

detention

2 Very minor 2 Very low 2 Very high Very high detention

1 None No effect 1 Remote Failure is unlikely

1 Almost certain

Almost certain

detention

Page 58: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

45

In order to obtain the maximum score, the collaborator needs to have a college degree and work

experience equal or over 3 years. If the collaborator has a college degree but work experience under 3

years, the given score is the same as a collaborator with a technical course with work experience equal

or over 3 years. The minimum score is given to a collaborator who has a technical course and work

experience under 3 years.

The weighted mode was created as a necessity to distinguish knowledge background from the

collaborators and to give more value to those who have a college degree, since collaborators with a

higher education understood and responded to the surveys more easily and with more carefully chosen

answers then those who did not possessed that type of education. If the results were only analysed

trough simple arithmetic mean and SD the results would not make sense in cases which the SD was

too elevated. For that reason the mode is preferred over the mean when describing categorical data.

The greatest frequency of responses is important for describing categorical data because, for instance,

classifying a risk with severity 3 is very different than classifying as 4, the first one is considered to have

a minimum effect in the quality of the product as the second, demonstrating slight dissatisfaction. Twelve

collaborators responded to the surveys tacking into account their function in the manufacturing process.

The FMEA and the ISO 14971:2007 qualitative analyses of the manufacturing process of HA 0.15% and

0.30% is presented in the Appendix in Table A 1, with the calculation of the RPN and CRIT for each

failure mode detected.

After the calculation of the risk score, the results of the level or priority of the risk were estimated using

a Pareto chart (Figure 12). Using the RPN scores of the failure modes, they may be ordered from who

possesses the highest risk to the lowest risk. The Pareto chart facilitates the visualization of the different

risks degrees, which helps the identification of the highest risks and the decision making of in which

value of RPN is considered to be a high level risk.

Page 59: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

46

Figure 12- Pareto chart of every failure mode of the manufacturing process of HA 0.15% and HA 0.30%.

0,00%

10,00%

20,00%

30,00%

40,00%

50,00%

60,00%

70,00%

80,00%

90,00%

100,00%

0

20

40

60

80

100

120

RP

N

Failure Modes

Risk Prioritization

Accumulated Percentage

Page 60: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

47

Form the analysis of the Figure 12 it was decided that the values in which the risk is considered to be

unacceptable starts at RPN 80 and intolerable at RPN 300 (Table 12). Over this RPN value every failure

mode needs to be mitigated, thus, risks below this value are considered to be acceptable risks. From

this decision the construction of a RPN matrix was possible. The ISO 14971:2007 Qualitative Analysis

already presents a risk matrix, using the probability as rows and the severity as columns a 3x3 risk

matrix which is represented in Table 13. This classification is already present in the FMEA table.

Table 12- RPN matrix.

Seve

rity

1 1 2 3 4 5 6 7 8 9 10 1

Detectab

ility

2 4 8 12 16 20 24 28 32 36 40 2

3 9 18 27 36 45 54 63 72 81 90 3

4 16 32 48 64 80 96 112 128 144 160 4

5 25 50 75 100 125 150 175 200 225 250 5

6 36 72 108 144 180 216 252 288 324 360 6

7 49 98 147 196 245 294 343 392 441 490 7

8 64 128 192 256 320 384 448 512 576 640 8

9 81 162 243 324 405 486 567 648 729 810 9

10 100 200 300 400 500 600 700 800 900 1000 10 1 2 3 4 5 6 7 8 9 10

Occurrence

Acceptable Risk Unacceptable Risk Intolerable Risk

Table 13- ISO 14971:2007 risk matrix [70].

Severity

Negligible Moderate Significant

Occurrence

High High Risk

Medium Low Risk

Low

Page 61: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

48

3.2.3. Failure Modes Prioritization and Corrective Actions

After the classification of risk level for each failure mode according to the RPN matrix and risk matrix

tables, prioritization of all the detected risks was made. Using the FMEA method the failure modes were

distinguished by their RPN values and CRIT values, the higher the RPN and the CRIT value the higher

the risk. In some cases the CRIT values were equal, in this case the highest risk chosen was the one

who presented a higher severity value. The risks obtained through the ISO 14971:2007 are not possible

to distinguish, since the only criteria available is the classification, which is not a numeric value. The

FMEA and the ISO 14971:2007 methods have detected two risks in common, however the FMEA

detects two more unacceptable risks comparable to the ISO 14971:2007 method. Corrective actions in

order to reduce the risk of each failure mode were applied.

Table 14- Unacceptable risks obtained through FMEA and current and corrective action.

FMEA RPN CRIT Current Process Control Corrective Action

Elevated Microbial

concentration/ non-

sterile (IPC and FPC) 108 36

The bioburden is determined/ Sterility test

Bioburden of the Validation Batch top and bottom at 0h and

24h after the manufacture

Procedure failures

(Weighting) 84 28 Required filling of the Batch

Master Record Triple check of all weights

Velocity Failure

(Mixing) 84 21 Backup shaker Visual verification of the

dissolution and registration of the same

Misuse use of uniform

(Mixing) 84 21 Uniform procedure Extra environmental controls

Product harvest in an

inappropriate container

(IPC and FPC) 81 27 The bioburden is determined

Creation of a product harvesting procedure where the type of

flask to be used is explicit

False Results (IPC and

FPC) 80 20 All methods are previously

validated Verification by the Quality

Control manager

Raw material loss

during transference

(Mixing) 80 20 Proper training

Specific training for the manufacture of the product /

Monitoring of the manufacture by a technician

Contaminated Filter

(Pre and Sterile

Filtration) 80 16

Sterilization of the filter in an autoclave with a valid load whenever there is a filling

Verification by the Production manager

IPC/FPC - In Process Control/Finished Product Control

Page 62: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

49

Table 15- Unacceptable risks obtained through ISO 14971 qualitative analyses and current and

corrective action.

ISO 14971 Classification Current Process Control Corrective Action

Elevated Microbial

concentration/ non-

sterile (IPC and FPC) High Risk

The bioburden is determined/ Sterility test

Bioburden of the Validation Batch top and bottom at 0h and

24h after the manufacture

Procedure failures

(Weighting) High Risk Required filling of the Batch

Master Record Triple check of all weights

High/Low pH (IPC and

FPC) High Risk

pH meter is calibrated daily. pH specified in batch record and pH

readings are double verified. After all adjustments Quality Control

confirms

Identification of an OOS. An investigation must be performed

High/Low osmolaliry

(IPC and FPC) High Risk

Osmometer is calibrated monthly against osmolality patterns

solutions (purified water, 300 mOsm/kg and 150 mOsm/kg). It

is calibrated daily against a reference solution with 300

mOsm/Kg. Osmolality specified in batch record and osmolalities

readings are double verified. After all adjustments Quality Control

confirms

Identification of an OOS. An investigation must be performed

High/Low Viscosity

(IPC and FPC) High Risk

Viscometer is calibrated annually by a certified external entity.

Viscosity specified in batch record and viscosities readings are

double verified. After all adjustments Quality Control

confirms

Identification of an OOS. An investigation must be performed

Sodium Hyaluronate

UV Assay out of range

(IPC and FPC) High Risk

Sodium Hyaluronate percentage specified in batch record is

determined by a validated UV/Vis method. Assay is performed

twice.

Identification of an OOS. An investigation must be performed

IPC/FPC - In Process Control/Finished Product Control; OOS - Out of Specification.

3.2.4. Risk Review

The implementation of the corrective actions for each failure mode has the aim of reducing the SEV,

OCC or the DET parameters. All the failure modes detected by the FMEA method and the first two

detected by the ISO 14971:2007, the corrective actions effects the DET value, since they present a

detective nature. In the risk detected in the Mixing Process by the FMEA method “Raw material loss

during transference” the implemented corrective action not only effects the DET value but also the OCC,

since implementing specific training diminishes the probability of occurrence. The rest of the risks

identified by the ISO 14971:2007 are not possible to implement corrective action since they are

analytical results. When a result is an OOS it is not expected, meaning that the value obtained does not

obey the respective specification, in this case an investigation must be opened. This investigation should

result in a well-documented and exhaustive report in order to determine the causes of the result.

Page 63: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

50

The ISO 14971:2007 quantitative analysis does not suffer alteration because it only classifies the risk

according the severity and the occurrence. In the FMEA method by reducing the DET and the OCC

parameter by one value the RPN was once again calculated, the results are presented in Table 16.

Table 16- RPN calculation after the implementation of the corrective actions.

FMEA SEV OCC DET Previous

RPN New RPN

Reduction (%)

Elevated Microbial concentration/ non-sterile (IPC and FPC)

9 4 2 108 72 33

Procedure failures (Weighting) 7 4 2 84 56 33

Velocity Failure (Mixing) 7 3 3 84 63 25

Inadequate clothing (Mixing) 7 3 3 84 63 25

Product harvest in an inappropriate container (IPC and FPC)

9 3 2 81 54 33

False Results (IPC and FPC) 10 2 3 80 60 25

Raw material loss during transference (Mixing)

5 3 3 80 45 44

Contaminated Filter (Pre and Sterile Filtration)

8 2 4 80 64 20

All the new calculated RPN are below 80 meaning that the previous unacceptable risks were reduced

to acceptable risks after the implementation of the corrective actions.

3.3. Discussion

The risk classification responses on the surveys among the collaborators presented some discrepancy

due to the fact some collaborators have shown more difficulty in answering the surveys than others.

This variance of values resulted from the fact that not all collaborators have the same background

education nor the same work experience. Other important factor was the risk analysis knowledge that

some collaborators possessed, while others did not. This discrepancy of responses in some cases led

to a very high SD, for example, the failure mode “Filling material with defects” the DET parameter present

a SD of 4.2 (Appendix Table A 1). In risk classification a 4 value difference has different meanings which

alter the type of risk. It was decided to do a weighted mode in which some collaborators responses had

more weight than others, depending on their education and work experience (Table 11). The final value

of each parameter, SEV, OCC and DET is the most frequent response of the survey with the respective

given weight. These values were used to calculate the RPN in the FMEA method and to classify the

type of risk using the ISO 14971:2007 qualitative analysis (Table 13). Thought the RPN it was possible

to order the failure modes from which represents a greater risk to the lowest risk by constituting a Pareto

chart. This exercise was necessary in order to decide the limit of RPN from which all failure modes are

considered unacceptable. In Figure 12 it is visible que maximum value of RPN and de minimum, 108

and 9 respectively. Various failure modes presented equal RPN, being the most common the value 32

with seventeen failure modes and the less common 108 with only one failure mode. The first eight at

left bars stand out from the chart in comparison with the other bars with RPN of 108, 84, 81 and 80. This

Page 64: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

51

highlight was the decision maker in defining the limit in which a risk should be considered as

unacceptable, this decision was fundamental to determinate which failure modes should be submitted

to corrective actions. For that reason, it was decided that a risk is considered to be unacceptable at RPN

80 and intolerable at 300 (Table 12). This exercise was not necessary to do to the ISO 14971 method

since it already presents a risk matrix (Table 13).

The results have shown that the FMEA method detected eight unacceptable risks and the ISO 14971

only detected six with two risk in common with the FMEA method. The FMEA method not only detected

more risks, but detected in four different processes (Weighting, Mixing, IPC and FPC and Pre and Sterile

Filtration), while the ISO 14971 method only detected in two processes (Weighting and IPC and FPC).

The RPN and the CRIT values helps understand the difference between unacceptable risks since it

uses numeric values, while the ISO only distinguishes high from low risks. We can argue that the FMEA

is a more sensible analysis than the ISO since it detects more unacceptable risks in more processes

and the differences between them (Table 14 and Table 15).

The next step was to implement corrective actions to all the unacceptable risks detected both by the

FMEA and the ISO method (Table 14 and Table 15). Once again, the aim of the implementation is to

reduce the RPN of the detected risks by mitigate the classification given to the SEV, OCC and DET

parameters. All the corrective actions given to the FMEA method are of a detective nature except one

who also has occurrence nature, meaning all the DET and one OCC parameter suffered a reduction. In

the ISO 14971 method the risks in common with de FMEA method present the same corrective action,

since the corrective action only affects the DET parameter. The risk classification given to the failure

mode trough this method does not suffers alteration, because the ISO only studies the SEV and the

OCC parameters. The rest of the unacceptable risks detected by the ISO are all from the IPC and FPC,

being analytical results. When an analytical result out of specification is obtained and it is not expected

it is classified as an OOS and an investigation must be performed in order to determine the cause of the

result. The final report of the investigation must refer if the anomaly is due to problems during the quality

control analysis, the manufacturing process or from the quality of the raw material. Thus, implement

corrective action in order to reduce the probability of occurrence in future analysis is necessary [110].

The problem with this product is that is new, and it was never manufactured before in Edol’s facilities,

meaning that there are no documents with previous OOS and respective corrective actions since an

investigation was never performed. For that reason, it is very difficult or impossible to implement a

corrective action since the possibilities are infinite. The corrective actions in this case are dependent of

the process in which the failure occurred, examples of possible corrective actions are maintenance of

the equipment used in a specific process and increase the anticipation of their maintenance, adequate

training and sensitization of the operators, validation of the mixing time, reanalysis of the raw material

or reformulation of the product.

The FMEA method is therefore the only method possible to perform a risk review, by implementing the

corrective actions the DET and the OCC value was reduced by one value. It was decided to reduce only

one value since these corrective actions are not extreme actions. By calculating once again the RPN

the once before unacceptable risk are now considered to be acceptable risks. The reduction in all cases

were less than 50% (Table 16) due to the fact that the previous unacceptable risks presented values

Page 65: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

52

close to the minimum limit of the Unacceptable Risk classification (Table 12), therefore it was not

necessary to do a dramatic corrective action in order to the risk become acceptable.

3.4. Conclusion

Risk Analysis is a complex and time consuming process with the necessity to understand in detail every

aspect of the process in question. In the case of the HA 0.15% and 0.30% an intensive study had to be

performed in every process, necessary to the production of the product in order to detect all the possible

risks that may occur. Two tools were chosen to detect and evaluate failure modes existent in the

manufacturing process of HA 0.15% and 0.30%, the FMEA and the ISO 14971:2007 Qualitative

Analysis. From the above evaluation of risk assessment based on FMEA and the ISO 14971:2007

Qualitative Analysis, the majority of the detected risks were considered to be acceptable risks, were the

FMEA method detected approximately 14% unacceptable risks and the ISO 14971:2007 detected 11%.

It was concluded that the FMEA method is more sensitive and precise than the ISO method, detecting

more risks in more processes and with the capacity to distinguish the differences between risks, which

makes it possible to understand the specific seriousness of each risk. Through the FMEA method is was

possible to implement corrective actions and mitigate every unacceptable risk detected, actions that the

ISO was not capable to do. By using two tools in one process it was possible to conclude that different

tools give different answers. Although the ISO 14971:2007 is an application of risk management to MD,

the FMEA demonstrated to be a most suitable risk management tool in this product.

Page 66: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

53

Chapter Four – Pilot Scale Batches Process Validation 4. Pilot Scale Batches Process Validation - Introduction

Validation is the act of demonstrating and documenting that a procedure operates effectively. Process

validation ensures and provides documentary evidence that the concerned processes, within their

specified design parameters/specifications, is capable of consistently producing a finished product of

the required quality. Therefore, it is essential to have a deep understanding of the process in order to

perform a risk analysis and identify the critical steps that may be incurred on the product quality [111].

