159
Universidad del Turabo Detection of Escherichia coli ATCC® 8739™ and Aspergillus brasiliensis ATCC®16404™ in Raw Materials and Pharmaceutical Products Using the Real- Time PCR in Comparison with Standard Conventional Microbiological Methods By Elsie Jacqueline Hernández García BS, Biology, Interamerican University of Puerto Rico MS, Science, University of Puerto Rico Mayagüez Campus Dissertation Submitted to the School of Science and Technology of the Universidad del Turabo In partial fulfillment of the requirements for the degree of Doctor of Philosophy In Environmental Science Biology Option Gurabo, Puerto Rico November, 2015

Detection of Escherichia coli ATCC® 8739™ and …bv.ut.suagm.edu/sites/default/files/resource_files/Elsie Jacqueline... · to 10¯¹⁵mL by the sensitivity test and detected the

  • Upload
    lamnhi

  • View
    220

  • Download
    0

Embed Size (px)

Citation preview

Universidad del Turabo

Detection of Escherichia coli ATCC® 8739™ and Aspergillus brasiliensis

ATCC®16404™ in Raw Materials and Pharmaceutical Products Using the Real-

Time PCR in Comparison with Standard Conventional Microbiological Methods

By

Elsie Jacqueline Hernández García BS, Biology, Interamerican University of Puerto Rico

MS, Science, University of Puerto Rico Mayagüez Campus

Dissertation

Submitted to the School of Science and Technology of the Universidad del Turabo

In partial fulfillment of the requirements for the degree of Doctor of Philosophy

In Environmental Science

Biology Option

Gurabo, Puerto Rico

November, 2015

UNIVERSIDAD DEL TURABO

CERTIFICATION OF DISSERTATION APPROVAL

The dissertation of Elsie Jacqueline Hernández García, was reviewed and

approved by the members of the Dissertation Committee. The Doctoral Academic

Requirements Compliance form, signed by the committee members, has been deposited

in the Register’s Office and at the Graduate Studies & Research Center in the

Universidad del Turabo.

DISSERTATION COMMITTEE MEMBERS Adalberto Bosque, Ph.D., MBA., REM CEA, CESCO Public Health Program Ponce Health Sciences University Research Advisor Teresa Lipsett, Ph.D. Universidad del Turabo Supervising Professor Ángel Rivera, MD, MBA Universidad del Turabo Member Eileen Villafañe, Ph.D. Research Laboratory of PR Environmental Quality Board Member Santander Nieto, Ph.D. Universidad del Turabo Member

© Copyright 2015

Elsie Jacqueline Hernández García. All Rights Reserved.

iv

Abstract Elsie Jacqueline Hernández (Ph.D., Environmental Science)

Detection of Escherichia coli ATCC® 8739™ and Aspergillus brasiliensis

ATCC®16404™ in Raw Materials and Pharmaceutical Products Using the Real-Time

PCR in Comparison with Standard Conventional Microbiological Methods

(November/2015)

Abstract of a doctoral dissertation at the Universidad del Turabo

Dissertation supervised by Adalberto Bosque, Ph.D., MBA., REM

No. of pages in text: 163.

Pharmaceutical products are susceptible to microbial contamination. This type of

contamination could represent a risk to consumers health. Furthermore, microbial

contamination can cause degradation, changes in the aesthetic of the product, and loss

of drug effectiveness by reducing or inactivate the therapeutic activity of the product. The

techniques used in the pharmaceutical industry are conventional techniques where the

practice of methods of transference of cultures, phenotypic observation of the colony, and

biochemical tests for its final identification prevails. These conventional techniques are

time-consuming, not specific, and lack accuracy and precision to demonstrate the present

of specified organisms in a sample. This analytical methodology results in delays for the

final approval of products.

A sensitive Real-time Polymerase Chain Reaction method with TaqMan® MGM

probe was developed in this research for precision, specificity and rapid detection of

objectionable microorganisms in raw material and finished product. The Real-time PCR

method had amplification of Escherichia coli ATCC® 8739™ DNA in all dilutions from 10¯¹

to 10¯¹⁵mL by the sensitivity test and detected the bacteria in the raw materials and OTC

v

samples analyzed at 10¯⁷ and 10¯¹⁵mL for Test for Specified Microorganisms (TSM).

Also, in Aspergillus brasiliensis ATCC®16404™ was obtained amplification in all samples

from sample 10¯¹ to 10¯¹⁵mL for sensitivity test and 10¯³ and 10¯¹⁵mL for detection of the

presence of the fungi in TSM. There is a statistically significant differences in the detection

of Escherichia coli ATCC® 8739™ and ATCC® 16404™ Aspergillus brasiliensis in

conventional and the rt-PCR methods. The conventional method did not have the ability

to detect small traces of the 10¯¹⁵ sample dilutions while the Real-time PCR method was

able to detect at this dilution. This research demonstrates how quickly, precisely, and

accurately detect the presence of these objectionable organisms in terms of the minimum

traces as 4 μl of pharmaceutical sample, something not possible under the conventional

USP pour plate method. The Real-time PCR methodology contributes to the rapid

detection of objectionable organisms in a pharmaceutical sample preventing the risk of

exposure of humans and animals to contaminated drugs that do not meet the FDA and

USP quality standards.

vi

Resumen

Elsie Jacqueline Hernández (Ph.D., Environmental Science)

Detección de Escherichia coli ATCC® 8739™ y Aspergillus brasiliensis ATCC®16404™

en Materia Prima y Producto Farmaceútico usando PCR en Tiempo Real en

Comparación con el Método Estándar Convencional Microbiológico

(noviembre/2015)

Resumen de una disertación doctoral en la Universidad del Turabo.

Disertación supervisada por Adalberto Bosque, Ph.D., MBA.,

No. de páginas en texto: 163.

Los productos farmacéuticos son susceptibles a contaminación microbiana. Este

tipo de contaminación representa un riesgo a la salud de los consumidores. Ocasionando

degradación, cambios en la estética del producto y la posible pérdida de la efectividad de

la droga. La presencia de ciertos microorganismos en preparaciones no estériles pueden

tener el potencial de reducir o inactivar la actividad terapéutica del producto. Las técnicas

utilizadas en la industria farmacéutica descrita en la Farmacopea son convencionales

donde prevalece la práctica de métodos de transferencia de cultivos, observación

fenotípica de la colonia y pruebas bioquímicas para su identificación final. Estas técnicas

convencionales consumen tiempo, no son específicas, carecen de exactitud y precisión

para demostrar la presencia de organismos específicos en una muestra resultando en el

retraso para la aprobación del producto.

Un método sensitivo de PCR en tiempo real (PCR-rt) con sonda TaqMan® MGM

fue desarrollado en esta investigación para precisión, especificidad y detección rápida de

microorganismos objetables en materia prima y producto terminado. Este método

amplificó el ADN de Escherichia coli ATCC® 8739 ™ en todas las diluciones desde 10¯¹

vii

hasta 10¯¹⁵mL para la prueba de sensitividad. Además,detectó la bacteria en las

muestras analizadas de materia prima y OTC en las diluciones 10¯⁷ y 10¯¹⁵mL en la

prueba de microorganismos especificos (TSM). En Aspergillus brasiliensis

ATCC®16404™ se obtuvo amplificación en todas las muestras desde 10¯¹ hasta 10¯¹⁵ mL

en la prueba de sensitividad y 10¯³ y 10¯¹⁵ mL en la prueba para detección del hongo en

TSM. Hay unas diferencias estadísticamente significativas en la detección de Escherichia

coli ATCC® 8739 ™ y ATCC® 16404 ™ Aspergillus brasiliensis tanto en el métodos

convencional de vertido en plato y el método molecular de PCR-rt. El método

convencional no tuvo la capacidad de detectar pequeñas trazas de la muestra contenida

en la dilución 10¯¹⁵mL, mientras que el método de PCR-rt fue capaz de detectar la

muestra en esta dilución. La metodología de PCR-rt desarrollada en esta investigación

contribuye a la rápida detección de objetables en muestras farmacéuticas previniendo el

riesgo de exposición de humanos y animales a medicamentos contaminados que no

cumplen con los estándares de calidad.

viii

Vita

Elsie Jacqueline Hernández García was born in the community of Captain Correa

in Arecibo, Puerto Rico. She holds a Bachelor of Science in Biology (BS) of the Inter

american University of Puerto Rico and obtained Master of Science (MS) degree with a

concentration in Biology specializing in mycology at the University of Puerto Rico,

Mayagüez Campus. She was recognized as the first graduate student in 1987 who did her

research on a topic of Chemical Engineering still a student of the department of Biology.

Elsie's research was directed to pharmaceutical biotechnology for the production of

benzylpenicillin for cell immobilization. She has 23 years of experience managing quality

control in a microbiology laboratory, manufacturing processes following FDA guidelines,

pharmacopoeias, government guidelines for parenteral drugs, including oral and semisolid

products. Her primary responsibility has been conducting audits and evaluating the

microbiological criteria for acceptance in pharmaceutical preparations by microbiological

testing, water analysis for pharmaceutical purposes, monitoring of controlled

environments, qualification, and validation processes to achieve compliance with the

acceptance criteria for finished products.

Ms. Hernández is a senior consultant in the area of microbiology in

pharmaceuticals and manufacturing, primarily doing consulting work in the area of

microbiological laboratories, manufacturing, and training in technical and regulatory

guidelines. She has had taught at various universities in Puerto Rico, including the

University of Puerto Rico, the Pontifical Catholic University of Puerto Rico, and at the Ana

G. Méndez University System. She has taught courses related to biotechnology,

pharmaceutical, and biopharmaceutical manufacturing, including manufacturing

biotechnology processes, validation processes, GMP’s and regulations, industrial

microbiology, mycology, genetics, microbiology, molecular biology, and general biology.

ix

Dedications

To God, for giving me the wisdom and the knowledge that was by my side every

step of this huge project.

To my greatest love, my daughter Ninotchka, my biggest inspiration, who gave

me the strength to transcend any adversity.

To my parents, Dr. Enrique Hernández Santos and Prof. Elsie D. García Lugo,

for teaching me what it was to have passion for learning and the faith that the reward

will always be to enjoy the harvest of hard work.

To all my friends and family who supported me in any way and were part of this

journey, I dedicate this disertation.

x

Acknowledgments

The goal of completing a doctoral degree was a major endeavor requiring passion

for education, the wisdom to understand the effort and dedication to succeed, and the

tenacity to achieve the degree. This goal has been possible with the advice and time from

others who have supported me.

I want to express my gratitude to my research advisor, Dr. Adalberto Bosque, for

supporting me in this research project and for his dedication, availability, goodness,

advice, and humanity. Without his direction, it would not have been possible to carry out

this dissertation.

I am grateful to my supervisor Professor, Dr. Teresa Lipsett for her time, and

willingness to guide my steps through the process of the doctoral degree. Thanks a lot, for

their unwavering support throughout. I want to express my appreciation to Dr. Eileen

Villafaňe for providing motivation, time, advice and support. To Dr. Angel L. Rivera, I am

grateful for your time spent; to Dr. Santander Nieto, thank you for being part of my

graduate committee. I thank to Ms. Ana Lliteras for technical molecular support and

Bioanalytical Instruments for supplying essential equipment.

I extend many thanks to Prof. Juan F. Acevedo of the Catholic University, Ponce

facilitating materials and equipment for the conventional part of this research. Thanks to

the Pontifical Catholic University of Arecibo for letting me use the facilities and laboratory

equipment for the experimental phase of this dissertation.

Many thanks to Dr. Dallas Alston of the Pontifical Catholic University of Ponce for

time, advise and dedication spent editing of this manuscript. I thank Dr. Abner Colón for

his guidance, support and time in the statistical analysis in this investigation.

xi

Table of Contents

page

List of Tables ............................................................................................................ iv

List of Figures ........................................................................................................... vii

Chapter One. Introduction ....................................................................................... 1

1.1. Nature of the Problem ...................................................................................... 4

1.2. Problem Justification ........................................................................................ 8

1.3. Research Goals and Objectives ....................................................................... 10

1.3.1. Goals .................................................................................................. 10

1.3.1.1. Environmental Science Goal ................................................ 10

1.3.1.2. Pharmaceuticals Industry Goal ............................................. 10

1.3.2. Specific Objectives .............................................................................. 11

Chapter Two. Literature Review ............................................................................... 13

2.1. Background ....................................................................................................... 13

2.2. Health Risks from Objectionable Organisms ...................................................... 34

2.2.1. From GMP’s Perspective .................................................................... 37

2.2.2. Non-compliance of Product ................................................................. 40

2.2.3. From the United States Phamacopoeia (USP) Perspective ................. 46

2.3. Contribution to Procedures, Costs and Benefits for Pharmaceuticals

Industries .......................................................................................................... 48

2.4. Advantages and Disadvantages of the Techniques Described in the

Pharmacopeia vs. Molecular Method ................................................................ 58

xii

page

2.4.1. USP 38-NF 33 <Chapter <61> Microbiological Examination of

Non-sterile Product: Test of Microbial Enumeration ........................... 59

2.4.1.1. Preparation of the Sample .................................................... 59

2.4.1.2. Total Aerobic Microorganisms Count (TAMC) ....................... 60

2.4.1.3. USP Results Interpretation ................................................... 62

2.5. USP 38-NF33 Chapter <62> Microbiological Examination of Non-sterile

Products: Tests for Specified Microorganisms ................................................... 63

2.5.1. Product Analysis ................................................................................. 63

2.6. Advantages and Disadvantages of the Techniques Described by USP 38-NF

33<61> .............................................................................................................. 65

2.7. Applications Present and Future ........................................................................ 68

2.8. Advantage of the Real-time PCR ....................................................................... 69

2.8.1. Options of the Real-time PCR Equipment’s ......................................... 70

2.8.2. Advantages of the Real-time PCR Technique ..................................... 70

2.9. Real-time PCR Comparison versus Microbiological Methods ............................ 72

2.10. Taxonomic change of Aspergillus niger to Aspergillus brasiliensis ................... 73

2.10.1. Morphologic Characteristics of Genus Aspergillus ............................. 73

2.10.2. Aspergillus: Importance in Industry, Agriculture and Medicine ........... 77

2.10.3. Taxonomy: Approaches to Distinguish A. niger from A. brasiliensis .. 78

2.10.4. Reclassification of Strain ATCC®16404™ from Aspergillus niger to

Aspergillus brasiliensis ...................................................................... 79

Chapter Three. Materials and Methods..................................................................... 85