Pilot batch size may be used in the process development or optimization step, to support formal stability

studies and also to support pre-clinical and clinical evaluation. It should correspond to 10% of production

scale and provides the data predictive of the production scale product. After the results from the

Validation Plan, it may be necessary optimise the manufacturing process. Therefore, the pilot batch is

the link between process development and industrial production of the product. The purpose of the pilot

batch is to analyse and evaluate the difficulties and critical points of the manufacturing process and to

determine the most appropriate large-scale production, providing a high level of insurance that the

product will have the best quality and that the process will be feasible on an industrial scale [111].

The HA 0.15% and HA 0.30% were never before produced in Edol’s facilities being the validation plan

an obligatory requirement confirm if the manufacturing process is suitable or if it needs some

adjustments.

4.1. Materials and Methods

4.1.1. Materials

The materials used are described in Chapter Three – Risk Analysis, section 3.1.1.

4.1.2. Methods

4.1.2.1. Production of three batches of Hyaluronic Acid 0.15% and HA 0.30%

The manufacturing process of 3 batches of 50 L of each product starts with the introduction of highly

purified water in the mixer (DB 110 A FW, Seite-werke, Germany) and the addition of sodium

hyaluronate, stirred at 680 rpm during 120 minutes or until complete dispersion. Afterwards occurs the

addition of potassium chloride, magnesium chloride hexahydrated and calcium chloride hexahydrated

and stirred at the same velocity during 10 minutes or, again, until complete dissolution. Sodium chloride,

boric acid, sodium tetraborate and EDTA are added posteriorly and stirred at the same conditions as

before or until complete dissolution. The final step is the addition of the preservative Suttocide (N-

hydroxymethylglycinate 50%), which is stirred at 680 rpm during 15 minutes or, once again, until

complete homogenization.

4.1.2.2. Appearance

The macroscopic appearance was visually analysed by placing a sample in an appropriate container

and observed at room light.

Page 67: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

54

4.1.2.3. Determination of the pH values

The materials used are described in Chapter Two – Previous Work, section 2.1.2.2.

4.1.2.4. Determination of the Osmolality values

The materials used are described in Chapter Two – Previous Work, section 2.1.2.3.

4.1.2.5. Determination of the Viscosity values

The materials used are described in Chapter Two – Previous Work, section 2.1.2.4.

4.1.2.6. Determination of the Density values

The pycnometer was tarred with the cap and afterwards filled with the sample. The excess was removed

when the cap was introduced. Then the pycnometer was weighted, and the value recorded, the value

of the density corresponds to the radio between the sample weight and the pycnometer calibration value.

The density value obtain is only informative and the test is made at room temperature.

4.1.2.7. Bioburden

The bioburden test was performed in a laminar-air-flux cabinet (The Baker Company SG503A-HE,

USA), using a filtration system (Milifex Plus, Merck, Germany). The cups utilized were a ready-to-use

sterilized filtration devices for microbial enumeration with a 0.45 µm PVDF membrane (EZ-Fit™ Filtration

Unit, Merck, Germany). A volume of 100 mL of sample was transferred into the cup and filtered

immediately, rinsing the membrane filter with 600 mL of fluid D (Merck, Germany). The membrane was

transferred to the surface of typticase soy agar (TSA, Merck, Germany) and incubated at 20-25°C for 5

days. After the first incubation, the plates were transferred to 30-35ºC and incubated for 2 days. The

test was performed in duplicate and with a negative control.

4.1.2.8. Sodium Hyaluronate Assay

The Sodium Hyaluronate Assay was performed using cetyltrimethylammonium bromide (CTAB)

turbidimetric method in the ultraviolet-visible spectrophotometer (Evolution 201 ÛV-Visible

Spectrophotometer, Thermo Fisher Scientific, USA). A volume of 2 mL of Acetate buffer pH 6.0 and 2

mL of standard solution or sample solution were introduced in tubes properly stoppered. The tubes were

stirred and incubated at 37°C in an incubator (Vaciotem-t, JP Selecta, Spain) in order to synchronize

the reaction temperature. CTBA was added next to each tube and incubated during 6 minutes. The

absorbance of the sample and the standard solution were read in the ultraviolet-visible

spectrophotometer at the absorption maximum of 600 nm, using the blank solution prepared previously.

4.1.2.9. Sterility Test

The sterility test was performed according to the European Pharmacopeia, Chapter 2.6.1. by Membrane

Filtration [112]. Briefly, the sterility test was performed in a laminar-air-flux cabinet (ADS Laminaire

OPTMALE 12, France) using a Steritest™ Symbio Pumps with Steritest™ EZ Canister Device for

Antibiotics (Merck, Germany), because the product is very viscous which difficult the filtration process.

This method is closed filtration method. It starts with prewetting the membranes with the rinse fluid A

Page 68: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

55

(Merck, Germany). Next, after the decontamination of the bottles with isopropyl alcohol 70% v/v, the

Steritest needle was inserted into the first product container allowing the product to be transferred to the

canisters, this process was repeated for the rest of the product containers. After the filtration of the

product, the membrane was rinsed with fluid A. Afterwards, one of the canisters was filled with

thioglycollate media (Merck, Germany) and the other with typticase soy broth (TSB, Merck, Germany).

The canister with TSB media was incubated at 20-25°C for 14 days and the canister with thioglycollate

media at 30-35 for 14 days.

4.2. Critical Steps to Control

In the scope of the manufacturing process validation, the steps and the process controls defined as

essential for the evaluation and obtainment of a finished product with the required quality, are defined

on Table 17.

Table 17 - Steps, Process Controls and Acceptance Criteria considered for the validation plan

of HA 0.15% and HA 0.30% eye drops solution, 8 mL.

Step Process control Acceptance Criteria

Mixture Velocity and time of the mixtures

Stir until completely dissolved

In Process Control Holding time validation: 0h3

Appearance The solution must be limpid, transparent and odourless.

pH Between 7.0 and 7.6 (temperature 20-25ºC)

Osmolality Between 280 and 320 mOsm/Kg

Viscosity In study1

Sodium Hyaluronate assay Between 90% and 110% (UV-Vis)

Density Not Apply2

Bioburden < 10 CFU/10mL

Sample collected before the sterilizing filtration Holding time validation: 48h3

Appearance The solution must be limpid, transparent and odourless.

pH Between 7.0 and 7.6 (temperature 20-25ºC)

Osmolality Between 280 and 320 mOsm/Kg

Viscosity In study1

Sodium Hyaluronate assay Between 90% and 110% (HPLC)

Bioburden < 10 CFU/10mL

Sterilizing Filtration Bubble Point In study1

Filling Average weight/volume (filling quantity)

8.0 mL – 8.5 mL

Leak test Bonfiglioli PKV 212 On line leak test Conform

Control of packaging operation

Visual control Conform

Finished Product Control (analytical demand for the product release)

Appearance The solution must be limpid, transparent and odourless.

pH Between 7.0 and 7.6 (temperature 20-25ºC)

Osmolality Between 280 and 320 mOsm/Kg

Viscosity In study1

Sodium Hyaluronate assay Between 90% and 110% (UV-Vis)

Filling Volume 8.0 mL – 8.5 mL

Sterility test Absence of growth 1 These specifications will be determined during the manufacture of the pilot batches. 2 Extra analysis for the determination of the filling average. 3 These values will be determined only for the batches used in the process validation.

Page 69: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

56

4.2.1. Acceptance Criteria

These tests aim to control the procedures considered key to the quality of the finished product, allowing

to evaluate the influence of each production step on the finished product. The results of the tests

performed in the scope of the process control should be in accordance with the acceptance criteria,

previously defined for each parameter, in each one of the manufacturing steps and should reflect the

reproducibility of the process.

In order to consider the manufacturing procedure validated, the results must be in accordance with the

defined acceptance criteria in the tests performed, in 3 pilot batches.

4.2.2. Validation Plan

The validation involves the evaluation of certain characteristics of the product in manufacturing steps

considered as potential influents. Therefore, process control test is needed to ensure the conform

specifications of the final product.

4.2.2.1. Preparation of HA 0.15% and HA 0.30% Eye Drops Solution

During the mixing process, the velocity and the mixing time of the mixer at each mixture step are

recorded. The results are evaluated and the influence on the manufacturing process and on the finished

product are defined.

4.2.2.2. In Process Control and Holding Time Validation of HA 0.15% and HA 0.30%

eye drops

After the complete homogenization of the solution in the Mixing Process, two samples of 200 mL for

microbiological control and 25 mL of physicochemical control are collected from the top and the bottom

of the mixer. The obtained results must be within the defined acceptance criteria.

Holding time study can demonstrate how much time is suitable for hold the blend or bulk stage before

processing to the next stage [113]. The defined time is 48h after the manufactured process, reflecting

the worst case scenario of waiting time between the end of the preparation and the start of the sterilizing

filtration. In this test also two sample are collected from the top and the bottom of the mixer, 200 mL for

microbiological control and also 200 mL for the physicochemical control.

These two tests have the same Process Control and the same Acceptance Criteria, represented in Table

17.

4.2.2.3. Control of the Sterilizing Filtration

The bubble point test is determined before and after the sterilizing filtration of the solution, in order to

control the suitability of sterilizing process defined for the manufacturing process. The test is made with

5L of the product and with purified water and it is made 3 times during the process. The filter must

remain intact, and the minimum pressure to rupture the membrane is 50 psi.

4.2.2.4. Filling Control

The Filling Process is controlled by determining the average weight with the determination of the density

of each batch and it must be between 8.0 mL and 8.5 mL.

Page 70: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

57

4.2.2.5. Finished Product Control

To evaluate the influence of the manufacturing procedures on the specifications of the finished product,

on the repeatability of the filtration step and on the consistency of the analytical results for the product

release a control of the finished product is made. After the sterilizing filtration and the filling/packaging

process 61 samples of finished product will be collected: 10 from the beginning, 10 from the middle and

11 from the end of the filling process, also a pack of 30 units for placement in the sample room. For a

physicochemical analysis, 15 units were collected (5 units from the beginning, 5 from middle and 5 from

end of the filling) and for the microbiologic analysis, an average sample of 10 units (3 units from de

beginning, 3 units from the middle and 4 units from the end of the filling) for sterility testing. The Process

Control and the Acceptance Criteria are once again represented in Table 17.

Page 71: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

58

4.3. Results

The results of the Validation Plan obtained in the IPC and FPC are presented in Table 18, Table 19 and Figure 13 for both the HA 0.15% and HA 0.30%. Three

batches of each product were produced to perform this study and the results in Table 18 correspond to the mean of the top and bottom samples.

The velocity and mixing time were recorded in all three batches from both products and it was concluded they were suitable for the manufacturing process.

Table 18- Results obtained in the IPC and FPC analysis with respective specifications of HA 0.15% and HA 0.30%.

HA 0.15 HA 0.30

Specification PB1 0.15 PB2 0.15 PB3 0.15 PB1 0.30 PB2 0.30 PB3 0.30

IPC

Aspect Limpid,

transparent and odourless

Conformed Conformed Conformed Conformed Conformed Conformed

pH (20°C - 25°C) 7.0 - 7.6 7.16 (23.2 °C ) 7.17 (24.2 °C ) 7.17 (24.2 °C ) 7.32 (20.8 °C ) 7.32 (20.9 °C ) 7.17 (24.2 °C )

Osmolality 280 - 320 mOsm/Kg

308 mOsm/Kg 302 mOsm/Kg 302 mOsm/Kg 304 mOsm/Kg 296 mOsm/Kg 297 mOsm/Kg

Viscosity (20°C - 25°C) Under study 59.4 cP (21.3 °C )

12 rpm 67.4 cP (21.4 °C )

12 rpm 42.7 cP ( 24.5°C )

12 rpm 504 cP (23.6 °C )

0.6 rpm 601.5 cP (22.2 °C )

0.6 rpm 646 cP (24.8 °C )

0.6 rpm

Sodium Hyaluronate Assay

90% - 110% 104.14% 108.45% 103.23% 100.77% 106.49% 105.90%

Bioburden < 10 CFU/10 mL 0 UFC/100 mL 0 UFC/100 mL 0 UFC/100 mL 0 UFC/100 mL 0 UFC/100 mL 0 UFC/100 mL

Density (20°C - 25°C) Not Apply 1.0049 g/mL 1.0051 g/mL 1.0045 g/mL 1.0057 g/mL 1.0059 g/mL 1.0074 g/mL

FPC

Aspect Limpid,

transparent and odourless

Conformed Conformed Conformed Conformed Conformed Conformed

pH(20°C - 25°C) 7.0 - 7.6 7.19 (24.3 °C ) 7.19 (24.3 °C ) 7.21 (23.9 °C ) 7.41 (24.2 °C ) 7.42 (2 4.2 °C ) 7.42 (24.3 °C )

Osmolality 280 - 320 mOsm/Kg

305 mOsm/Kg 298 mOsm/Kg 305 mOsm/Kg 299 mOsm/Kg 301 mOsm/Kg 296 mOsm/Kg

Viscosity (20°C - 25°C) Under study 31100 cP (21.3

°C ) 12 rpm 37933 cP (22.2 °C )

12 rpm 36483 cP (22.1 °C )

12 rpm 655 cP (22.1 °C )

0.6 rpm 537 cP (22.6 °C )

0.6 rpm 520 cP (22.6 °C )

0.6 rpm

Sodium Hyaluronate Assay

90% - 110% 101.35% 100.83% 101.03% 97.32% 99.27% 98.57%

Sterility Test Absence of

growth Absence of

growth Absence of growth Absence of growth * * *

*Results non-existent.

Page 72: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

59

Table 19- Results obtained in the Bubble Point and Bonfiglioli test for HA 0.15% and HA 0.30%.

Batch number

Bubble Point HA 0.15%

Bubble Point HA 0.30%

Bonfiglioli HA 0.15% Bonfiglioli HA 0.30%

Before filtration process

(Psi)

After filtration process

(Psi)

Before filtration process

(Psi)

After filtration process

(Psi)

Tested Rejected Rejected

(%) Tested Rejected

Rejected (%)

PB1 56.60 56.90 56.10 56.20 4789 7 0.15 3033 8 0.26

PB 2 57.50 58.60 58.90 56.20 5035 5 0.10 4600 16 0.35

PB 3 55.30 55.30 56.70 58.40 4868 10 0.21 4362 10 0.23

Mean 56.47 56.93 57.23 56.93 4897 7 0.15 3998 11 0.28

SD 1.11 1.65 1.47 1.27 126 3 0.05 844 4 0.06

4.4. Discussion

The aim of the present study was to validate the effectiveness of the manufacturing process by analysing

the physical-chemical and microbiological aspects of the finished product. Three pilot batches of the two

products, HA 0.15% and HA 0.30% eye drops solution, were produce in order to verify if their

characteristics were persistent among each other and between their IPC and FPC, to see if the results

respected the required specifications of each analysis. Overall the results descendent from the process

controls in each pilot batch have respected the required specifications, the aspect always presented a

limpid, transparent and odourless appearance, the pH, the osmolality, the sodium hyaluronate assay,

the bioburden and the sterility test, this last aspect only applied to the HA 0.15%, all presented values

within the limit of the required specification. The density assay which was only performed for the

determination of the filling average presented identical values in all the manufactured batches.