3.1. Microbiological Analysis ..................................................................................... 85

3.1.1. Identification Method ........................................................................... 85

xiii

page

3.1.2. Negative Control ................................................................................. 86

3.1.3. Growth Promotion Test ....................................................................... 86

3.1.4. Suitability of the Counting Method in Presence of Product .................. 86

3.1.5. Specified Microorganisms ................................................................... 87

3.1.6. Preparation of Test Strains .................................................................. 87

3.1.7. Inoculum Preparation-determination of the Population Known

to Contaminate Pharmaceutical Sample ............................................. 88

3.1.8. Preparation of the Pharmaceutical Sample ......................................... 89

3.1.9. TBC and TYMC Test: Plate Method .................................................... 92

3.1.10. Identification of the Preparation of the Inoculum ................................ 92

3.2. Molecular Method Real-time PCR...................................................................... 93

3.2.1. Preparing a PCR Reagent ................................................................... 94

3.2.2. Preparation and Extraction Samples for Escherichia coli ATCC®

8739™ and Apergillus. brasiliensis ATCC® 16404™ Testing ............. 96

3.2.3. Real-time PCR Amplification Process ................................................. 98

4.0. Chapter Four: Results and Discussion ............................................................... 101

4.1. Data Analysis .................................................................................................... 122

4.1.1. Inferential Data Analysis ...................................................................... 122

4.1.2. Inferential Analysis Methods ............................................................... 124

5.0. Chapter Five: Conclusions and Recommendations ........................................... 126

Literature Cited ......................................................................................................... 128

xiv

List of Tables

page

Table 2.01. Major Risk for Non-GMP Compliance .................................... 39

Table 2.02. Product Recall Definition ........................................................ 41

Table 2.03. Products Withdrawn from Market in 2013 for Microbial

Contamination……………………………………………………..43

Table 2.04. CDC and FDA Laboratory-confirmed Organisms from

Product Samples…………………………………………………..44

Table 2.05. Summary of Organisms that are More Frequently a Health

Threat .................................................................................... 48

Table 2.06. Advantages and Disadvantages of Culture and Molecular

Methods for Screening of MRSA ............................................ 49

Table 2.07. Contribution and Benefits to SOPs ......................................... 53

Table 2.08. Generic and Practical Approach to Microbial Measurements

Methods ................................................................................. 56

Table 2.09. Contribution in Terms of Cost for the Pharmaceutical

Industry .................................................................................. 57

Table 2.10. Costs of Molecular Reactives Translated to Costs per

Reaction ................................................................................ 58

Table 2.11. Products Examination Techniques ......................................... 61

Table 2.12. Test for the Absence of Specified Microorganisms ................ 64

Table 2.13. Advantages and Disadvantages of the Techniques

Described in the USP38 <61> ................................................ 67

Table 2.14. Advantages and Disadvantages of the Real-time PCR .......... 71

xv

page

Table 2.15. Advantages and Disadvantages of Conventional

Microbiology Methods ............................................................ 73

Table 2.16. The Difference in ITS Sequences .......................................... 80

Table 3.01. Selected dilutions from Test for Specified Microorganisms

(TSM) .................................................................................... 91

Table 3.02. Amount of Samples per Objectionable Organisms ................. 92

Table 3.03. Preparation of the Reaction Mix ............................................. 95

Table 3.04. Primers and Probe per Organisms ......................................... 96

Table 3.05. Steps Performed to extract sample with PrepMan ................. 97

Table 3.06. Preparation of the 48 Wells Plate ........................................... 99

Table 3.07. Reaction of Real-time PCR Quantities of Reagents by

Reaction ................................................................................ 99

Table 3.08. Quantities for Reaction .......................................................... 100

Table 4.01. Sensitivity Test for minimal Detection of Eschericha coli

ATCC® 8739™ by Conventional Pour-plate Method ............. 103

Table 4.02. Confirmatory Test for E. coli ATCC® 8739™ Identification

Detected in USP Conventional Pour-plate and Real-time

PCR Methods ........................................................................ 104

Table 4.03. Confirmatory Test for A. brasiliensis ATCC® 16404™

Identification Detected in USP Conventional Pour-plate and

Real-time PCR Methods ........................................................ 106

xvi

Table 4.04. Sensitivity test for minimal detection of

Aspergillus brasiliensis ATCC® 16604™ by Conventional

Pour-plate Method ................................................................. 109

Table 4.05. Real-time PCR Treshold Cycle (Ct) Values of E. coli at

minimum detection (Sensitivity Test) ...................................... 111

Table 4.06. Real-time PCR Ct Values of A. brasiliensis ATCC® 16404™ at

Minimum Detection Sensitivity Tests ...................................... 115

Table 4.07. Live and Dead Cells of Escherichia coli ATCC® 8739™ with

Cellometer®Auto M10 ............................................................ 116

Table 4.08. Live and Dead Cells of Aspergillus brasiliensis ATCC®

16404™ with Cellometer®Auto M10 ...................................... 117

Table 4.09. Presence in CFU, Cells and Ct Value .................................... 119

Table 4.10. Test for Specified Microorganisms Detection by USP

Conventional Pour-plate and Real-time PCR141 ................... 121

Table 4.11. Mann-Whitney test for Detection of Escherichia coli

ATCC®8739™ and Aspergillus brasiliensis ATCC®16404™

for Each of the Two Different Methods ................................... 122

Table 4.12. Mann-Whitney Test for Real-time PCR .................................. 123

Table 4.13. Mann-Whitney test ................................................................. 123

Table 4.14. Frequiencies .......................................................................... 124

Table 4.15. Tests Statistic ........................................................................ 125

xvii

List of Figures

page

Figure 2.01. Algorithm for the Identification of Nonfermenting

Gram-negative Bacilli ............................................................. 27

Figure 2.02. Causes for Non-sterile Recalls ............................................... 36

Figure 2.03. Morphological Structures of the Genus Aspergillus A-B:

conidiophores C-D: conidial heads ........................................ 74

Figure 2.04. Aspergillus brasiliensis sp. nov. CBS 101740T ...................... 75

Figure 2.05. Colony Morphologies of Type Strains of Species assigned

to Aspergillus section Nigri ..................................................... 76

Figure 2.06. A Neighbor Joining Tree of Black Aspergilli Based on

Their ITS DNA Sequences ..................................................... 81

Figure 2.07. The sequence data for A. brasiliensis ATCC®16404™ was

18S ribosomal RNA gene, partial sequence ........................... 82

Figure 3.01. Computer Screen Template Real-time PCR Run Method

Parameters ............................................................................ 99

Figure 3.02. Real-time PCR Protocol Used-Reconstitution of the Forward

and Reverse Primers to 10,000 pmol ..................................... 100

Figure 3.03. Protocol Used for Dilutions of the TaqMan® Probe to

100µM in 50µL ....................................................................... 101

Figure 4.01. Sensitivity Test for Minimal Detection of E. coli ATCC®8739™

the CFU Conventional Method ............................................... 102

Figure 4.02. A. brasiliensis Results in CFU for Sensitivity Test by

Conventional Pour-plate Method ............................................ 108

Figure 4.03. Amplification Plot for a 10 fold Dilution for

E.coli ATCC®8739™ and A. brasiliensis ATCC®16404™ ..... 113

1

Chapter One

Introduction

Microbiology laboratories in the pharmaceutical industries conduct the

identification of objectionable organisms with conventional cultivation methods, selective

and differential media, phenotypical characteristics, and biochemical tests as traditional

manual methods. Today’s microbiological quality control laboratory still uses fundamental

tools that were developed centuries ago. It was interesting that such instruments as

autoclaves, agar plates, broth tubes, erlenmeyer flasks, incubators, inoculation loops, and

microscopes are popular when many new detection and enumeration devices provide

alternative and rapid microbiological methods.

Traditional instruments are being compared to alternative or rapid microbiology

systems in terms of functionality and sensitivity (Denoya 2014). The microbiological test

methods can be grouped into three broad categories based on their function. They are (1)

detection of the presence or absence of microorganisms in a test sample; (2) enumeration

of microorganisms present in a sample; and (3) identification of microorganisms either

present in a test sample or from a pure culture isolated from a test sample (Madsen 2001).

The term most commonly used for the conventional microbial population counting was

“Colony Forming Unit” (CFU), which was an artifact-based count relying on cellular

replication to produce a visible colonies of cells on growth medium. If the physical

conditions or growth medium are not suitable, no colonies appear. Also, if a clump of many

cells lands in one place and only a single colony forms, then the count of “one” would

underestimate the total.

Therefore, plate counts are not always accurate or precise because they involve a

lot of manipulation of the analyst. The microbiological tests represent a small portion of a

2

pharmaceutical quality testing program and although their importance was critical

to pharmaceutical product safety. New technology is needed for microbiological methods

that offer advantages of speed and precision for solving microbiological problems

associated with materials, excipients or environmental factors during manufacturing

proceses or samples analyses .

Many new methods use technologies developed for aerospace research, clinical

studies, and the food industry (Hussong & Mello 2006). While it may seem odd that the

pharmaceutical industry lags behind in implementing new microbiological technologies,

the resistance to change was reinforced, in part, by regulatory pressure and the complexity

of assays. Automated methods are among the most frequently used miniaturized systems,

including API systems Inc., Enterotube™, Minitek®, Crystal™ ID system, Micro-ID®, RaID

systems, Biolog Microbial ID system and Vitek®. The most successful and sophisticated

miniaturized automated identification system include the Vitek® system (bioMérieux)

(Fung 2002). These standard miniaturized tests’ method are time-consuming, not specific,

and delay the release of the product. These automated systems do not provide information

about identification characteristics of many of the isolated organisms in their computerized

information system. As a result, many isolated organisms cannot be identified. These

automated systems are limited in the identification of specific fungi, which was the reason

why industries regularly use external contractors for the identification (Matsuda et al.

2007).

In recent years, the focus of multiple investigations of the pharmaceutical

microbiology has been toward improving isolation and early detection of pathogenic

organisms, including their characterization and enumeration from various environmental

and products origin. Each of these methods falls into the classification of rapid

microbiological methods (RMM). These approaches are categorized according to the type

3

of technology used, including microorganism growth, organism viability, presence of the

microorganism, cellular component or “artifact” effect, methods of nucleic acid, traditional

methods combined with computerized image assistance, and combinations of several

methods (Clontz 2009). These strategies serve as the basis for determining the identity of

a microorganism as part of an investigation of product failure by not complying with the

specifications for environmental or product sample.

Recent microbiological detection technologies frequently require fewer microbial

cells to obtain results in less time than those obtained with traditional methods. Some of

the technologies are more accurate and informative data related to the potential

contaminant than the information obtained using conventional assays (Saghee et al.

2010). The evolution of microbiological testing from classical procedures to RMM will

provide new and increased data to assist in rapid decision-making to facilitate real-time

release or to complete on time with the determination of the root-cause analysis for failure

investigations, thus improving product safety. With the successful use of these

technologies, the quality control microbiology laboratories incorporate these new tools to

provide product and process knowledge to improve their quality goals (Duguid et al. 2011).

The analyses according, to United States Pharmacopoeia (USP) chapter <62>

Microbiological Examination of Non-Sterile Products: Test for Specified Microorganisms

(TSM), must comply with the absence of: Staphylococcus aureus, Pseudomonas

aeruginosa, E. coli, Salmonella spp., and Candida albicans for non-sterile products and

raw materials (USP38-NF33. 2015). This research study is based on the detection of

Escherichia coli ATCC®8739™ (bacteria) and Aspergillus brasiliensis (fungus).

Aspergillus brasiliensis ATCC®16604™ for the experimental phase in this investigation

was chosed from Chapter <61> Microbiological Examinations of Nonsterile Products:

Microbial Enumeration Test (MET). In accordance to these requirements, this study was

presented as a possible alternative to accurately detect the presence and genetic activity

4

of these objectionable organisms in pharmaceutical product samples. The Real-time PCR

technique was accurate, effective, simple, and cost-effective for the analyses, not solely

for bacteria, but also for fungi and yeasts required by the USP chapter <62>: Test for

Specied Microorganisms (TSM). The fundamental objective of the investigation was

detection, sensitivity, and precision in terms of presence or absence of objectionable

organisms through the utilization of molecular methods (Real-time PCR) by comparing

conventional to modern identification techniques to identify E. coli ATCC® 8739™ (USP38-

NF33 Chapter <62>. 2015) and A. brasiliensis ATCC®16404™ for the United States

Pharmacopeia (USP) <61> by using the molecular technique Real-time PCR. The

conventional microbiological methods, including long-established techniques described in

Europe, Japan, and the United States pharmacopoeia have benefited microbiologists

during the past century and have helped to ensure the production of microbiologically safe

products (Sandle 2014). For example, a wide range of microbiological techniques have

verified by using plate-count methods in the enumeration and identification of

microorganisms within an acceptable margin of error according to conventional

methodology and type of product (Sutton 2011).

1.1. Nature of the Problem

Pharmaceuticals industries conduct lab tests for the compliance of product quality

in terms of purity, safety, and efficacy (GMP 2015). For compliance purposes, most

microbiological methodologies analyses rely on traditional techniques (USP38-NF33

2015). The USP are standards that describe minimal expectations for the quality of

finished products. If a pharmaceutical product does not meet requirements of the

appropriate USP monograph, it was designated “mislabeled” or “adulterated”, based on

provisions by the Federal Food Drug, and Cosmetic Act, in which case the Food and Drug

Administration (FDA) can take enforcement actions (Sutton et al. 2011)

5

The type of analyses to confirm the presence or absence of specific organisms in

a pharmaceutical sample is critical to achieve the approval of the product to be used by

the consumer. The role of USP, a nongovernmental, not-for-profit organization, was to

develop microbiological public standards that guarantee the uniformity of products from

batch to batch, as well as the microbiological quality of the products (Sutton et al. 2001).

The test for specified microorganisms (TSM) was performed in the pharmaceutical

industries to guarantee that the organisms considered objectionable were not present,

either in the raw material or in the finished product. The microorganisms identified as

objectionable organisms by the FDA must not be present in the pharmaceutical

preparations that may pose a risk to consumer health (Jiménez 2007).

In this type of test, TSM was the predominant practice among microbiological

methods for the cultivation of microorganisms, phenotypic observation of the colony, and

biochemical tests to determine its final identification. These time-consuming tests were

not specific, and lack accuracy and precision to show the entire content of the presence

or absence of these specific organisms in a sample. Time-consuming tests delay final

approval of the pharmaceutical product (Ragheb et al. 2012). The TSM was required

within the pharmaceutical industries to ensure that objectionable organisms were not

present in the raw material, nor in the finished product. To avoid consumer-health risks,

microorganisms identified as objectionable by the FDA should not be present in

pharmaceutical preparations (Jiménez 2007).