7,9

8,0

8,1

8,2

8,3

8,4

8,5

8,6

Avera

ge s

am

ple

s v

olu

me (

mL)

Hours in Process Control

Filling Control HA 0.15% and HA 0.30%

Upper specification limit Lower specification limit

Average samples volume HA 0.15% PB1 (mL) Average samples volume PB2 HA 0.15% (mL)

Average samples volume HA 0.15% PB3 (mL) Average samples volume HA 0.30% PB1 (mL)

Average samples volume HA 0.15% PB2 (mL) Average samples volume HA 0.15% PB3 (mL)

Figure 13- Results from the filling control test for HA 0.15% and HA 0.30%.

Page 73: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

60

Between each batch of the two products it is visible that the values of the IPC and the FPC kept similar

results for the process controls with a defined specification. The viscosity component, which is in study,

showed similar values between the three batches both in the IPC and the FPC. However, the values of

the IPC and the FPC for each batch in HA 0.15% increased significantly while in the HA 0.30% only the

PB1 presented a slight increase while the other batches diminished lightly. These unexpected values

between the IPC and the FPC of each batch are maybe due to the fact that the polymer in question is a

dry polymer and one of the most important steps in the preparation of dry polymer is polymer wetting.

The moment the polymer and the dilution water first contact it is crucial to disperse the polymer into the

water effectively wetting each individual polymer particle. In this step a high shear mixing energy is

required to avoid polymer agglomeration. The rate of hydration is dependent of the particle size and

wetting time, in other words, the smaller the polymer particle the faster it hydrates. This parameter can

be measure through viscosity, higher polymer hydration increases solution viscosity [114]. The viscosity

of the three batches of HA 0.15% had an increasement of approximately 500 times between the IPC

and the FPC. It is important to mention that the time that the IPC was performed was in July while the

FPC was in September, which means that the product was at rest during two months. The HA is a dry

powder polymer that takes some time to hydrate and for a period of two months the product had a large

amount of time to hydrate each particle individually, resulting in a viscosity increasement. This

phenomenon may have also affect the osmolality of the product. These results were not visible in the

HA 0.30% since the time between the IPC and the FCP was not so extensive. The IPC of PB1 0.30 was

performed in August and the FPC was in September, while the IPC and the FPC of both PB2 0.30 and

PB3 0.30 were performed in September. The PB1 0.30 was at rest for one month which means the

polymer had some time to hydrate, being the reasons of the slight increase. However the PB2 0.30 and

PB3 0.30 only rested for some days, not having enough time to hydrate properly and the polymer was

still in a readjustment stage, which causes the slight decrease of viscosity. Although the PB1 0.30

presented a slight increased and the PB2 0.30 and PB3 0.30 presented a slight decrease these changes

were not significant.

Regarding the sterility test, being the product an eye solution, it is demand that the product is sterile to

guarantee a safe use. The HA 0.15% three batches all contained absence of growth meaning that the

product was not contaminated. The HA 0.30% however the sterility test was not performed.

The Bubble Point and Bonfiglioli test results (Table 19) show the filter integrity records for the filters used

(before and after filtration) were in accordance with the required parameters as well as the leak tests

(Bonfiglioli). The filling control (Figure 13) shows all three batches (HA 0.15% and HA 0.30%) are within

the limited specifications

4.5. Conclusion

The results of the Validation Plan obtained in the IPC and the FPC have showed to be consistent and

respectful of the limit established for each process control, indicating homogeneity between batches, as

well as a good reproducibility of the manufacturing process. It is not possible to conclude if the

manufacturing process can be validated or if it is necessary to optimise the manufacturing process since

the sterility test of HA 0.30% is still yet to be performed.

Page 74: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

61

Chapter Five – Product Characterisation 5. Product Characterization - Introduction

In order to develop a proper MD it is essential to verify if it is safe, effective and easy to use. To achieve

this goal usability testes are performed to scrutinize device design for potential safety issues and validate

device user interface such that errors that could occur during the use of the device are either eliminated

or reduced as far as possible [69].

When it comes to eye drops, its characteristics may determine the effectiveness and compliance of the

therapy. The solution has to assure comfort, convenience and absence of adverse effects to guarantee

treatment success [115]. Polymers are used in eye drops to regulate the surface tension and wettability

which promotes the hydration of the cornea surface [116], therefore it is important to study the strength

of the interaction between the polymer and the mucin, which is one of the layers that constitutes the tear

film.

Nowadays to study the efficacy of a product in vitro tests are quite used, for instance, though rheological

methods such as tensile strength measurements, flow and oscillation methods providing information

concerning the concentration of the polymer, the mucin and physical properties aspects of the product

[46,52]. To evaluate the products’ safety, in vitro tests cell viability assays using specific cell lines are

widely used to study cell behaviour after product administration.

5.1. Material and Methods

5.1.1. Material

The materials used are described in Chapter Three – Risk Analysis, section 3.1.1. In addition, it was

use dried mucin from porcine stomach type II (Sigma-Aldrich, USA). Human retinal pigment epithelial

cell lines ARPE-19 (ATCC® CRL-2302™) were obtained from American Type Cell Culture collection

(USA), and they were used for cell viability and dry eye assays. Cell culture medium and supplements

were from Gibco (Thermo Fisher Scientific, UK).

5.1.2. Methods

5.1.2.1. Viscosity Measurements

5.1.2.1.1. Brookfield viscometer

Shear rate against shear stress measurements were obtained using a DV-II + Pro Brookfield (Brookfield,

USA) viscometer equipped with spindle nº 21 at room temperature (20 - 25ºC). A shear rate sweep from

0.5 to 100/s and up and down for 7.5 min was assessed.

5.1.2.2. Mucoadhesion studies

The mucoadhesion was evaluated by viscosity, rheology and zeta potential (ZP) measurements. The

mucin used in this study was hydrated with water by gentle stirring until complete dissolution to yield a

dispersion of 10% (w/w) at 20-25°C.

Page 75: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

62

5.1.2.2.1. Ostwald viscometer

The viscosity properties of the solutions were determined at room temperature by using the Ostwald

viscometer (Fisher Sientific, USA) using the following equation:

ƞ1 = ƞ2. ρ1t1/ρ2t2 (5) Where ƞ1 and ƞ2 are viscosity coefficients of the solution and water, ρ1 and ρ2 are the densities of the

solution and water, and t1 and t2 are the flow times measured in the viscometer of the solution and water,

respectively.

The viscosities of each individual component, HA 0.15%, HA 0.30% and mucin, were measure first in

triplicate, a mean of the values of each component was made. To evaluate the effect of the interaction

of the mucin with the solutions three samples were prepared: 1) 5 % w/w mucin suspension diluted in

water; 2) Mucin and HA 0.15% solution (1:1); 3) Mucin and HA 0.30% solution (1:1).

The mucoadhesion was expressed through the following equation:

Δ(%) = [ƞmuc+HA – (ƞmuc + ƞHA)] / (ƞmuc + ƞHA) x 100 (6)

Where Δ(%) is the mucoadhesion index, ƞmuc, ƞHA and ƞmuc+HA is the mucin’s, the product’s and the

solution containing mucin and product dynamic viscosity, respectively. For a mucoadhesive polymer,

which is the case of HA, the ƞmuc+Ha is higher than (ƞmuc + ƞHA) due to the interactions occurring between

the polymer and mucin. The mucoadhesive index is a measure of the mucoadhesive strength [56].

5.1.2.2.2. Rotational Rheometer

The rheological characteristics of the formulations were examined at high shear rates using continuous

shear techniques and in the viscoelastic region using oscillation techniques. These experiments were

performed with a controlled stress Malvern Kinexus Rheometer (Malvern Instruments, Malvern, UK)

using cone and plate geometry (truncated cone angle 4° and radius 40mm). The frequency sweep

method was performed between 0.1Hz and 10Hz, with a shear strain of 0.8%, at 25ᵒC, while the table

of shear rate method was performed by increasing the shear rate from 0.1s-1 to 100s-1, at 25ᵒC. The

shear stress was measured by this method and the apparent viscosity was calculated by dividing the

shear stress by the shear rate.

An oscillatory amplitude sweep and frequency testing was performed using this equipment. The

amplitude sweep conditions used were shear strain between 0.01% and 100% with the frequency of 1

Hz. It was concluded that the LVER (linear-viscoelastic region) was at shear strain of 0.25%. In the

frequency testing the frequency range used was between 0.1 - 10 Hz with a shear strain of 0.25%.

A time sweep test was also performed using this equipment with a shear strain of 0.25% and a frequency

of 1Hz during 30 minutes at room temperature (20 - 25ºC).

The adhesive strength was also measured using the same equipment and a plate and plate geometry.

It was used a toolkit with the conditions of 0.1 mm/s, 5 mm and 0.15 GAP. The same protocol was

performed using pig eyes obtained from a local slaughterhouse, instead of mucin.

Page 76: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

63

5.1.2.2.3. Zeta Potential

The mucoadhesion interaction was determined by measuring the ZP of the mixtures of mucin and each

solution using a Zetasizer Nanoseries Nano Z (Malvern Instruments, Malvern, UK). A volume of 40 µL

of all samples were diluted in 2 mL of filtered purified water and the cell was filled verifying for the

existence of bubbles that could cause interference in the ZP measurements. All experiments were done

in triplicate.

5.1.2.3. In Vitro Assay

5.1.2.3.1. Cell Culture Condition

The ARPE-19 cell line (ATCC, CRL-2302™) was grown in DMEM/F12 culture medium (Gibco, UK)

supplemented with 10 % (w/v) fetal bovine serum (FBS, Life Technologies. Inc., UK), penicillin (100

IU/mL) and streptomycin (100 μg/mL) in a humidified 95 % O2, 5 % CO2 environment at 37°C.

5.1.2.3.2. Cell Viability of HA 0.15% and HA 0.30%

The cell viability was quantitatively evaluated in vitro using general cell viability endpoint MTT reduction

(3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl- 2H-tetrazolium bromide) assay. MTT is a yellow and water-

soluble tetrazolium dye that is converted by viable cells to a water-insoluble, purple formazan.

Cell viability was assessed after 24h of incubation of ARPE-19 cell line with different concentrations of

each sample. The negative control was the culture medium and positive control sodium dodecyl sulfate

(SDS) at 0.1 mg/mL. After the time of exposition (24 h), the culture medium was replaced by medium

containing 0.5 mg/mL MTT. The cells were further incubated for 3h. In the plates containing reduced

MTT, the media was removed, and the intracellular formazan crystals were solubilized and extracted

with dimethylsulfoxide (DMSO). After 15 min at room temperature the absorbance was measured at 570

nm in the same microplate reader.

The relative cell viability (%) compared to control cells was calculated by the following equations:

𝐶𝑒𝑙𝑙 𝑉𝑖𝑎𝑏𝑖𝑙𝑖𝑡𝑦 (%) 𝑓𝑜𝑟 𝑡ℎ𝑒 𝑀𝑇𝑇 𝑎𝑠𝑠𝑎𝑦 = [𝐴𝑏𝑠𝑜𝑟𝑣𝑎𝑛𝑐𝑒 570 𝑛𝑚]𝑠𝑎𝑚𝑝𝑙𝑒

[𝐴𝑏𝑠𝑜𝑟𝑏𝑎𝑛𝑐𝑒 570 𝑛𝑚]𝑐𝑜𝑛𝑡𝑟𝑜𝑙 × 100 (7)

5.1.2.3.3. 2D model - Evaluation of Cell Morphology and Cell Viability After

Dehydration

The protective effect of the selected formulas against dehydration was evaluated using previously

reported protocols, with modifications [56]. Specifically, cells were seeded in 24-multiwellplates (5 x

104/well) and in DMEM/F12 until 70% confluence was reached. The medium was then replaced with

selected HA formulations (HA 0.15%, HA 0.30% and CR 0.30% wt% solutions prepared in cell culture

medium) and with the same solutions diluted 1:5. For the positive and negative controls, the medium

was replaced with fresh medium not containing HA. Cells were incubated under cell culture conditions

for 2 h. Cells treated with the HA samples and not treated (negative control, NC) were then dehydrated:

the medium was removed and the multiwells were incubated at 37°C without the lid until a stress

response (morphological change) was evident in the NC (about 20 min). The positive control (PC, not

Page 77: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

64

treated with HA), was not dehydrated (cells were kept in the presence of the medium during all

experiments.

Cell viability was evaluated using the AlamarBlue assay (Cat. N. A13261, Invitrogen, GIBCO) according

to manufacturer’s instructions. When added to cells, the cell-permeable AlamarBlue reagent, resazurin,

is reduced by the dehydrogenases into resarufin by viable cells. The conversion is proportional to

metabolically active cells and was quantitatively determined by fluorescence measurements. Cell

viability (%) was calculated with respect to the positive control (100% viability). Results were reported

as means ± SD.

5.1.2.3.4. 3D model - Dry Eye Model and Cell Viability

For the 3D dry eye assay, cells were cultured on filters following the protocol described by Dunn et al.

with some modifications [117]. Briefly, the cells were seeded at a density of 1×105 cells/cm2 on

ThinCert™ cell culture inserts (Greiner, 3 μm, 12 wells, UK). The culture medium was supplemented

with l-ascorbic acid (50 µg/mL), β-glycerolphosphate (10 mM) and dexamethasone (10 nM) in order to

enhance the barrier properties and facilitate expression of RPE-specific genes [117]. Fresh medium with

supplements was changed twice a week.

The progress of epithelial barrier formation and polarization was followed by measuring transepithelial

electrical resistance (TEER) with a Millicell-ERS device (Millipore, Germany) and chopstick-style

electrode. The combined resistance of the filter was subtracted from the values of filter-cultured ARPE-

19 cells in order to calculate the resistance of the cell layer. The plateau in TER was reached in two

weeks and, thereafter, it remained essentially unchanged. The cells were used for experiments after

culturing them for three weeks. In the permeability experiments the resistance was determined before

and after the experiments.

After the three weeks ARPE-19 cells were placed under controlled environmental conditions to mimic

dryness (without lid, <40% relative humidity, 37 °C ± 5 °C temperature and 5% CO2). Cells were

investigated for cell viability at 48 h after establishment of dry eye conditions, using the MTT reduction

assay (see section 5.1.2.3.2). Cell viability was assessed after 24h of incubation of ARPE-19 cell line

with 20 mg/mL concentration of each sample. The negative control was the culture medium and positive

control sodium dodecyl sulfate (SDS) at 0.1 mg/mL.

5.1.2.4. Statistical Data Analysis

The data was expressed as mean and standard deviation (mean ± SD) of experiments. Tukey–Kramer

multiple comparison test (GraphPad PRISM 5 32 software, USA), was used to compare the significance

of the difference between the groups, a p <0.05 was accepted as significant.

5.2. Results

5.2.1. Viscosity Measurements of HA 0.15% and HA 0.30%

The viscosity of the three pilot batches of each product, HA 0.15% and HA 0.30%, were measured. The

results are represented in Figure 14 which shows the representative flow curves (shear stress function

of shear rate).

Page 78: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

65

Figure 14 – Typical flow curve of shear stress as function of shear rate for HA 0.15% and HA

0.30% eye drops solution.

All batches of each product present the same rheological profile demonstrating that the relation between

shear stress and shear rate is not constant, which shows that the apparent viscosity increases with the

increase of the shear rate in a non-linear form (Figure 14). This profile shows that both products are

shear thinning fluids. It is evident that the shear stress values of HA.030% are much higher than the

shear stress values of HA 0.15% at the same rate. At the highest shear rate (122.36 sec-1) the apparent

viscosity of HA 0.30% was 83.9 mPa.s and 22.5 mPa.s for HA 0.15%.