Conventional microbiological methods are labor-intensive and time-consuming

with lengthy incubation periods, resulting in delaying the release of products. Ragheb et

al. (2012) tested and validated a polymerase chain reaction identification approach to

detect the following indicator bacteria: E. coli, P. aeruginosa, Salmonella spp., and

Staphylococcus aureus in pharmaceutical preparations. Their method depended on

amplification of specific conserved genes located in four bacteria. The method could be

6

performed individually or collectively to detect indicator bacteria in a single reaction in

different forms of pharmaceutical products. The method provides a high-performance

screening method to test different pharmaceutical preparations to detect specific

microorganism contamination (Ragheb et al. 2012).

The quality test compliance of the raw material and the finished product depends

on the results obtained in terms of detection, precision, and sensitivity to the entire process

of pharmaceutical manufacture. Certain standardized tests were carried out in

microbiological testing laboratories in each pharmaceutical production facility to ensure

quality according to standard pharmacopeia recommendations. It was essential that

nonsterile products were free of specified microorganisms. For example, topical

preparations must be free of P. aeruginosa and S. aureus, and oral preparations without

Salmonella spp. and E coli. The existent conventional methodologies analyze the limits of

microbial growth in a sample of raw material and finished product, as described by the

USP (USP38-NF33 2015). Rapid release of samples optimizes manufacturing, product

testing, and release, thus permitting high throughput, with simultaneous analysis of

pharmaceutical formulations (Jiménez 2001). Conventional microbiological methods

generate-during and at the end of each analysis-significant quantities of biological waste,

thus increasing the risk of biological hazards as compared to the Real-time PCR

methodology which generates less biological waste. The detection-level in the Real-time

PCR test is in nanograms of the concentration of sample DNA or this methodology can

used to detect specific organisms. Identification of bacteria and fungi using traditional

techniques requires a lot of time, effort, excessive expense of materials, excess of

biological waste, and experienced analysts in bacterial and mycological identification.

A sensitive rRNA targeted reverse transcription quantitative PCR method was

developed for exact and sensitive enumeration of subdominant bacterial populations using

specific primers for 16S or 23S rRNA, with corresponding analytical curves for E. coli,

7

Enterococcus faecalis, S. aureus, Clostridium perfringes and P. aeruginosa. The number

of cultured bacteria was determined by Real-time -qPCR; the results correlated with the

CFU (Colony Forming Unit) count over the range from 10˚ to 10¯5 CFU. The bacteria

counts obtained by Real-time -qPCR methodology were the same as the CFU count,

regardless of the in vitro growth phase, except for C. perfringes during starvation periods.

The viable cell counts obtained were in agreement with Real-time -qPCR counts rather

than with the CFU counts (Matsuda et al. 2007).

During the manufacturing process of a pharmaceutical product, microbiological

contamination can originate from raw material or can be introduced during manufacture

by means of contaminated equipment, operators, from the air, or from packaging material

(GMP - 21CFR211 2013). It can also be originated during the storage and handling of

material. The majority of the raw material (including water) used in the pharmaceutical

industry may contain several types of polluting organisms. The microbial growth potential

of a pharmaceutical drug product is the potential of microorganisms to survive and

proliferate within the product. Each drug formulation can support or inhibit microbial

growth, depending on its components (Lolas and Metcalfe. 2011). Depending on the

manufacturing process, microbial contaminants can be reduced or eliminated (Clontz

2009). The microbiological quality of the pharmaceutical excipients used to manufacture

pharmaceutical and over-the-counter (OTC) drug products may significantly affect the

outcome of individual processing steps and the microbiological attributes of the final drug

products (Cundell 2006). Today’s microbiological quality control laboratory still uses

fundamental tools such as agar plates, autoclaves, broth tubes, Erlenmeyer flasks,

incubators, inoculation loops, and microscopes. However, many new detection and

enumeration devices were applicable in alternative and rapid microbiological methods.

Traditional instruments will be compared to alternative or rapid microbiology systems in

terms of functionality and sensitivity.

8

The modern pharmaceutical industry must use rigorous and sensitive methods of

microbial identification to detect pathogenic organisms in a minimal sample size.

Molecular technologies use genetic sequences to rapidly detect microbial contamination,

provide an accurate microorganism identification in the pharmaceutical environment, and

improve efficiency (Jiménez 2011). Analyses must provide information assuring the

absence of specified or objectionable organisms belonging to the pharmacopoeia

(USP38-NF33 2015). The objectionable organisms stipulated in the USP Chapter <62>

are E. coli, Salmonella spp., S. aureus, P. aeruginosa, Clostridium spp. and C. albicans.

The indicator organisms used for the suitability test of the enumeration method in the

presence of product are S. aureus, P. aeruginosa, B. subtilis, C. albicans and A.

brasiliensis (Chapter <61> USP38-NF33 2015).

1.2. Problem Justification

The purpose of this investigation work was to evaluate the detection of E. coli

ATCC® 8739™ and A. brasiliensis ATCC® 16404™ in OTC pharmaceutical preparations

using the Real-time PCR methodology as a viable alternative so that the pharmaceutical

industries will be confident to use this methodology for routine analyses systems in quality

control laboratories. This investigation will evaluate that Real-time PCR analyses are

precise and rapid for the detection and identification of E. coli ATCC ® 8739™ and A.

brasiliensis ATCC ® 16404™. For instance, this method only required thirty-minutes to

analyze each sample, which was not possible using conventional microbiological methods

that take from 24 to 48 hr for bacteria and from 5 to 7 days for fungi. Decreasing analysis-

time to detect contaminant microorganisms in raw material, excipients, environmental

monitoring, and finished products will accelerate the final approval of Real-time PCR

methodology. This research focus on the following three types of products of finished

product: solids, liquids, and cream, which will be analyzed strictly according to the criteria

established in the chapter <61> and <62> of the Compendium of the USP38-NF33 (2015).

9

The hypothesis therefore was that the molecular methods of Real-time PCR

provides the detection of minimal traces of objectionable organisms present in the

samples, contrary to standard conventional microbiological method that measure the

presence of viable organisms that could be counted when reading test results. The

standard conventional microbiological methods are the standard methodologies that

involves the growth of colonies on a nutrient agar surface during a specific period of

incubation (Postagate 1969). In the pharmaceutical industry, detection and quantification

of viable cells of well-characterized species are important for quality control purposes.

Determination of microbial viability by the plate count method was routine in microbiology

laboratories worldwide (Davey 2011).

The technique Real-time PCR was expected to evaluate how the physiological

capabilities of these organisms can mask the detection of these objectionable organisms.

The use of the molecular technique Real-time PCR was projected as a molecular method

with a high potential to detect the presence and activity of living cells of objectionable

organisms in a pharmaceutical sample.

This research proposed to evaluate if the Real-time PCR technique was more

sensitive, accurate, and precise in the detection of E. coli ATCC®8739™ and A.

brasiliensis ATCC®16404™ in the TSM test, as compared to the conventional method of

identification. A secondary objective was to evaluate the cost effectiveness of this

technique compared to conventional methods in terms of preparation time, obtaining

results, reproducible results, and ease of use. The goal of this study was to compare the

conventional USP techniques of detection and identification for E. coli ATCC® 8739™ and

A. brasiliensis ATCC® 16404™ using the TSM pour-plate method with the molecular

technique of Real-time PCR.

1.3. Research Goals and Objectives

1.3.1. Goals

10

1.3.1.1. Environmental Science Goal

The goal of this study was to contribute with an alternative to the analysis of raw

materials and finished products for the pharmaceutical industries with a capacity to detect

minimal concentrations of objectionable organisms contamination in their sample. This

helps to reduce the risk of exposure of humans and animals to contaminated drugs that

do not meet the FDA and USP quality standards. The early and rapid detection of a

precise and effective methodology as the Real-time PCR helps in protecting public health

by avoiding outbreak of nosocomial infections.

1.3.1.2. Pharmaceuticals Industry Goal

The goal of this investigation was to evaluate that the Real-time PCR methodology

as a precise, rapid, and specific technique to detect objectionable and specified organisms

for the TSM test, as compared to conventional microbiological current methods specified

in chapter <62> of the USP (2014). It evaluate its advantages in molecular microbiological

analyses for pharmaceutical industries versus the conventional methodologies,

specifically in terms of precision, specificity, and test duration. It compare the cellular

viability of the inoculum analysis used for both skills (molecular and conventional pour-

plate methods) in terms of detection of DNA target (Ct value) versus CFU/mL. It evaluate

that results obtained from this investigation can be used by the pharmaceutical industries

to validate the Real-time PCR methodology as an alternative to TSM methods to

determine the presence, quantity of cells, and genetic activity of the objectionable

organisms, including S. aureus, P. aeruginosa, E. coli, Clostridia spp., Salmonella spp.

and C. albicans (USP38-NF33 2015). The cost effectiveness of the Real-time PCR vs.

USP pour-plate methods will be compared among test duration, risk of biological

exposure, materials, and equipment during the execution of the methods.

11

This investigation directly impacts the conventional methodology established by

the USP (2015), thus offering the Real-time PCR methodology as a viable alternative

microbiological laboratory technique suitable for the pharmaceutical industry.

1.3.2. Specific Objectives

This study will use the molecular technique Real-time PCR for the identification of

bacteria and fungi to compare its effectiveness, precision (reproducibility), and rapid

analysis to conventional techniques used to culture and identify these organisms. The

Real-time PCR, which involves a single-step based on the sequencing of the conserved

DNA to target specific microorganisms, also will be compared to data provided from

conventional techniques. The methodology performs a millions of copies of the target DNA

of Escherichia coli ATCC® 8739™ and Aspergillus brasiliensis ATCC® 16404™ in 30 min

at real time in this study.

This investigation outlines the following five principal targets.

Compare the minimum capacity of the method to detect the contaminant in

the pharmaceutical sample so that the output of the product to market is

prevented avoiding risk to public health .

Compare the methodology of microbiological conventional analysis for the

TSM test with the molecular methodology of Real-time PCR in terms of

sensitivity, precision, accuracy, specificity, reproducibility, and test duration

to detect objectionable or specified organisms.

Evaluate the Real-time PCR possesses high sensitivity in the detection of

minimal traces of pharmaceutical product, as compared to the USP

conventional method pour-plate culture techniques.

12

Compare the genetic activity for the minimal trace of DNA at the molecular

level versus CFU/mL using the conventional microbiological pour-plate

method.

Evaluate the Real-time PCR method can identify objectionable bacteria

and fungi in reference to time invested in the analysis, amount of materials

used for preparation, analysis and biological waste.

13

Chapter Two

Literature Review

2.1. Background

Microbial contamination is still one of the major causes for global product recalls,

in particular in developing tropical countries (Okeke et al. 2001; Jimenez 2007). Therefore,

it was important to improve the preservative system and refine non-invasive packaging

to inhibit the growth of contaminating microorganisms during manufacturing, storage and

use by consumers (Farrington et al. 1994; Linter and Genet 1998). Contamination leads

to product degradation or, if it was contaminated with pathogens, allows the product to act

as a fomite to potentially spread infection to susceptible users (Brannan and Dille 1990).

Contamination by microorganisms can cause infections after the organisms are dead and

thus are harder to detect. Consequently, the presence of objectionable microorganisms in

non-sterile products was indicative of the absence control protocols (Jiménez 2001).

Pharmaceutical products are prone to microbial contamination at every stage

during their manufacture. Identification of microbial contaminants in product recalls and

environmental samples provides important information on the possible contamination

sources and distribution of microbial species in pharmaceutical environments (Jiménez

2007). The presence of some microorganisms in non-sterile preparations can reduce or

inactivate the therapeutic activity of a pharmaceutical product, thereby adversely affecting

patients’ health (cGMP 2014).

Current rapid method technologies can detect the presence of diverse types of

microorganisms or specific microbial species, enumerate the number of sample

microorganisms, and identify microbial cultures to the genus, species, and sub-species

levels. Each microorganism detected, quantified, or identified was dependent on the

specific technologies and instrumentations employed (Miller 2005). Pharmaceutical

14

industries for non-sterile products must reduce the microbiological load in its raw materials

and final product.

The acceptance criteria established for non-sterile products was based on the

number of total aerobic microbial count (TAMC) and the combination of the amount of total

yeast and mold (TYMC) (USP38 <1111> 2015). These specifications vary according to

the dosage form and use of the product. The dosage forms of a pharmaceutical product

include tablet, capsule, liquid, and ointment/cream. For purposes of microbiological

laboratory testing, the products have been divided into four categories (PMM. 2014):

Category 1 – Injections and other parenteral including otic products, emulsions,

sterile nasal products, and ophthalmic products made with aqueous bases or

vehicles.

Category 2 – Topical products made with aqueous bases or vehicles,

non-sterile nasal products, and emulsions, including emulsions applied to mucous

membranes.

Category 3 – Oral products other than antacids that are made with aqueous bases

or vehicles.

Category 4 – Antacids that are made with aqueous bases or vehicles.

The analysis method known as the TSM (Test for Specified Microorganisms)

provides the general instructions for the microbiological examination of non-sterile

products (USP38 <62> 2015). This test involves the execution of two basic methods: (1)

tests for the microbiological quantitative enumeration and (2) tests for specified

microorganisms, performed as determined in Chapters <61>, <62> of USP 38-NF33

(2015). The TSM test was primarily designed to determine if a substance or

pharmaceutical preparation complies with the established specifications in terms of

microbiological quality (USP <1227> 2015). The role of United States Pharmacopeia, a

15

nongovernmental, not-for-profit organization, was to develop microbiological public

standards that, along with other requirements, ensure the consistency of products from

batch to batch, as well as assure the microbiological quality of products (Sutton et al.

2001). The TSM describes the quantitative enumeration of bacteria, fungi, and yeast from

non-sterile products that can grow under aerobic conditions. It also describes the

determination of presence or absence of indicators organisms like S. aureus, P.

aeruginosa, E. coli, Salmonella spp., Aspergillus niger, and C. albicans (Clontz 1998). The

TSM test must demonstrate that the specimen analyzed does not inhibit growth and

multiplication of the microorganisms contained in the sample.

The analysis methods described in USP <61>, <62> (2015) are conventional

methodologies that include the transference of cultures, phenotypic colony observation,

and biochemical tests for final identification. The plate method outlined in Chapters <61>

and <62>Microbiological examination of non-sterile product of the US Pharmacopeia

include guidance for assessing total aerobic bioburden in pharmaceutical products. This

comprehensive methodology lacks the flexibility for a variety of situations. It uses a

mathematical model to express the plate-counting procedure as a statistical framework

concerning the total aerobic bioburden. This framework allows the laboratory scientist to

adjust USP <61> and <62> chapters methods to meet specific practical constraints. The

plate method can performed with acceptance criteria using a test-specimen quantity

smaller than the prescribed 10 g or 10 mL (Kai 2004).