5.2.2. Mucoadhesive Studies

5.2.2.1. Viscosity Measurements

A study using an Ostwald viscometer was also performed to evaluate the viscosity of the products and

to study the mucoadhesion, in other words, the interaction of HA 0.15% and HA.30% in solution in the

presence of mucin. By measuring the viscosity it is possible to evaluate the interaction of the product

with the mucin, because a higher interaction with the mucin is related with higher viscosity.

0

2000

4000

6000

8000

10000

12000

0 20 40 60 80 100 120 140

Sh

ear

str

ess (

Pa)

Shear rate (sec-1)

HA0.30% Batch 1

HA0.30% Batch 2

HA0.30% Batch 3

HA0.15% Batch 1

HA0.15% Batch 2

HA0.15% Batch 3

Page 79: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

66

Figure 15- Viscosity determination for HA 0.15% and HA 0.30% in absence and presence of Mucin

5% (w/w) (mean SD, n=3).

The mucoadhesive index was calculated for the HA 0.15 + Mucin and HA 0.30% + Mucin solutions in

order to evaluate the mucoadhesive strength gain due to the interactions between the polymer and the

mucin.

The results obtained with an Ostwald viscometer presented in Figure 15 supports the results obtained

in the Brookfield viscometer, HA 0.30% presents a much higher viscosity than HA 0.15%. An increase

of viscosity of the product in the presence of mucin when compared with their individual viscosity is also

observed. This increase is more evident for the HA 0.30%, with a Mucoadhesive index of 298.07% ±

6.24% in viscosity (Equation 6) than for HA 0.15% with 67.44% ± 19.90%.

5.2.2.2. Rheology Measurements

5.2.2.2.1. Tackiness Testing

Tackiness in the context of material behaviour is associated with stickiness and may result from

adhesive forces between two materials in contact. It is measured at the maximum force needed to break

the resultant bond. In this test the peak force which is a negative normal force can be attributed to tack

and the area under the force-time curve represents the adhesive strength [118]. These parameters are

measured and the results of the prepared solutions are represented in Table 20. Two samples used are

commercial reference (CR) of HA eye drops solution, with different concentrations of HA (CR 0.15%

and CR 0.30%) available in the market.

0

50

100

150

200

250

300

350

400

450

Mucin HA 0.15% HA 0.30% HA 0.15% +Mucin

HA 0.30% +Mucin

Vis

co

sit

y (

mP

a.s

)

Page 80: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

67

Table 20- Normal force and area under force time curve results for HA 0.15% and HA 0.30% and

their interactions with Mucin and with Pig Eye.

*1 Mean ± SD, n=6 *2 Mean ± SD, three different eyes, n=3

The results of the tack testing on the seven samples show that the Mucin + HA 0.30% appears to be

the tackiest of the seven samples analysed with a peak normal force of -0.287 N, followed by the HA

0.30% (-0.287 N), Mucin (-0.228N), CR 0.30% (-0.220 N), Mucin + HA 0.15% (-0.216 N), HA 0.15% (-

0.178 N) and CR 0.15% (-0.168 N). For the Area under force time curve the results did not show the

same profile, the CR 0.30% appears to be the strongest of all samples (1.051 N.s) and the HA 0.15%

the weakest (0.438 N.s).

A similar study was performed but instead of mucin was used pig’s eye (Figure 16), three samples were

used and attached to the probe and the adhesive force between the eye and samples of HA 0.15% and

HA 0.30% was measured. The results show there are significant differences between HA 0.15% and

HA 0.30%, were the HA 0.30% appears to be tackiest with -0.134 N and the HA 0.15% with -0.078 N,

respectively. The Area under force time curve also shows the same profile were HA 0.30% appears to

be the strongest with 1.0110 N.s and the HA 0.15% with 0.891 N.s.

Figure 16- Three samples of pig eye used in the frequency sweep assay (A) and one of the

samples attached to the probe (B).

Peak normal force - Normal Force (N)

Area under force time curve (N.s)

HA 0.15%*1 -0.178 ± 0.003 0.438 ± 0.058

HA 0.30%*1 -0.229 ± 0.013 0.775 ± 0.091

CR 0.15%*1 -0.168 ± 0.017 0.904 ± 0.069

CR 0.30%*1 -0.220 ± 0.007 1.051 ± 0.043

Mucin*1 -0.228 ± 0.004 1.012 ± 0.065

Mucin + HA 0.15%*1 -0.216 ± 0.019 0.573 ± 0.152

Mucin + HA 0.30%*1 -0.287 ± 0.030 0.747 ± 0.066

Pig Eye + HA 0.15%*2 -0.078 ± 0.029 0.891 ± 0.060

Pig Eye + HA 0.30% *2 -0.134 ± 0.034 1.010 ± 0.059

Page 81: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

68

5.2.2.2.2. Oscillation Frequency Sweep

Before the oscillation frequency sweep, an amplitude sweep test was performed to define the fluid’s

linear viscoelastic region (LVER), and the results showed that this region was at 0.25% shear strain.

With this results the product’s structure can be further characterized using a frequency sweep proving

more information about the effect of colloidal forces, interactions among particles or droplets [119].

Figure 17- Frequency sweep with shear moduli as function of frequency of HA 0.15%, Mucin and

Mucin + HA 0.15% at room temperature.

Both Figure 17 and Figure 18 represent the frequency behaviour of the product, HA 0.15 % and HA

0.30% respectively, compared to system obtained with mucin. It is evident that with the mucin the elastic

modulus G’ and the viscoelastic modulus G’’ increased in both products. At lower frequencies both

products exhibited fluid-like mechanism spectra with G’’ modulus greater than G’, being both frequency

dependent. As the frequency increases occurs a cross-over at approximately 2 - 5 Hz, turning the G’

modules greater than the G’ indicating both products started to have a more elastic behaviour.

0,1

1

10

0,1 1 10

G' an

d G

'' (

Pa)

f(Hz)

G´HA0.15%

G´´HA0.15%

G´Mucin

G´´Mucin

G´Mucin+HA0.15%

G´´Mucin+HA0.15%

Page 82: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

69

Figure 18- Frequency sweep with shear moduli as function of frequency of HA 0.30%, Mucin and

Mucin + HA 0.30% at room temperature.

5.2.2.2.3. Time Sweep

The test was performed to understand the changes in the sample with time at constant temperature,

stress and frequency with a time range of 30 minutes. The results show that all sample except mucin

are time independent with viscoelastic modulus (G’’) greater than the elastic modulus (G’), meaning that

their structure do not suffer changes overtime (Figure 19).

0,1

1

10

0,1 1 10

G' an

d G

'' (

Pa)

f(Hz)

G´HA0.30%

G´´HA0.30%

G´Mucin

G´´Mucin

G´Mucin+HA0.30%

G´´Mucin+HA0.30%

Figure 19- Time Sweep Test for HA 0.15% and HA 0.30% with and without mucin.

0,1

1

10

100 1000

G' an

d G

'' (

Pa)

Time (s)

G'Mucin + HA0.30% G´´Mucin + HA0.30% G'Mucin + HA0.15% G´´Mucin + HA 0.15%

G'Mucin G´´Mucin G'HA0.30% G´´HA0.30%

G'HA0.15% G´´HA0.15%

Page 83: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

70

5.2.2.2.4. Zeta Potential

This study demonstrated that the ZP values of HA 0.15% and HA 0.30% eye drops solution are similar

to their market equivalent, CR 0.15% and CR 0.30% respectively. However, comparing the values of

the two products their values are quite different being HA 0.15% ZP values much more negative that

HA 0.30% values, which in absolute is higher (Figure 20). This negative values are in accordance to the

fact the HA presents an anionic nature due to the presence of carboxylic groups. The mucin also

presents negative charge due to the oligosaccharide chains, which confer negative charge to the mucins

through carboxyl and sulfate groups. When the mucin is added to both products an increase of the

negative charge is observed, being the ZP value more negative in Mucin + HA 0.30% than with HA

0.15%.

An overtime study was made to investigate if the interactions alter overtime or if they maintain stable.

The measurements of the samples were performed at 0, 5 10, 15 and 20 minutes and it was concluded

that the values do not suffer significant alteration overtime.

Figure 20- Determination of ZP for HA 0.15%, HA 0.30%, Mucin and both products with mucin

(Mean ± SD, n=3).

5.2.2.3. In Vitro Assay

5.2.2.3.1. Cell Viability of HA 0.15% and HA 0.30%

An initial test to evaluate the potential irritant of the HA formulations was performed in order to choose

the most suitable dilution to be used on the next assays. Four dilutions were prepared 100 µg/mL (1:1),

-40

-35

-30

-25

-20

-15

-10

-5

0

ZP

(m

V)

Page 84: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

71

50 µg/mL (1:2), 33 µg/mL (1:3) and 20 µg/mL (1:5). In Figure 21 it is evident that the 1:5 dilution presents

a high rate of survival in all samples with cell viability above 80%. The 1:3 dilution although the CR

samples demonstrated high survival rate (approximately 100%), the HA samples did not show the same

results. For that reason it was decided to use the 1:5 dilution.

Figure 21- Results of cell viability on ARPE-19 cell lines testing CR 0.30%, CR 0.15%, HA 0.30%

and HA 0.15% at various concentrations (mean ± SD, n=8).

5.2.2.3.2. 2D model - Evaluation of Cell Morphology and Cell Viability After

Dehydration

In Table 21 it is shown optical microscope images of ARPE-19 cells stained with Crystal Violet and

exposed to desiccation under no protective conditions (Dry Eye, negative control), after being treated

with HA 0.15%, HA 0.30% and CR 0.30% and of cells that were not exposed to dehydration (Medium,

positive control). The respective cell viability determination of the samples is also present. In the Dry

eye images it is evident the cells exhibited a disintegrated and dry membrane morphology and an

increase of cell mortality. The cells treated with HA formulations, did not show the same results, the

typical morphology and high survival rate could still be observed. The results of cell viability confirmed

the microscopic observation. The dehydration was responsible for almost 50% of mortality rate in the

Dry Eye sample, while the cells pre-treated with HA formulations presented higher survival rates, 60 –

70%, confirming a protective effect displayed by the HA formulations.

.

0

20

40

60

80

100

120

CR 0.30% CR 0.15% HA 0.30% HA 0.15%

Cell v

iab

ilit

y (

%) SDS

100 µg/mL

50 µg/mL

33 µg/mL

20 µg/mL

Medium

Page 85: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

72

Table 21- Optical microscope images of ARPE-19 after dehydration in no protective conditions

(Dry Eye), after dehydration preceded by treatment with HA formulations and cells not submitted

to dehydration (Medium). Without Die

Magnification 100x Crystal Violet

Magnification 200x Crystal Violet

Magnification 400x Cell

Viability (%)

Dry

Eye

50.7 ± 6.8

CR

0.3

0%

73.4 ± 12.6

HA

0.1

5%

62.7 ± 9.5

HA

0.3

0%

72.5 ± 6.2

Med

ium

100.0 ± 11.0

5.2.2.3.3. 3D model - Dry Eye Model and Cell Viability

The results of cell viability for the different tested samples are presented in Figure 22 showing a good

cell viability profile of ARPE-19 cell line. When applied CR 0.30%, HA 0.30% and HA 0.15% cell viability

increases when compared to the Dry Eye model. The survival rates were over 100% indicating that the

formulations were non-toxic so much so that provided a good environment for cell proliferation.

Page 86: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

73

Figure 22-Cell viability ARPE-19 cell line after dehydration treatment exposed for 24h to HA

0.15% and HA 0.30% eye drops solution and commercial formulation CR 0.30% (mean ± SD, n=9).

5.3. Discussion

The viscosity is one of the most important parameter for an eye drop solution, since it compromises the

efficacy of the treatment. A product with low viscosity may not assure the suitable retention time to treat

or relief symptoms of DED, but a too much viscosity may cause unwanted visual disturbances or some

blurriness. Several studies were performed in order to understand the difference in viscosity of the two

products, HA 0.15% and HA 0.30%, and to understand the mucoadhesion properties of this polymer

and eye drops solution [36].

The first study was the determination of the rheological profile of the two products. Observing the Figure

14 the values of shear stress are much higher for HA 0.30% when compared to HA 0.15%, which

indicates that it is much more viscous. These results make sense since HA 0.30% has the double of HA

concentration. The viscosity of the HA is due to the fact that is it a semi-flexible polymer, the higher the

concentration the higher the viscosity [120]. The three batches of each product present similar

rheological profile, both products do not present a constant relation between shear stress and shear

rate. The shear stress increases along the increase of the shear rate in a non-linear form, meaning that

this fluid behaves as a shear thinning fluid. The viscosity was also measured using an Ostwald

viscometer. The results shown that HA 0.30% is much more viscous than HA 0.15% with 71.20 mPa.s

and 6.83 mPa.s, respectively (Figure 15). These results support the outcomes obtained with the

Brookfield viscometer. The suspension of mucin prepared for this research work (5%, w/w) presents

viscosity since it is a high glycosylated protein with high MW.

To study the mucoadhesive properties of HA, samples of mucin with HA 0.15% and mucin with HA

0.30% were prepared and the results have shown there are indeed interactions between mucin and HA,

since the viscosity increased significantly when compared with the viscosity of HA 0.15% and HA 0.30%

eye drops solution or mucin alone. The mucoadhesive index was also determined and it demonstrated

that although both product have an increase superior to 50%, HA 0.30% + Mucin presented a massively

increase of 298.07% ± 19.90%. This increase suggests that a strong interaction between mucin and HA

0

20

40

60

80

100

120

CR 0.30% HA 0.30% HA 0.15% Dry Eye Medium SDS

Cell V

iab

ilit

y (

%)

Page 87: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

74

occurred, since HA 0.30% presents more concentration of HA, more interactions with the mucins were

possible. This increase of viscosity given by the interactions between mucin and the polymer are

possibly due to the formation of hydrogen bounding between the hydroxyl and carboxyl groups with

mucin’s amino groups. The HA is a linear molecule and can easily interpenetrate a mucin random coil

and that high molecular weight polymers can increase the probability of interfacial interactions with

mucin, creating a more stable connexion. These conclusions were given by Hassan and Gallo [121] and

their study with chitosan and mucin were the viscosity was measured at different pH levels. The

adhesive capacity was not solely due to electrostatic bonding, which showed to be ambiguous, but also

other types of bounding and interactions.

The adhesive properties of polymer are highly influenced by the viscosity as well the surface and

interfacial tensions of the polymer and substrate. In the pull away assay the adhesion was measured

throught a tack or probe testing, where by bringing a probe into contact with the surface of the polymer,

the mucin and the system polymer + mucin under a specific force and pulling the probe at a constant

velocity, the adhesive force was measured. While the probe is raised at a constant velocity it is measured

the necessary force in which the sample dissociates from the probe, resulting in a force versus distance

curve. The integral of that curve corresponding to the Area Under Force Time Curve represents the

adhesive strength of the sample [122]. The results show that are significant differences (p<0.05)

between the Normal Peak Force of HA 0.15% vs HA 0.30%, HA 0.15% vs Mucin + HA 0.15% and HA

0.30% vs Mucin + HA 0.30% (Table 20). The first case makes sense since HA 0.30% has the double

concentration of polymer, which makes it more viscous as discussed before. The significant difference

between HA 0.15% vs Mucin + HA 0.15% and HA 0.30% vs Mucin + HA 0.30% is an indication that the

addition of mucin created an interference with the polymer which formed a more viscous system,

concluding that a mucoadhesion occurred. The same conclusion was obtained with the pig eye, both

Normal Peak Force and Area Under Force Time Curve were higher with HA 0.30% than with HA 0.15%.