Many rapid microbiological method (RMM) technologies provide more sensitive,

accurate, precise, and reproducible test results when compared with conventional, growth-

based methodologies. They may be fully automated, offer increased sample throughput,

operate in a continuous data-collecting mode, reduce duration (e.g., from days or weeks

to hours or minutes), and for some RMM platforms, provide Real-time results (Miller 2012).

16

The pharmaceutical industry utilizes precise and sensitive methods for microbial

identification so that pathogenic or specific organisms can be detected with a minimal

sample size. Results of a sample must demonstrate an absence of indicative or

objectionable organisms which include the following: Salmonella spp., S. aureus, E. coli,

P. aeruginosa, C. albicans, and A. niger. These are organisms that could be harmful by

themselves or by toxins produced by them; they also can cause diseases and infections

in humans (FDA 2014). Objectionable organisms can cause disease or degradation of

product. Other examples are Pseudomonas spp, such as P. putida and P. maltophila.

Opportunistic organisms can cause disease in immune compromised patients, and these

organisms include most of the microbial flora found in pharmaceutical raw materials,

recipients, and the manufacturing environment (Clontz 1998).

The most frequent microbial contaminants of pharmaceutical products and raw

materials are bacteria, yeast, and filamentous fungi. To analyze the microbial content of

pharmaceutical raw materials in finished pharmaceutical products, Martínez et al. 1991

determined the aerobic bacteria, anaerobic bacteria, and fungi. Few or one of pathogenic

microorganisms were found in most analyzed products, but in some materials, especially

those of natural origin, they detected high bacterial and fungal contamination.

Microorganisms of the genus Bacillus were the most frequent aerobic bacteria isolated;

Bifidobacterium and Clostridium were the most common anaerobic bacteria; and

Penicillium and Aspergillus fungi had the highest frequency. Because of their enzymatic

or toxigenic activities, these microorganisms are problematic in pharmaceutical finished

products.

Many of the ingredients used in formulations can become substrates for

microorganisms with optimum conditions, including pH, temperature, and nutrients. Thus,

the pharmaceutical industry should move away from traditional methods of testing of raw

materials and the finished product to a new technological pharmaceutical industrial era.

17

Moving away from the traditional pharmaceutical manufacturing becomes evident when a

retrospective evaluation compares traditional methods to modern technologies. For

instance, many organisms cannot be cultured with traditional methods. However, by using

new technologies, organisms which cannot be cultured can still be related to different

environments. By correlating organisms to different environments or soil, we can correlate

them to the origins of sterile products, non-sterile products, ointments, creams, and

biological products.

Regardless of the classification, it was important to detect the bioburden because

the vast majority of the pharmaceutical preparations have pH values of 6.5-7.5, ideal pH

values to sustain the ample microbial growth. According to the ingredients that compose

each formula, the susceptibility to microbial growth can be determined. Products unable

to suppress the growth of several microorganisms represent a potential health hazard

(Campana et al. 2006; Clontz 2009).

The evolution of the pharmaceutical industry during the last decade was evidenced

in the incorporation of molecular techniques in sensitive analyses to maintain quality

control of the finished product. This incorporation was performed with the principal

objective of obtaining positive and reliable results. Molecular techniques in microbiological

analysis were presented as an ideal alternative for those cases when no visible signs of

contamination were observed, but that certain types of organisms may be present in high

numbers. Physical changes in raw materials and finished product are usually associated

with microbial contamination, changes such as breakdown of emulsions, bio-pellicles,

surface growth, and production of gas, odors, unwanted texture, colors or flavors. In recent

years, several publications have encouraged the application of molecular techniques in

the microbiological assessment of pharmaceuticals. One of these techniques is

polymerase chain reaction (PCR). The successful application of PCR in the

pharmaceutical industry in developing countries was governed by technical factors and

18

regulatory requirements. These components include the development of a PCR laboratory

and the choice of appropriate equipment and reagents, including the presence of well-

trained analysts and the establishment of quality control and quality assurance programs

(Ragheb et al. 2014).

Different methodologies of molecular application have been applied toward the

pharmaceutical industry microbiological laboratories. One of these methods has been the

diagnosis by adenosine tri-phosphate (ATP), bioluminescence, and chain reaction of the

polymerase (PCR). When fitted to the methods of standard analysis, these methods

provide a rapid result regarding the quality control screening of cosmetics, and finished

products (Jiménez 2001).

The rapidity and alternatives for the "screening" in the microbiological analyses will

depend on the alternative techniques, the organisms involved, and the economic factor,

which can determine what methodology appears viable for routine use in the

pharmaceutical environment. These alternative methods can be classified by the

principles on which they are based, such as bioluminescence, DNA techniques,

immunological techniques, instrumental measurement of bacterial metabolism, and

modified conventional methods or other combinations of these techniques. Test kits must

be accurate, easy to use, labor-saving, sensitive, specific, and rapid (24 h or less). They

must also offer the possibility of a low detection limit, computerization, and low investment

and operational costs. (Van der Zee and Huis int’t Veld. 1997).

The pharmaceutical industries use conventional methods of culture techniques for

the enumeration of different populations using the selective culture media following the

ways of isolating pure cultures and use of confirmatory tests (Jiménez 2001). Other

methods used by pharmaceuticals industries for identification are the automated systems,

including BIOLOG, MIDI™, Phoenix, and VITEK® Systems, based principally on

19

phenotypical tests with a database of genus and species, which are limited for bacteria

and much more limited for identification of fungi (Cundell 2006). In research of the clinical

isolates, the sensitivity of the tests was 99.5% for Vitek® and 95.3% for the Phoenix

system. There were no significant differences between the 2 systems in the control strains,

with the Phoenix system obtaining 100% sensitivity. The Vitek® 2 expert systems obtained

seven strains with ESBL-positive tests, but were considered to be incoherent (Treño et al.

2009).

Cundell (2006) suggested that diverse methods and strategies must be evaluated

to characterize and, identify the genus and species of the organisms isolated in different

pharmaceutical industry environments. This reaffirms the need of the pharmaceutical

industry to justify programs of microbial identification and development of identification

techniques. Cundell suggests the evaluation of new, precise methods in microbiological

laboratories, especially for molecular genetics for bacteria and fungi. The conventional

Microbiological tests based on the activity of microbial growth represent a limiting factor in

speed for obtaining the results of quality control.

During the last 30 years, implementation of Good Manufacturing Practices (GMP)

has been the foundation for improving industrial quality control analysis. As part of GMP,

the United States Pharmacopoeia (USP) Microbial Enumeration Test (MET) provides

methods for the determination of total microbial counts for bacteria, yeast, and mold (USP

2015). The detection of microbial contaminants has been traditionally performed using

cultivation based methods (Mestrandrea L.W. (1997); Baird R. (1998). However, new

molecular methods are available that can rapidly detect microorganisms in contaminated

samples.