According to Hägerström and Edsman, who performed a similar assay, the strengthening might arise

from the entanglement of the polymer chains and the mucous glycoproteins, the formation of chemical

bonds and/or from dehydration of the mucous layer [46]. However, the Area Under Force Time Curve

values did not meet the same profile as the Normal Force, there were no significant difference between

HA 0.15% vs Mucin + HA 0.15% and HA 0.30% vs Mucin + HA 0.30%. These results may be due to the

fact that the samples variability was higher in this measurement and when performing the statistical data

analysis, it was not capable of detecting differences. Rather than having a p < 0.05, a p < 0.1 could be

sufficient to detect differences in those results. Values obtained with mucin were higher than the ones

with pig eye since the concentration of mucin is much higher in solution (5% w/w) than the concentration

existent in the eye.

In non-ideal fluids the response of the polymer will depend on frequency with both shear moduli (G’ and

G’’) increasing with frequency. In both products, HA 0.15% and HA 0.30% eye drops solution, the G’’

modulus is grater at low frequencies which indicates a fluid-like system (Figure 17 and Figure 18). With

the addition of mucin this profile still remains, but the values of both shear moduli increase. This means

that it is necessary a greater amount of force or stress to deform the sample along the plane of the

direction of the force, which indicates that some type of interaction has been established. According to

Page 88: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

75

Ludwig [8] the interaction between a mucoadhesive polymer and the mucin may occur by the following

mechanisms: physical entanglements, Van der Walls bonds, electrostatic forces and hydrogen bonds.

At low frequencies, the products and the system HA + Mucin suffer a rearranging due to Brownian

motion, physical entanglements are created and broken quickly compared to the rate of deformation, so

they do not store elastic energy. At high frequencies the polymer/polymer + mucin system does not have

time to rearrange causing the physical entanglements to persist longer than the oscillation frequency

constraining the polymer. The elastic energy is stored and the viscous dissipates that being the reason

why at high frequencies the G’ moduli is higher than G’’. The cross-over is than the point of passage

where the formulation stops presenting fluid-like characteristics and behaves more gel-like. The shear

moduli present greater values in HA 0.30% in comparison with HA 0.15%, which indicates that the

strength of the formulation/mucin interaction increases with HA concentration [56,122].

The ZP is related to the measurement of the surface charge that a specific material possesses or

acquires when suspended in a fluid. The results have shown that HA 0.15% and CR 0.15% ZP values

are more negatively charged than HA 0.30% and CR 0.30% (Figure 20). As said before the negative

charge of HA is due to the presence of carboxyl groups who dissociate at physiological pH [123]. By this

logic, the higher the HA concentration the higher the negative charge. However, the HA in this product

is at the form of sodium hyaluronate, which means it is in salt form. Positively charged ions from the

monovalent alkali metal series such as Na+ act as counter ions on anionic structures like HA, absorbing

on the surface-dominating negative sites decreasing the absolute value of the ZP. These results are in

accordance with Romero et al. and their work on silica with salts which demonstrated that the ZP value

of silica in absolute decreased with the addition of salts. The addition of salts reduces electrostatic

repulsion which facilitates the formation of H bounds, and thus an increase in viscosity occurs [124].

With the addiction of mucin both HA 0.15% and HA 30% ZP values increased in absolute, being the

Mucin + HA 0.30% a more negatively charged system. Mucin can be described as a double-globular

protein region connected by highly glycosylated linkers containing carboxylic and sialic acid, which

confers the negative charge at physiologic pH. The overall net charge is negative, but it may also exist

positively-charge regions in the non-glycosylated globular region containing histidine, arginine and

lysine residues. A study performed by Menchicchi et al. concluded that the polymers like chitosan

presents mucoadhesive properties due to the electrostatic interactions between positively-charged

polymer and negatively-charged mucin. However negatively-charge polymers such as alginates, pectins

and acrylic acid also show mucoadhesive properties, similar to the HA case. This means that the reason

for mucoadhesion on polymers is not solely due to electrostatic interactions but also due to other types

of interactions. Mucin forms a complex macromolecular network with available functional groups such

as sialic acid, therefore it is possible to interact with the polymer by hydrogen bounding between sialic

acid and the polymer’s carboxylate residue. It is also possible to form hydrophobic interactions with

mucin’s amino acids and entanglement of the polymer. The changes in ZP value were more significant

in HA 0.30% than HA 0.15% with the addition of mucin, because the first one presents double

concentration of HA, more HA allows more interactions and thus an increase of viscosity [57].

The time sweep test (Figure 19) shows that both shear moduli of the products and HA + Mucin systems

do not suffer alteration gradually with time. This indicates that overtime it does not occur changes of

Page 89: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

76

structure at a given oscillation frequency, concluding that stability is time independent. The same

happened in the ZP overtime study. The unchanged structure overtime and the increase of viscosity are

also indicators that the system HA + Mucin is stable. Stable interactions increase retention time, which

means that the contact time in the corneal surface remains longer. Since the product in study is a MD,

its action is of physical nature and not pharmacological, for that matter the longer the retention time the

more efficient is the treatment [36,43].

Cell viability was performed using ARPE-19 cell line in a 2D and 3D in vitro model that mimicked DED

conditions. By testing in this conditions, it is possible to study if HA 0.15% and HA 0.30% are promising

candidates for the treatment of DED. Frist, cell viability was performed using different dilutions of HA

0.15% and HA 0.30% to study which dilution is most suitable to use on the following assays, in other

words the less cytotoxic dilution (Figure 21). Four dilutions were made, 1:1, 1:2, 1:3 and 1:5. The results

concluded that the most suitable dilution was the formulation 1:5 dilution due to the high survival rate

(above 80%) in all tested samples.

A 2D in vitro assay was performed secondly to study the differences in the morphology and cell viability

of dehydrated cells who received a pre-treatment with HA formulations with cells who were not submitted

to the treatment (Table 21). It was also tested the commercial formulation CR 0.30% since this MD is

used in severer cases of DED. The results have shown that the cells who were not treated with HA

presented a disintegrated and dry membrane with a cell mortality rate close do 50% when compared to

the non-dehydrated cells. The pre-treated cells showed a morphology more similar to the hydrated cells

in both products with a high survival rate. Cells treated with HA 0.30% showed a higher cellular viability

rate of 72.5% ± 6.2%, almost equal to the CR 0.30% with 73.4% ± 12.6, when comparing to the ones

treated with HA 0.15% with 62.7% ± 9.5%. In the 3D model the results show that the application of all

three formulations obtained over 100% cell viability meaning that the application of these DM provided

a more suitable environment for cell proliferation. These results are very similar to the one performed

by Salzillo et al., were it was performed a study of cellular response of primary porcine corneal epithelial

cells when administrated different HA formulations [56]. They justified their results with the hypothesis

that the protective effect displayed by the HA formulation on the cells is related to the polymer water

retaining capacity. HA 0.30% presented higher cell viability values since more concentrated formulations

retain more water promoting higher hydration. This findings are important in the view of potential forms

of treatment for DED, the higher the HA concentration the higher the efficacy, which concludes that HA

0.30% may be more indicated for more severe cases of DED.

When comparing the 2D with the 3D in vitro assay the results of cell viability performed in both methods

were quite different. This is due to the fact that the 2D cell culture model is more sensitive than the 3D

model, this method only has a single layer which means when submitted to dehydration all cells were

exposed since all medium was removed. In the 3D model the dehydration is partial, the medium is

removed in the insert but it still exists in the well, simulating DED in vivo conditions.

Page 90: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

77

5.4. Conclusion

The aim of product characterization in this case was to evaluate the differences between the two

products, HA 0.15% and HA 0.30%, and to study their interactions with mucin, a form to predict their

behaviour in a biological environment.

All results obtained showed that HA 0.30% presented a higher viscosity than HA 0.15%.

The mucoadhesion between the HA and the mucin was tested and all results indicated that some kind

of interaction occurred between the mucin and the HA being stronger with HA 0.30%. Physical

entanglements and hydrogen bounding are possible forms of interaction.

The increase of viscosity when the mucin is added and the unchanged structure overtime are indicators

that the system HA + Mucin is a stable interaction, which increases the retention time in the corneal

surface, improving the efficacy of the treatment.

The cell viability test was performed with a 2D and 3D in vitro Dry Eye model. In the 2D model it was

concluded that the cells pre-treated with HA preserved the cell’s morphology after the dehydration

process and maintained a high survival rate. The 3D model demonstrated that the administration of HA

0.15% and HA 0.30% increased the cell viability over 100%. These values indicate that the application

of HA favours cell proliferation by creating an optimum environment. The reason for that environment

may be due to the hydration caused by the water retention by the HA molecules. From these results the

products, HA 0.15% and HA 0.30%, are potential candidates for becoming suitable MD for DED

treatment, being HA 0.30% most suited for more severe cases.

Page 91: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

78

Page 92: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

79

Chapter Six – Conclusion and Future Work

6. Concluding Remarks and Future Work

6.1. Concluding Remarks

The ophthalmic administration is the most common and well-accepted form of administration for the

treatment of eye disorders, such as DED. Symptoms of discomfort and visual disturbances causes by

DED may interfere with patience quality of life, being the application of some kind of treatment who

immediately attenuates the symptoms a big necessity. In modern society were smoking, alcohol

consumption, computer-related tasks, use of contact lenses and prolonged working hours in air-

conditioned facilities are part of some people daily activities can cause dry eye, which is why DED is the

one of the most common diagnosed ocular disease. The first–line therapy are lubricants also known as

eye drop comfort solutions or artificial tears which are non-prescription eye drops, classified as a MD

[36,43].

Polymers with water binding properties and high viscosity are highly used in ophthalmic preparations.

In artificial tears, polymers sooth, protect, lubricate and possess mucoadhesive properties with mucin

presented in the ocular surface, relieving the symptoms of dry eye [83]. One of the most popular and

effective is HA, the chosen polymer of Edol’s project in developing safe and efficient eye drops solution.

It was decided to invest in two products: HA 0.15% for daily use and 0.3% for more severe cases or

before sleep application. For the selection of the excipients these had to obey some required

specification to avoid ocular irritation or even damage. The pH value, osmolality and electrolyte

composition must be similar to the lacrimal fluid and the solution must be sterile. The preservative is one

of the most important excipients because it is a multidose MD, the solution must be able to kill

microorganisms or avoid microbial growth [39]. The chosen buffer was borate, potassium, magnesium

and calcium chloride as electrolytes and sodium chloride for osmolality adjustment. The selected

preservative was Suttocide (N-hydroxymethylglycinate 50%) with EDTA. This decision led to the

conclusion of the manufacturing process which conduce the design of the scale-up pilot batch

manufacturing process.

The Risk Analysis performed on the pilot batch manufacturing process demonstrated to be a complex

and a time consuming study since an intensive study of all the process steps had to be performed. The

two methods used, FMEA and Qualitative Analysis according to the ISO 14971:2007, presented very

different results from each other. From a long list of possible failure modes, FMEA was able to detect

14% unacceptable risks, while the ISO method only classified 11% as unacceptable. This demonstrated

that the FMEA is a more sensitive tool, detecting more risks and was capable of distinguish different

types of risks trough the RPN. After the detention, corrective actions to attenuate the risk had to be

applied in both methods. In the FMEA method it was possible to implement corrective actions in all the

unacceptable risks detected which diminished their risk level from unacceptable to acceptable. The ISO

method however was not capable of implementing corrective actions meaning that it was not also

capable of mitigating the unacceptable risks detected. This concludes that the FMEA besides of being

a generic tool was a much more useful tool, detecting and correcting unacceptable risks than the ISO

method, which is an application of risk management to MD.

Page 93: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

80

The Validation Plan was a required study since it is a new product and the manufacturing process must

produce consistently product within the specified requirements and quality. From the results obtained in

the IPC and FPC it was concluded that all values were consistent and respectful of the limit establish

for each process control. To conclude if the process can be validated the sterility test of HA 0.30% must

be performed.

The results from the Product Characterization showed that the viscosity increases with the increase of

HA concentration. This was proven by measuring viscosity through the Brookfield and Ostwald

viscometers, tack test, oscillatory frequency test and ZP measurements were in all cases the HA 0.30%

presented higher values than HA 0.15%. This proves that the viscosity increases with the increase of

concentration. The mucoadhesion was also measured trough the previous tests except the Brookfield

viscometer. The results showed that the viscosity, Normal force, both shear moduli (G’ and G’’) and ZP

value in absolute all increased in value once the mucin was in contact with HA, for both HA 0.15% and

HA 0.30%. This is a confirmation that interactions in fact occurred between the mucin and HA though

possibly entanglements and hydrogen bounding. The unchanged structure overtime time observed in

the time frequency test is an evidence that the structure is stable. Cell viability test in 2D results

demonstrated that cells pre-treated with both HA 0.15% and 0.30% maintained their morphology after

dehydration and that there was an increase of cell viability, demonstrating a protective effect. In the 3D

model the results showed that the cells treated with HA presented more than 100% cell viability

compared with the control Medium, indicating that they are in fact promising and effective MD.

6.2. Future Work

In future works besides from the cell viability test performed in ARPE-19 cell line, which are cells from

the retina, a new study of cell viability should be performed with cells from the corneal epithelium for

example HCE-T cell line, since DED effects the cornea and not so much the retina. Other in vitro test to

evaluate the effectiveness of the products could be through the quantification of the expression of

biomarkers related to different dry eye pathways such as: inflammatory mechanism at celular and matrix

level (TNF-α and MMP-19); adaptive and defence mechanism in case of lipidic film loss (MUC-4); water

channel protein that regulates water transport (AQP-3); and tight junction biomarkers for epithelial

integrity (OCLN and ZO-1), for example [125].

Mucoadhesion testing by ex vivo adhesion wash-off test method should be also interesting to perform

to compare the retention time of both HA 0.15% + Mucin and HA 0.30% + Mucin systems. This could

be performed using for example freshly excised pieces of intestinal mucosa from pig, a method used by

Veerareddy et al.[126].

Draize eye irritation in vivo test to observe changes of cornea, conjunctiva and iris in rabbit eye ball

following the exposure to the products should be also performed [127]. If the product passes this test,

human trials should be performed.

Page 94: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

81

References

1. Kumar A, Malviya R, Sharma PK. Recent Trends in Ocular Drug Delivery : A Short Review. Eur J Appl Sci. 2011;3(3):86–92.

2. Kim Y-C, Chiang B, Wu X, Prausnitz MR. Ocular delivery of macromolecules. Natl Institutes Heal. 2014;(September):172–81.

3. Morrison PWJ, Khutoryanskiy V V. Advances in ophthalmic drug delivery. Ther Deliv. 2014;5:1297–315.

4. “Facts About the Cornea and Corneal Disease- https://nei.nih.gov/health/cornealdisease”[Online].[Accessed 02-Out-2017].

5. “Understanding The Eye: Anatomy - http://healthlifemedia.com/healthy/understanding-the-eye-anatomy/ ”[Online].[Assessed: 12-Sep-2017].

6. “Understanding the Tear Film and Dry Eeye - http://www.collinsoptometrists.com.au/dry-eye-clinic/understanding-the-tear-film-and-dry-eye/”[Online].[Assessed: 11-Out-2017].

7. Bron AJ, Tiffany JM, Gouveia SM, Yokoi N, Voon LW. Functional aspects of the tear film lipid layer. Exp Eye Res. 2004;78:347–60.

8. Ludwig A. The use of mucoadhesive polymers in ocular drug delivery. Adv Drug Deliv Rev. 2005;57(11):1595–639.

9. Foulks GN. The Correlation Between the Tear Film Lipid Layer and Dry Eye Disease. Surv Ophthalmol. 2007;52(4).

10. Dartt D, Willcox M. Complexity of the tear film: Importance in homeostasis and dysfunction during disease. Exp Eye Res. 2013;(December):1–3.

11. Cwiklik L. Tear film lipid layer: A molecular level view. BBA - Biomembr [Internet]. 2016;1858(10):2421–30. Available from: http://dx.doi.org/10.1016/j.bbamem.2016.02.020

12. George S. On the nature and the role of the subsurface vesicles in the outer epithelial cells of the conjunctiva. Br J Ophthalmol. 1985;69:477–81.

13. Holly J, Mass USA. Formation and Rupture of the Tear Film. Exp Eye Res. 1973;(1):515–25.

14. Hodges RR, Dartt DA. Tear Film Mucins: Front Line Defenders of the Ocular Surface; Comparison with Airway and Gastrointestinal Tract Mucins. Exp Eye Res. 2013;117(December):62–78.

15. Tavares FDP, Fernandes RS, Bernardes TF, Bonfioli AA, Jorge E, Soares C. Dry Eye Disease. Inf Healthc. 2010;25(3):84–93.

16. Javadi M, Feizi S. Dry Eye Syndrome. J Ophthalmic Vis Res. 2011;6(3):192–8.

17. Whitcher JP. The treatment of dry eyes. Br J Ophthalmol. 2004;603–4.

18. Bernauer W, Broadway DC, Wright P. Chronic progressive conjunctival cicatrisation. Eye. 1993;7(3):371–8.

19. The Definition and Classification of Dry Eye Disease: Report of the Definition and Classification Subcommittee of the International Dry Eye Workshop (2007). Ocul Surf [Internet]. 2007;5(2):75–92. Available from: http://linkinghub.elsevier.com/retrieve/pii/S1542012412700812

Page 95: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

82

20. Colligris B, Alkozi HA, Pintor J. Recent developments on dry eye disease treatment compounds. Saudi J Ophthalmol [Internet]. 2014;28(1):19–30. Available from: http://dx.doi.org/10.1016/j.sjopt.2013.12.003

21. “Palovarotene for the Potential Treatment of Dry Eye Disease - http://clementiapharma.com/our-pipeline/palovarotene-for-dry-eye-disease/”[Online].[Accessed 17-Out-2017].

22. Yavuz B, Bozdağ Pehlivan S, Ünlü N. An Overview on Dry Eye Treatment: Approaches for Cyclosporin A Delivery. Sci World J [Internet]. 2012;2012:1–11. Available from: http://www.hindawi.com/journals/tswj/2012/194848/

23. “Food Supplements - https://ec.europa.eu/food/safety/labelling_nutrition/supplements_en ”[Online].[Accessed 06-Jan-2018].

24. Rand AL, York N. Current Opinion in Ophthalmology Nutritional Supplements for Dry Eye Syndrome. Natl Institutes Heal. 2012;22(4):279–82.

25. Akhtar S, Ahmed A, Randhawa MA, Atukorala S, Arlappa N, Ismail T, et al. Prevalence of vitamin A deficiency in South Asia: causes, outcomes, and possible remedies. J Health Popul Nutr. 2013;31(4):413–23.

26. Yildirim P, Garip Y, Karci AA, Guler T. Dry eye in vitamin D deficiency: More than an incidental association. Int J Rheum Dis. 2016;19(1):49–54.

27. Yang SF, Roberts JE, Liu QH, Pang J, Sarna T. Zeaxanthin and Lutein in the Management of Eye Diseases. J Ophthalmol. 2016;2016:1–2.

28. Shen G, Qi Q, Ma X. Effect of Moisture Chamber Spectacles on Tear Functions in Dry Eye Disease. Optom Vis Sci [Internet]. 2016;93(2):158–64. Available from: http://content.wkhealth.com/linkback/openurl?sid=WKPTLP:landingpage&an=00006324-201602000-00008

29. “Medical Devices - http://www.who.int/medical_devices/full_deffinition/en/”[Online].[Accessed 09-Jan-2018].

30. “European Commission: Comunicado de imprensa- http://europa.eu/rapid/press-release_IP-12-1011_pt.htm”[Online].[Accessed 20-Out-2017].

31. “Medical Devices - https://ec.europa.eu/growth/sectors/medical-devices_pt”[Online].[Accessed 09-Jan-2018].

32. INFARMED - Autoridade Nacional do Medicamento, e Produtos de Saúde IP. Dispositivos médicos. 2008; Available from: http://www.infarmed.pt/web/infarmed/entidades/dispositivos-medicos

33. Ttopstart. Certification and registration: Medical Devices on the European market. Spottlight [Internet]. 2013;(October). Available from: http://www.ttopstart.com/uploads/ttopstart_-_spottlight_report_-_Certification_and_registration_of_medical_devices.pdf

34. “Nova legislação aplicável aos Dispositivos Médicos – Infarmed - http://www.aenfermagemeasleis.pt/2017/05/05/nova-legislacao-aplicavel-aos-dispositivos-medicos-infarmed”[Online].[Accessed 06-Jan-2018].

35. British Standards Institution. A guide to the In Vitro Diagnostic Directive. 2012;1–8. Available from: http://www.bsigroup.com/meddev/LocalFiles/en-IN/Technologies/BSI-md-ivd-diagnostic-directive-guide-brochure-UK-EN.pdf

36. Tong L, Petznick A, Lee S, Tan J. Choice of Artificial Tear Formulation for Patients With Dry Eye. Cornea. 2012;31(11):S32–6.

Page 96: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

83

37. Schmidl D, Schmetterer L, Witkowska KJ, Unterhuber A, Aranha V, Kaya S, et al. Tear Film Thickness After Treatment With Artificial Tears in Patients With Moderate Dry Eye Disease. Cornea. 2015;34(4):421–6.

38. Freeman PD, Kahook MY. Preservatives in topical ophthalmic medications: historical and clinical perspectives. Expert Rev Ophthalmol. 2009;4(1):59–64.

39. Baudouin C, Labbé A, Liang H, Pauly A. Preservatives in eyedrops : The good , the bad and the ugly q. Prog Retin Eye Res [Internet]. 2010;29(4):312–34. Available from: http://dx.doi.org/10.1016/j.preteyeres.2010.03.001

40. Dutescu RM, Panfil C, Schrage N. Osmolarity of Prevalent Eye Drops, Side Effects, and Therapeutic Approaches. Cornea [Internet]. 2015;34(5):560–6. Available from: http://content.wkhealth.com/linkback/openurl?sid=WKPTLP:landingpage&an=00003226-201505000-00015

41. Schuerer N, Stein E, Inic-kanada A, Pucher M, Hohenadl C, Bintner N, et al. Implications for Ophthalmic Formulations : Ocular Buffers Show Varied Cytotoxic Impact on Human Corneal – Limbal. Cornea. 2017;36(6):712–8.

42. Ivarsson D, Wahlgren M. Comparison of in vitro methods of measuring mucoadhesion: Ellipsometry, tensile strength and rheological measurements. Colloids Surfaces B Biointerfaces [Internet]. 2012;92:353–9. Available from: http://dx.doi.org/10.1016/j.colsurfb.2011.12.020

43. Kashikar VS. Review Article Ophthalmic Mucoadhesive Polymers - A Literature Review. Ophthalmic Res. 2011;7(1):68–73.

44. Saraswathi B, Balaji A, Umashankar MS. Polymers in mucoadhesive drug delivery system-latest updates. Int J Pharm Pharm Sci. 2013;5(SUPPL 3):423–30.

45. Chickering DE, Mathiowitz E. Definitions, Mechanisms, and Theories of Bioadhesion. In: Mathiowitz E, Chickering DE, Lehr C-M, editors. Bioadhesive Drug Delivery Systems: Fundamentals, Novel Approaches and Develpment. New York: Marcel Dekker, Inc.; 1999. p. 1–10.

46. Hägerström H, Edsman K. Interpretation of mucoadhesive properties of polymer. Jorna. 2001;1589–99.

47. Benedetto DA, Shah DO, Kaufman HE. The instilled fluid dynamics and surface chemistry of polymers in the preocular tear film. Investig Ophthalmol. 1975;(December):887–902.

48. Russo E, Selmin F, Baldassari S, Gennari CGM, Caviglioli G, Cilurzo F, et al. A focus on mucoadhesive polymers and their application in buccal dosage forms. J Drug Deliv Sci Technol [Internet]. 2016;32:113–25. Available from: http://dx.doi.org/10.1016/j.jddst.2015.06.016

49. Boddeda B, Ratna JV, Battu H. A review on mucoadhesive polymers in ophthalmics. Int J Pharm Sci Rev Res. 2014;24(1):237–45.

50. Johnson ME, Murphy PJ, Boulton M. Effectiveness of sodium hyaluronate eyedrops in the treatment of dry eye. Graefe’s Arch Clin Exp Ophthalmol. 2006;244(1):109–12.

51. Shaikh R, Raghu T, Singh R, Garland MJ, David A, Donnelly RF. Mucoadhesive drug delivery systems. J Pharm Bioallied Sci. 2011;3(1):89–100.

52. Yu T, Andrews GP, Jones DS. Mucoadhesion and Characterization of Mucoadsive Properties. In: Neves J das, Sarmento B, editors. Mucosal Delivery of Biopharmaceuticals: Biology, Challenges and Strategies. Springer; 2014. p. 35–58.

53. Thirawong N, Nunthanid J, Puttipipatkhachorn S, Sriamornsak P. Mucoadhesive properties of various pectins on gastrointestinal mucosa : An in vitro evaluation using texture analyzer. Eur J

Page 97: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

84

Pharm Biopharm. 2007;67:132–40.

54. Bonacucina G, Martelli S, Palmieri GF. Rheological , mucoadhesive and release properties of Carbopol gels in hydrophilic cosolvents. Int J Pharm. 2004;282:115–30.

55. Andrews GP, Donnelly L, Jones DS, Curran RM, Morrow RJ, Woolfson AD, et al. Characterization of the Rheological , Mucoadhesive , and Drug Release Properties of Highly Structured Gel Platforms for Intravaginal Drug Delivery. Biomacromolecules. 2009;10:2427–35.

56. Salzillo R, Schiraldi C, Corsuto L, Agostino AD, Filosa R, Rosa M De, et al. Optimization of hyaluronan-based eye drop formulations. Carbohydr Polym [Internet]. 2016;153:275–83. Available from: http://dx.doi.org/10.1016/j.carbpol.2016.07.106

57. Menchicchi B, Fuenzalida JP, Hensel A, Swamy MJ, David L, Rochas C, et al. Biophysical Analysis of the Molecular Interactions between Polysaccharides and Mucin. Biomacromolecules. 2015;16(3):924–35.

58. V ESPB, Reinhart T, Peppas A. Solute diffusion in swollen membranes. part ii. influence of crosslinking on diffusive properties. J Memb Sci. 1984;18:227–39.

59. Edmondson R, Broglie JJ, Adcock AF, Yang L. Three-Dimensional Cell Culture Systems and Their Applications in Drug Discovery and Cell-Based Biosensors. Assay Drug Dev Technol [Internet]. 2014;12(4):207–18. Available from: http://online.liebertpub.com/doi/abs/10.1089/adt.2014.573

60. Ravi M, Paramesh V, Kaviya SR, Anuradha E, Paul Solomon FD. 3D cell culture systems: Advantages and applications. J Cell Physiol. 2015;230(1):16–26.

61. “3D Cell Culture 101 : An Introduction to 3D Cell Culture Tools and Techniques - http://www.sigmaaldrich.com/technical-documents/articles/biology/3d-biomatrix-white-paper-3d-cell-culture-101.html”[Online].[Acessed 27-Out-2017].

62. Larson B. 3D Cell Culture: A Review of Current Techniques. BioTek. 2015;6:1–10.

63. Sanyal S. Culture and Assay Systems Used for 3D Cell Culture. Corning. 2014;

64. Shafaie S, Hutter V, Cook MT, Brown MB, Chau DYS. In Vitro Cell Models for Ophthalmic Drug Development Applications. Biores Open Access [Internet]. 2016;5(1):94–108. Available from: http://online.liebertpub.com/doi/10.1089/biores.2016.0008

65. Barar J, Asadi M, Mortazavi-Tabatabaei SA, Omidi Y. Ocular Drug Delivery; Impact of in vitro Cell Culture Models. J Ophthalmic Vis Res [Internet]. 2009;4(4):238–52. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23198080%5Cnhttp://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=PMC3498862

66. Meloni M, Servi B De, Marasco D, Prete S Del. Molecular mechanism of ocular surface damage : Application to an in vitro dry eye model on human corneal epithelium. Mol Vis. 2011;(January):113–26.

67. White E, White E. Risk Management for Aseptic Processing. :22–30.

68. International Conference on Harmonisation (ICH) Quality Risk Management, Q9, ICH, November 2005.

69. Norma Portuguesa ISO 13485:2016. Dispositivos médicos Sistemas de Gestão da Qualidade e Requisitos para Fins Regulamentares., Instituto Portugês da Qualidade., 2017.

70. ISO 14971:2007 Medical Devises - Application of Risk Management to Medical Devises., Internacional Standard., 2007.

Page 98: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

85

71. Norma Portuguesa ISO 31000. Gestão de Risco. Princípios de Linhas de Orientação., Instituto Português da Qualidade., 2013. 2012;

72. International Conference on Harmonisation (ICH) of Technical Requirement for Registration of Pharmaceuticals for Human Use, Pharmaceutical Development, Q8 (R2), ICH, August 2009.

73. Walters KA, Brain KR. Dermatological Formulation and Transdermal Systems. Dermatological Transdermal Formul. 2002;

74. Lei L. Development of a New Lubricant and Nutrient Tear Substitute [Internet]. Der Universität zu Lübeck Aus der Medizinischen Fakultät. 2004. Available from: http://d-nb.info/978081064/34

75. International Conference on Harmonisation (ICH) of Technical Requirement for Registration of Pharmaceuticals for Human Use, Stability Testing of new Drugs and Products, Q1A (R2), ICH, 2003.

76. United States Phamacopeia 31/National Formulary 26. United States Pharmacopeial Convention, Inc. 2012;(c):21–3.

77. Matthews BR, Wall GM. Stability Storage and Testing of Ophthalmic Products for Global Registration. Drug Dev Ind Pharm. 2000;26(12):1227–37.

78. European Pharmacopeia 9.0 Eficacy of Antimicrobial Preservation, European Department for the Quality of Medicines within the Council of Europe, Strasbourg, section 5.1.3;2017:577-579.

79. Montés-Micó R, Cerviño A, Ferrer-Blasco T, García-Lázaro S, Madrid-Costa D. The Tear Film and the Optical Quality of the Eye. 2010;8(4):185–92.

80. Potvin R, Makari S, Rapuano CJ. Tear film osmolarity and dry eye disease: a review of the literature. Clin Ophthalmol [Internet]. 2015;9:2039–47. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26586933%5Cnhttp://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=PMC4636089

81. Norn MS. Tear fluid pH in normals, contact lens wearers, and pathological cases. 1988;66:485–9.

82. White CJ, Thomas CR, Byrne ME. Bringing comfort to the masses : A novel evaluation of comfort agent solution properties. Contact Lens Anterior Eye [Internet]. 2014;37(2):81–91. Available from: http://dx.doi.org/10.1016/j.clae.2013.07.004

83. Guillaumie F, Furrer P, Felt-Baeyens O, Fuhlendorff BL, Nymand S, Westh P, et al. Comparative studies of various hyaluronic acids produced by microbial fermentation for potential topical ophthalmic applications. J Biomed Mater Res - Part A. 2010;92(4):1421–30.

84. Ghelardi E, Celandroni F, Gueye SA, Salvetti S, Campa M, Senesi S. Antimicrobial Activity of a New Preservative for Multiuse Ophthalmic Solutions. 2013;29(6):586–90.

85. Carney LG, Mauger TF, Hill RM. Buffering in Human Tears : pH Responses to Acid and Base Challenge. 1989;30(4):747–54.

86. Houlsby RD, Ghajar M, Chavez G. Antimicrobial Activity of Borate-Buffered Solutions. 1986;(May):803–6.

87. “PubChem - https://pubchem.ncbi.nlm.nih.gov/”.[Online].[Accessed: 02-Aug-2017 ].

88. Scientific Committee on Consumer Safety. The Safety of Poly(hexamethylene) biguanide hydrochloride (PHMB). 2014;

89. “Chemical Book - http://www.chemicalbook.com/ChemicalProductProperty_EN_CB6233954.htm”.[Online].[Acces

Page 99: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

86

sed: 02-Aug-2017].

90. Scientific Committee on Consumer Safety. Safety the safety of poly ( hexamethylene ) biguanide hydrochloride ( PHMB ). 2014.

91. “sodium hydroxymethylglycinate - http://www.thegoodscentscompany.com/data/rw1303211.html”.[Online].[Accessed 02-Aug-2017].

92. The Scientific Committee On Cosmetic Products And Non-Food Products Intended For Consumers. Evaluation And Opinion on : The Determination Of Certain Formaldehyde Releasers In Cosmetic Products. 2002;(December).

93. Strasbourg: Council Of Europe : European Directorate for the Quality of Medicines and Healthcare. EP. European Pharmacopoeia 9.0. In 2017.

94. Liu L, Shaabana N, Sheardown HD. Hydrogel Materials Interacting with Polysaccharide Wetting Agents for Contact Lens Application. 2008;5(2):2008.

95. Rayahin JE, Buhrman JS, Zhang Y, Koh TJ, Richard A, Regeneration T, et al. High and low molecular weight hyaluronic acid differentially influence macrophage activation. ACS Biomater Sci Eng. 2015;1(7):481–93.

96. Potenzone R, A.J.Hopfinger. Conformational Analysis of Conformational Properties of Hyaluronic Acid and Sodium Hyaluronate. Polym J. 1978;10(November):181–99.

97. Fujii T, Sun Y, An K, Luo Z. Mechanical properties of single hyaluronan molecules. J Biomech. 2002;35:527–31.

98. Meyer K, Palmer JW. Polysaccharide of Vitreous Humor. J Biol Chem. 1934;(107):629–34.

99. “Sodium Hyalutonat, 95%- https://aksci.com/item_detail.php?cat=H926”[Online].[Accessed 12-Aug-2017].

100. “Sodium Hyaluronate - https://en.wikipedia.org/wiki/Sodium_hyaluronate”.[Online].[Accessed: 10-Aug-2017].

101. The European Agency for the Evaluation of Medical Products. Note for Guidance on Quality of Water for Pharmaceutical Use. 2002;(May):0–5.

102. WHO Expert Committee on Specifications for Pharmaceutical Preparations. Annex 2 - WHO good manufacturing practices: water for pharmaceutical use. 2005;(929):67–89.

103. Fischbarg J. The Biology of the Eye. First Edit. Fischbarg J, editor. New York: Elsevier; 2006. 394 p.

104. Furrer P, Mayer JM, Gurny R. Ocular tolerance of preservatives and alternatives. 2002;53:263–80.

105. Michael Levin, Pharmaceutical Process Scale up, Second edition, Volume 157.

106. European Medicines Agency. Guideline on process validation for finished products - information and data to be provided in regulatory submissions. 2016;44(November):1–15.

107. Patel D, Kaul R, Bhavsar A. Manufacturing Risk Assessment Study for Sterile Dry Powder Injection of Ceftriaxone Sodium. WjppsCom [Internet]. 2016;5(6):1143–53. Available from: http://www.wjpps.com/download/article/1464675961.pdf

108. “FAILURE MODE EFFECTS ANALYSIS (FMEA) - http://asq.org/learn-about-quality/process-analysis-tools/overview/fmea.html”. [Online].[Accessed: 03-Jul-2017].

Page 100: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

87

109. Murray B, Craft S. Failure Modes and Effects Analysis Template. In: The National Ecological Observatory Network. 2013.

110. U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER). Guidance for Industry Guidance for Industry - Investigating Out-of-Specification (OOS) Test Results for Pharmaceutical Production. 2006;1(August):14. Available from: http://www.fda.gov/downloads/Drugs/.../Guidances/ucm070287.pdf

111. Commitee for Proprietary Medical Products and Committe for Veterinary Medical Products; EMEA. Note for Guidance on Process Validation. 2001.

112. European Pharmacopeia 9.0 Sterility, European Department for the Quality of Medicines within the Council of Europe, Strasbourg, section 2.6.1;2017:185-188. In.

113. Mallu UR, Nair AK, Bandaru S, Sankaraiah J. Hold Time Stability Studies in Pharmaceutical Industry: Review. Pharm Reg Aff. 2012;1(4):1–8.

114. Rao M. Taking the mystery out of polymer activaion. Tall Oaks Publ Inc. 1995;

115. Jampel HD. Patient Preferences for Eye Drop Characteristics. Arch Ophthalmol [Internet]. 2003;121(4):540. Available from: http://archopht.jamanetwork.com/article.aspx?doi=10.1001/archopht.121.4.540

116. Kedik SA, Yartsev EI, Levachev SM, Panov A V, Sakaeva I V, Grigor OA, et al. Approaches To Eye Drops Efficiency Evaluation Based On Physicochemical Characteristics. 2011;45(3).

117. Dunn KC, Aotaki-Keen AE, Putkey FR, Hjelmeland LM. ARPE-19, a human retinal pigment epithelial cell line with differentiated properties. Exp Eye Res. 1996;62(2):155–69.

118. Assessing tackiness and adhesion using a pull away test on a rotational rheometer. Malvern Instruments Worldw. 2015;

119. Franck A. Understanding rheology of structured fluids. B TA instruments [Internet]. 2004;1–11. Available from: http://scholar.google.com/scholar?hl=en&btnG=Search&q=intitle:Understanding+Rheology+of+Structured+Fluids#2%5Cnhttp://scholar.google.com/scholar?hl=en&btnG=Search&q=intitle:Understanding+rheology+of+structured+fluids#2

120. Cowman MK, Schmidt TA, Raghavan P, Stecco A. Viscoelastic Properties of Hyaluronan in Physiological Conditions. F1000Research [Internet]. 2015;(0):1–13. Available from: http://f1000research.com/articles/4-622/v1

121. Hassan EE, Gallo JM. A Simple Rheological Method for the in Vitro Assessment of Mucin- Polymer Bioadhesive Bond Strength. Pharm Res. 1990;7(5):491–5.

122. Grillet AM, Wyatt NB, Gloe LM. Polymer Gel Rheology and Adhesion. J Rheol (N Y N Y) [Internet]. 2012; Available from: http://www.intechopen.com/books/rheology/rheology-and-adhesion-of-polymer-gels

123. Becker LC, Bergfeld WF, Belsito D V., Klaassen CD, Marks JG, Shank RC, et al. Final Report of the Safety Assessment of Hyaluronic Acid, Potassium Hyaluronate, and Sodium Hyaluronate. Int J Toxicol. 2009;28(4):5–67.

124. Romero CP, Jeldres RI, Quezada GR, Concha F, Toledo PG. Zeta potential and viscosity of colloidal silica suspensions: Effect of seawater salts, pH, flocculant, and shear rate. Colloids Surfaces A Physicochem Eng Asp [Internet]. 2018;538:210–8. Available from: http://dx.doi.org/10.1016/j.colsurfa.2017.10.080

125. Servi B De, Olmiere C, Meloni M, Assay T, Death C. Hyperosmolar dryness stress on 3D HCE model : a new tool for pre-clinical assessment of tear substitutes. :6031.

Page 101: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

88

126. Veerareddy PR. Preparation and evaluation of mucoadhesive cefdinir microcapsules. Adv Pharm Technol Res. 2011;2(2).

127. Lee M, Hwang J, Lim K. Alternatives to In Vivo Draize Rabbit Eye and Skin Irritation Tests with a Focus on 3D Reconstructed Human Cornea-Like Epithelium and Epidermis Models. Toxicol Res. 2017;33(3):191–203.

Page 102: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

89

Appendix

Figure A 1- Ishikawa diagram for possible failures in Weighting.

Page 103: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

90

Figure A 2- Ishikawa diagram for possible failures in Mixing.

Page 104: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

91

Figure A 3-Ishikawa diagram for possible failures in Pre and Sterile Filtration.

Page 105: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

92

Figure A 4- Ishikawa diagram for possible failures in Filling and Packing.

Page 106: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

93

Table A 1- FMEA and the ISO 14971 qualitative analyses of the manufacturing process of HA 0.15% and HA 0.30%.

SEV OCC DET SxOxD SxO

Pro

cess

Failure Modes Failure Effects

SEV (Mean ± SD)

Mode ISO 14971 Failure Cause

OCC (Mean ± SD)

Mode ISO 14971 Current Process Controls

DET (Mean ±

SD) Mode RPN CRIT

ISO 14971 Result

Weig

hti

ng

Raw Material Contamination during receipt

and transferring

Inappropriate quality of raw

material which directly

affects the quality of finished

dosage form

7.5 ± 0.6

7 Significant

Due to environment,

human interference

or by the vendor

2.0 ± 0.0

2 Low

Batch verification by operators and supervisors

before manufacture

3.5 ± 0.6

4 56 14 Low Risk

Equipment Wrongly cleaned

Contaminated product

8.0 ± 0.0

8 Significant Bad cleaning

practices 2.0 ± 0.0

2 Low

Room and equipment verification

by operators and

supervisors before

manufacture

2.0 ± 0.0

2 32 16 Low Risk

Raw material loss during

transference

Affects the quality of finished

dosage form

6.0 ± 1.2

5 Moderate Operating

errors 3.5 ± 0.6

3 Low Proper training

3.5 ± 0.6

4 60 15 Low Risk

Absence of Raw Material

Affects the production schedule

8.5 ± 0.6

8 Significant Failures of

supplier 1.5 ± 0.6

2 Low

Verification of raw material two weeks

before manufacture

3.5 ± 1.7

2 32 16 Low Risk

Long operator exposure

during handling

Contaminated product

4.5 ± 0.6

5 Moderate Operating

errors 3.0 ± 0.0

3 Low

Utilization of Personal protective equipment

2.5 ± 0.6

3 45 15 Low Risk

Laminar Flux failure

Contaminated product

7.5 ± 0.6

8 Significant Equipment malfunction

2.5 ± 0.6

2 Low

Semestral verification

and every six months

2.5 ± 0.6

2 32 16 Low Risk

Page 107: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

94

Table A 1- FMEA and the ISO 14971 qualitative analyses of the manufacturing process of HA 0.15% and HA 0.30% (Continuation).

SEV OCC DET SxOxD SxO

Pro

cess

Failure Modes Failure Effects

SEV (Mean ± SD)

Mode ISO

14971 Failure Cause

OCC (Mean ± SD)

Mode ISO

14971

Current Process Controls

DET (Mean ± SD)

Mode RPN CRIT ISO

14971 Result

Weig

hti

ng

Uncalibrated scale

Affects the quality of finished

dosage form

6.0 ± 1.15

5 Moderate Equipment malfunction

2.5 ± 0.6

3 Low

Annual calibration and daily

verification with two

calibrated weights.

Fortnightly verification with more calibrated weights

2.5 ± 0.6

3 45 15 Low Risk

HEPA filter lack of integrity

Contaminated product

8.5 ± 0.6

8 Significant

Improper HEPA filter efficiency,

variations in velocity of air, flow

pattern of air or

leaking of the filter

2.5 ± 0.6

2 Low

Semestral verification

and every six months

2.0 ± 0.0

2 32 16 Low Risk

Procedure failures

Affects the quality of finished

dosage form

7.5 ± 0.6

7 Significant Invalidated or Unmet procedure

4.0 ± 0.0

4 Medium

Required filling of the

Batch Master Record

3.0 ± 0.0

3 84 28 High Risk

Non-verification of criteria

Inadequate weighing chamber

conditions result in poor final product

quality

6.5 ± 0.6

7 Significant Unmet

procedures 3.0 ± 0.0

3 Low

Verification of the

conditions within the

specifications

3.0 ± 0.0

3 63 21 Low Risk

Misuse use of uniform Contaminated

product 7.0 ± 0.0

7 Significant Wrong or

incomplete training

2.0 ± 0.0

2 Low Uniform

procedure 2.0 ± 0.0

2 28 14 Low Risk

Page 108: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

95

Table A 1- FMEA and the ISO 14971 qualitative analyses of the manufacturing process of HA 0.15% and HA 0.30% (Continuation).

SEV OCC DET SxOxD SxO

Pro

cess

Failure Modes Failure Effects

SEV (Mean ± SD)

Mode ISO

14971 Failure Cause

OCC (Mean ± SD)

Mode ISO

14971

Current Process Controls

DET (Mean ± SD)

Mode RPN CRIT ISO

14971 Result

Weig

hti

ng

Wrong amount of raw material

Affects the quality of finished

dosage form

5.5 ± 0.6

5 Moderate

Weighing errors/ Unmet

procedures

2.0 ± 0.0

2 Low

Raw materials are checked in duplicate

and preservatives

and API´s checked by supervisors

before manufacture

2.0 ± 0.0

2 20 10 Low Risk

Untrained operators

Affects the quality of finished

dosage form

6.5 ± 1.7

8 Significant Wrong or

incomplete training

2.5 ± 0.6

3 Low Initial and

continuous training

2.5 ± 0.6

3 72 24 Low Risk

Mix

ing

Incomplete dissolution/

Insufficient mixing time

Affects the quality of finished

dosage form

7.0 ± 0.0

7 Significant

Little mixing time,

mixing blade with little force or velocity

3.0 ± 1.2

3 Low

Visual control and validation

of the manufacturing

process

2.5 ± 0.6

2 42 21 Low Risk

Excess/Low Capacity of the mixer

Equipment malfunction, affects the quality of finished

dosage form

4.5 ± 0.6

5 Moderate Wrong batch

volume

3.0 ± 0.6

3 Low

Visual control and validation

of the manufacturing

process

3.5 ± 1.7

2 30 15 Low Risk

Velocity Failure

Incomplete dissolution, effects the quality of finished

dosage form

6.0 ± 1.2

7 Significant Equipment malfunction

3.5 ± 0.6

3 Low Backup shaker

4.0 ± 0.0

4 84 21 Low Risk

Change raw material placement order

Affects the quality of finished

dosage form

4.5 ± 0.6

5 Moderate Improper working of personnel

3.5 ± 0.0

3 Low

Batch Master Record with

all the necessary

steps

3.5 ± 0.6

4 60 15 Low Risk

Page 109: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

96

Table A 1- FMEA and the ISO 14971 qualitative analyses of the manufacturing process of HA 0.15% and HA 0.30% (Continuation).

SEV OCC DET SxOxD SxO

Pro

cess

Failure Modes Failure Effects

SEV (Mean ± SD)

Mode ISO

14971 Failure Cause

OCC (Mean ± SD)

Mode ISO

14971

Current Process Controls

DET (Mean ± SD)

Mode RPN CRIT ISO

14971 Result

Mix

ing

Raw material loss during transference

Affects the quality of finished

dosage form

5.5 ± 0.6

5 Moderate Operating

errors 3.3 ± 1.2

4 Medium Proper training

3.0 ± 1.2

4 80 20 Low Risk

Equipment Wrongly cleaned

Contaminated product

7.0 ± 0.0

5 Moderate Bad cleaning

practices 3.5 ± 0.6

3 Low

Room and equipment verification

by operators and

supervisors before

manufacture

3.0 ± 0.0

3 63 21 Low Risk

Misuse use of uniform Contaminated

product 7.0 ± 0.0

7 Significant Wrong or

incomplete training

3.5 ± 0.6

3 Low Uniform

procedure 2.5 ± 1.7

4 84 21 Low Risk

Procedure failures

Affects the quality of finished

dosage form

4.5 ± 0.6

5 Moderate Invalidated or

Unmet procedure

3.0 ± 0.6

3 Low

Required filling of the

Batch Master Record

5.0 ± 2.3

3 45 15 Low Risk

Incorrectly mounted equipment

Equipment malfunction, affects the quality of finished

dosage form

5.5 ± 0.6

5 Moderate Unmet

procedures 3.0 ± 0.6

3 Low Proper training

3.0 ± 0.0

3 45 15 Low Risk

IPC

/FP

C

Product harvest in an inappropriate container

Contaminated product

9.0 ± 0.0

9 Significant

Due to improper storage of containers,

environment, human

interference

3.0 ± 0.0

3 Low The

bioburden is determined

3.0 ± 0.0

3 81 27 Low Risk

Page 110: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

97

Table A 1- FMEA and the ISO 14971 qualitative analyses of the manufacturing process of HA 0.15% and HA 0.30% (Continuation).

SEV OCC DET SxOxD SxO

Pro

cess

Failure Modes Failure Effects

SEV (Mean ± SD)

Mode ISO

14971 Failure Cause

OCC (Mean ± SD)

Mode ISO

14971 Current Process

Controls

DET (Mean ± SD)

Mode RPN CRIT ISO

14971 Result

IPC

/FP

C

Presence of colour and odour

Affects the quality of finished dosage

form

7.3 ± 2.1

9 Significant Bad

manufacturing practices

2.3 ± 1.3

1 Low

Samples are taken from the top and bottom for visual

check

3.3 ± 1.3

1 9 9 Low Risk

High/Low pH

Affects the quality of finished dosage

form

7.3 ± 2.1

9 Significant

Failures in weighting or loss of raw

material

2.3 ± 1.3

4 Medium

pH meter is calibrated daily. pH specified in batch

record and readings are

double verified. QC confirms

3.3 ± 1.3

1 36 36 High Risk

High/Low osmolality

Affects the quality of finished dosage

form

8.5 ± 1.0

9 Significant

Failures in weighting or loss of raw

material

3.8 ± 1.3

4 Medium

Osmometer is calibrated monthly against patterns

solutions.Calibrated daily against a

reference solution. Osmolality

specified in batch record and osmolalities readings are

double verified. After all

adjustments QC confirms

1.8 ± 1.0

2 72 36 High Risk

Page 111: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

98

Table A 1- FMEA and the ISO 14971 qualitative analyses of the manufacturing process of HA 0.15% and HA 0.30% (Continuation).

SEV OCC DET RPN CRIT

Pro

cess

Failure Modes Failure Effects

SEV (Mean ± SD)

Mode ISO

14971 Failure Cause

OCC (Mean ± SD)

Mode ISO

14971

Current Process Controls

DET (Mean ± SD)

Mode RPN CRIT ISO

14971 Result

IPC

/FP

C

High/Low Viscosity

Affects the quality of finished

dosage form

7.5 ± 0.6

8 Significant

Failures in weighting or loss of raw

material

4.3 ± 1.3

4 Medium

Viscometer calibrated

annually by an external

entity. Viscosity

specified in batch record

and readings are

double verified. QC

confirms

1.8 ± 1.0

1 32 32 High Risk

Absence of Sodium Hyaluronate

Wrong formulation

8.5 ± 0.6

9 Significant

Failures in weighting or loss of raw

material

1.0 ± 0.0

1 Low

Identification of Sodium

Hyaluronate by IR

method

1.5 ± 1.0

1 9 9 Low Risk

Sodium Hyaluronate UV Assay out of range

Wrong formulation

8.5 ± 0.6

9 Significant

Failures in weighting or loss of raw

material

3.5 ± 1.3

4 Medium

Sodium Hyaluronate percentage specified in

batch record determined by UV/Vis method.

1.8 ± 0.5

2 72 36 High Risk

Elevated Microbial concentration/ non-

sterile

Contaminated product

9.0 ± 0.8

9 Significant

Operating errors in

past procedures

3.8 ± 1.3

4 Medium

The

bioburden is

determined/

Sterility test

3.3 ± 0.5

3 108 36 High Risk

False Results

Validated certificates with wrong

results

9.3 ± 1.0

10 Significant Poorly

performed analysis

1.8 ± 0.5

2 Low

All methods are

previously validated

3.5 ± 1.3

4 80 20 Low Risk

.

Page 112: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

99

Table A 1- FMEA and the ISO 14971 qualitative analyses of the manufacturing process of HA 0.15% and HA 0.30% (Continuation).

SEV OCC DET RPN CRIT

Pro

cess

Failure Modes Failure Effects

SEV (Mean ± SD)

Mode ISO 14971 Failure Cause

OCC (Mean ± SD)

Mode ISO 14971 Current Process Controls

DET (Mean ±

SD) Mode RPN CRIT

ISO 14971 Result

Pre

an

d S

teri

le

Fil

trati

on

HEPA filter lack of integrity

Contaminated product

7.5 ± 3.0

8 Significant

Improper HEPA filter efficiency,

variations in velocity of air, flow pattern of air or leaking of the filter

4.0 ± 4.0

2 Low Verification by

external entities

3.5 ± 2.4

2 32 16 Low Risk

Laminar Flux failure

Contaminated product

6.8 ± 2.5

8 Significant Equipment malfunction

2.3 ± 0.5

2 Low

Every six months

verification and room, equipment

and operators environmental

control

3.0 ± 2.7

2 32 16 Low Risk

Membrane Integrity failure

Unable to sterilized

8.3 ± 1.3

8 Significant

Defective membrane,

misplacement of the

membrane

3.4 ± 3.5

2 Low Bubble Point

Control 3.3 ± 2.5

2 32 16 Low Risk

Product obstruction

Bubble point failure

6.8 ± 2.5

5 Moderate

Improper equipment,

elevated product viscosity

5.0 ± 3.5

4 Medium

Deposit capable of

pressurizing at high

pressures

2.3 ± 0.5

2 40 20 Low Risk

Non-sterile equipment/parts

Contaminated product

5.0 ± 3.6

8 Significant Bad cleaning

practices 1.5 ± 0.6

2 Low

Validation of autoclave

loads. Well packaged material

3.8 ± 3.0

3 48 16 Low Risk

Page 113: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

100

Table A 1- FMEA and the ISO 14971 qualitative analyses of the manufacturing process of HA 0.15% and HA 0.30% (Continuation).

SEV OCC DET RPN CRIT

Pro

cess

Failure Modes Failure Effects

SEV (Mean ± SD)

Mode ISO

14971 Failure Cause

OCC (Mean ± SD)

Mode ISO

14971

Current Process Controls

DET (Mean ± SD)

Mode RPN CRIT ISO

14971 Result

Pre

an

d S

teri

le

Fil

trati

on

Contaminated Filter Contaminated

product 9.0 ± 1.2

8 Significant Poor filter

quality 3.8 ± 4.2

2 Low

Sterilization of the filter in an

autoclave with a valid

load whenever there is a

filling

7.5 ± 2.9

5 80 16 Low Risk

Broken/ Misplaced Filter

Unfiltered Product

8.0 ± 0.8

8 Significant Operating

errors/ Poor filter quality

3.8 ± 4.2

2 Low Bubble Point

Control 3.3 ± 2.5

2 32 16 Low Risk

Procedure failures

Affects the quality of finished

dosage form

7.7 ± 1.5

7 Significant Invalidated or Unmet procedure

4.3 ± 3.2

3 Low

Required filling of the

Batch Master Record

3.3 ± 0.5

3 63 21 Low Risk

Misuse use of uniform Contaminated

product 8.0 ± 0.8

8 Significant Wrong or

incomplete training

4.3 ± 3.3

2 Low

Uniform procedure

and operators environmental

control

1.5 ± 0.6

2 32 16 Low Risk

Incorrectly mounted filtration system

Equipment malfunction, affects the quality of finished

dosage form

8.0 ± 0.8

8 Significant Operating

errors 4.5 ± 3.7

3 Low Proper training

2.5 ± 1.0

2 48 24 Low Risk

Filli

ng

an

d P

ackin

g

HEPA filter lack of integrity

Contaminated product

9.0 ± 1.2

8 Significant

Improper HEPA filter efficiency,

variations in velocity of air, flow

pattern of air or

leaking of the filter

4.3 ± 3.9

2 Low Verification by

external entities

3.5 ± 1.9

2 32 16 Low Risk

Page 114: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

101

Table A 1- FMEA and the ISO 14971 qualitative analyses of the manufacturing process of HA 0.15% and HA 0.30% (Continuation).

SEV OCC DET RPN CRIT

Pro

cess

Failure Modes Failure Effects

SEV (Mean ± SD)

Mode ISO

14971 Failure Cause

OCC (Mean ± SD)

Mode ISO

14971

Current Process Controls

DET (Mean ± SD)

Mode RPN CRIT ISO

14971 Result

Filli

ng

an

d P

ackin

g

Laminar Flux failure Contaminated

product 6.8 ± 2.5

8 Significant Equipment malfunction

2.5 ± 0.6

2 Low

Every six months

verification and room, equipment

and operators environmental

control

2.6 ± 2.2

2 32 16 Low Risk

Non-sterile equipment/parts

Contaminated product

5.5 ± 3.3

8 Significant Bad

cleaning practices

2.5 ± 1.0

3 Low

Validation of autoclave

loads. Well packaged material

2.8 ± 3.5

1 24 24 Low Risk

Filling area unclean Loss of sterility

8.3 ± 0.5

8 Significant

Failure in maintenance

of area classification

3.5 ± 3.7

2 Low

Cleaning procedure and room

environmental control

2.3 ± 1.3

2 32 16 Low Risk

Uniformity of Volume Failure

Final product with wrong

volume

5.5 ± 2.5

5 Moderate

Uncalibrated equipment scale, non-verification of the scale

4.0 ± 3.4

2 Low

Tare of 12 empty bottles, 10 bottles for

weight adjustment, control of

liquid weight every 30 minutes

3.4 ± 2.0

2 20 10 Low Risk

Filling material with defects

Loss of sterility, loss of integrity

6.0 ± 2.5

6 Moderate Failures of

supplier 3.8 ± 4.2

2 Low

Inline sealing in all batch, defective

material is rejected

2.8 ± 1.5

4 48 12 Low Risk

Sealing defective Loss of sterility

8.3 ± 0.5

8 Significant

Defective sealant, machine failure

4.0 ± 3.4

2 Low

Inline sealing in all batch, defective

material is rejected

1.8 ± 0.5

2 32 16 Low Risk

Page 115: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

102

Table A 12- FMEA and the ISO 14971 qualitative analyses of the manufacturing process of HA 0.15% and HA 0.30% (Continuation).

SEV OCC DET RPN CRIT

Pro

cess

Failure Modes Failure Effects

SEV (Mean ± SD)

Mode ISO

14971 Failure Cause

OCC (Mean ± SD)

Mode ISO

14971

Current Process Controls

DET (Mean ± SD)

Mode RPN CRIT ISO

14971 Result

Filli

ng

an

d P

ackin

g

Incorrectly mounted equipment

Equipment malfunction, affects the quality of finished

dosage form

8.3 ± 0.5

8 Significant Unmet

procedures 4.3 ± 3.4

2 Low Proper training

2.5 ± 1.0

3 48 16 Low Risk

Procedure failures

Affects the quality of finished

dosage form

7.8 ± 1.0

7 Significant Invalidated or

Unmet procedure

4.3 ± 2.5

3 Low

Required filling of the

Batch Master Record

3.0 ± 0.0

3 63 21 Low Risk

Misuse use of uniform Contaminated

product 8.3 ± 0.5

8 Significant Wrong or

incomplete training

4.0 ± 3.4

2 Low

Uniform procedure

and operators environmental

control

1.5 ± 0.6

2 32 16 Low Risk

Labelling errors

Misleading labels and

market complains

6.8 ± 2.1

5 Moderate

Improper checking of

labels including

Names and amount of

active ingredients,

Storage requirements, Control or lot

number, Appropriate

auxiliary labelling

(including precautions)

4.5 ± 3.8

2 Low

Verification and approval by the Quality Control, batch

number confirmation,

date of manufacture and validity confirmation carried out

inline

3.5 ± 1.7

3 30 10 Low Risk

Page 116: Design, testing, and scale-up of medical device class IIb ... Angelica...Design, testing, and scale-up of medical device class IIb: risk analysis as a crucial tool for pharmaceutical

103

Table A 1- FMEA and the ISO 14971 qualitative analyses of the manufacturing process of HA 0.15% and HA 0.30% (Continuation). SEV OCC DET RPN CRIT

Pro

cess

Failure Modes Failure Effects

SEV (Mean ± SD)

Mode ISO

14971 Failure Cause

OCC (Mean ± SD)

Mode ISO

14971

Current Process Controls

DET (Mean ± SD)

Mode RPN CRIT ISO

14971 Result

Sto

rag

e

Mixing up two different types of raw material

Wrong product

storage and use

7.5 ± 0.6

7 Significant Improper working of personnel

2.0 ± 0.0

2 Low

Specific places to

avoid misplacement

2.3 ± 0.5

2 28 14 Low Risk

Mixing up two different types of packing

material

Wrong product

storage and use

7.0 ± 0.0

7 Significant Improper working of personnel

2.0 ± 0.0

2 Low

Specific places to

avoid misplacement

2.0 ± 0.0

2 28 14 Low Risk

Wrong stability test conditions

Wrong data regarding

the product stability

8.0 ± 0.0

8 Significant

Operating errors in product quantity,

storage time and type of incubator

2.3 ± 0.5

3 Low

Training of operators, instructions documented

in batch record. Storage

conditions verified in validation. Automatic email alert.

Temperature checked

twice a day by an

operator and registration on the form.

1.3 ± 0.5

2 48 24 Low Risk

Storage room conditions out of

specification

Degradation of product

7.0 ± 0.0

7 Significant

Storage room

temperature, humidity

2.0 ± 0.0

2 Low

Stability studies.

Automatic email alert.

Temperature checked

twice a day by an

operator and registration on the form.

2.5 ± 0.6

2 28 14 Low Risk