Chaubron, Martin and Groulon (2006) developed a one-step universal kit for real-

time RT-PCR method (Reverse Transcriptase Polymerase Chain Reaction). This kit detect

20

bacteria, fungi, and yeasts isolated in pharmaceutical preparations, cosmetics, and non-

clinical samples. Using only one step of RT-PCR, it perform a rapid RT-PCR to

simultaneously detect and quantify the presence of RNA of bacteria, fungi, and yeasts by

using fluorescent monitoring during amplification to reduce risk for false positives. The

false positives can caused by the opening of the pipe between the RT and PCR, as a

consequence of possible environmental contamination of the product due to the precedent

reaction of amplification in the laboratory.

To successfully treat a contamination caused by a bacteria or fungus-yeast in a

sterile or non-sterile product from industry, rapid and accurate detection was required.

Bacterial and fungus-yeast detection have traditionally been performed by pure culture

isolation, followed by identification procedures that incorporate biochemical characteristics

requirements and features, knowledge of the specimen source, visible (colony)

microscopic morphology. A rapid diagnostic method of less than 24 hr for detecting

bacteria and fungus-yeast in industrial samples with the equivalent sensitivity as culture

would be a significant improvement over currently used methods (Chaubron et al. 2006).

The majority of edible gelatin sold in Europe was derived from pigskin or extracted

from bovine tissue. Analytical methods are deficient that document the origin of gelatin or

more specifically, the animal species used as raw material sources in the finished product.

Several published species-specific PCR systems were evaluated as potential molecular

methods for determining the origin of the raw material used in making gelatin. A PCR

primer recently substantiated bovine species-specific material in gelatin by targeting the

ATPase 8 subunit gene in bovine mitochondrial DNA. This PCR primer set was enhanced

by both conventional and real-time PCR approaches and an evaluation of confirmed the

high specificity for the adopted primer set in various gelatin matrices of known origin. The

presence of bovine gelatin in pork or fish gelatin can be detected at 0.1 to 0.001%. Thus,

21

these two PCR assays were potential molecular detection tools to routinely detect bovine

gelatin either alone or as an inclusion in gelatin from other species (Tasara et al. 2005).

A multiplex PCR assay was devised and compared with standard conventional

methods for quality evaluation of pharmaceutical raw materials and finished products with

low levels of microbial contamination. Studies (Karanam et al. 2008) of artificially

contaminated with <10 colony forming units of E. coli, S. aureus, P. aeruginosa, and

Salmonella spp. and possibly contaminated samples were incubated for 16 h with different

enrichment media.

The detection limits for artificially contaminated products analyzed by multiplex

PCR was 1 CFU/g; the detection limit for conventional methodologies was >2 CFU/g.

Similarly, when tested with possibly contaminated samples, 35% were detected for E. coli,

Salmonella spp., S. aureus and P. aeruginosa with multiplex PCR, while only 21% were

detected with standard conventional microbial methods. Thus, multiplex PCR test

provides sensitive and reliable results and allows for the cost-effective detection of all four

bacterial pathogens in a single reaction tube. Karami et al. also conducted a rapid and

definite diagnosis of Salmonella enteritis using an ultra-rapid multiplex polymerase chain

reaction (PCR) detection method for major Salmonella serotypes, such as Salmonella

Typhi, Salmonella Typhimurium and Salmonella Havana. The results showed that all

reference and clinical isolates of Salmonella serovars Typhi and Paratyphi were accurately

identified by this assay. Specificity analysis revealed no cross reactions with other

Enterobacterial strains. Sensitivity of PCR and multiplex PCR assays was 1-10 cells.

Multiplex PCR preparation from sample to final result was 45-50 min (2006).

Abee and Wouters designed a method for the detection and identification of

microbial contaminants in pharmaceutical products, environments of pharmaceutical

production, cosmetics, and food (1999). This method was based on the selective

amplification for quantitative PCR “target” sequence of specific cDNA, particularly in

22

detecting the rRNA sequence of microbial contaminates. The benefits of this method was

the advantage of the conversion of the sequence of RNA in cDNA route with only one

step of reverse transcription and subsequently used in a PCR, in particular in TaqMan® X

PCR.

Schabereiter et al. developed a new test for the detection and differentiation of

eleven (11) species of Aspergillus spp. and Candida spp. in clinical specimens using the

molecular proficiency of RT-PCR. The detection of these organisms were collected from

patients for 33 clinical samples irrespective of suspecting the presence of infection by

fungi; the samples were analyzed using standard culture methods and by molecular real-

time-PCR. The RT-PCR not only detected fungi growing in samples, but also detected

Candida spp. as C. albicans and C. glabarata, as opposed to the conventional method

which could not detect Candida spp.(2006).

Klingspor and Jalal (2006), reported that 5-6 hr were required for the identification

of Candida spp. and Aspergillus spp. using the Rt Light Cycler® method. The

oligonucleotide primers and probes used for species identification were derived from DNA

sequences of the 18S rRNA genes of various fungal pathogens. Each sample was

screened for Aspergillus and Candida to the genus level in the real-time PCR assay. The

assay detected and identified most of the clinically relevant Aspergillus and Candida spp.

at 2 CFU/mL blood. Amplification was 100% specific for all Aspergillus and Candida spp.

tested. The use of RT-PCR in this study demonstrated sensitivity for the detection of the

DNA of the fungi in blood samples, fluids, and samples of biopsy within 6 hr. and identified

the majority of Candida spp. Also, the Real-time PCR assay allows sensitive and specific

detection and identification of fungal pathogens in vitro and in vivo.

Another method of Reverse Transcription-quantitative PCR (RT-qPCR) used by

Matsuda et al. (2007) focused on rRNAs as the target for precise and sensitive

23

quantification of commensal subdominant bacterial populations because rRNA was a

universal constituent of bacterial ribosomes and high copy numbers (103 to 104 molecules

per actively growing cell) are present as housekeeping genes, those required for basic

cellular function.

Targeting these molecules has the potential to increase the detection sensitivity,

as compared to the sensitivity of assays based on detection of a single or multiple copies

of genomic sequences. In this research the sensitive quantification of bacterial populations

with lower detection limits of 103 cells/g of feces and 100 cells/mL of peripheral blood

were detected with RT-qPCR by targeting rRNA, which has similar sensitivity to

conventional culture methods, but improved performance time. They demonstrated that

only 5 hr. was needed for RT-qPCR quantification, and suggested that rRNA-targeted RT-

qPCR assays provide a sensitive and convenient system for quantification of commensal

bacteria and for examining their possible invasion of a host.

Samadi et al. (2007) compared the conventional microbiological method currently

performed in pharmaceutical industry laboratories with the polymerase chain reaction

method. They suggested that the conventional methodology for detection of microbial

contamination in pharmaceutical products was generally based on culture by selective

media, microscopic examination of the suspected colonies, and biochemical tests, each

which was time-consuming and non-specific. In recent years attempts have been focused

on specific, sensitive and rapid methods because available documents are still insufficient

to integrate rapid methods in identification of microbial contaminations.

The study was directed towards the evaluation of a simple DNA lysis protocol,

coupled with a rapid PCR system for detection of low numbers of S. aureus in

pharmaceutical products. In the PCR assay of the experiment, the distinctive 241base-

pair (bp) fragments were obtained with universal primers and bacterial DNA templates,

thus indicating the efficiency of the extraction method and primers. The amplification of

24

the 108 bp fragment with specific primers using a DNA S. aureus template and negative

results with other staphylococcal species proved the specificity of the assay. The minimum

detection limit (MDL) of S. aureus in pharmaceutical samples was 102 CFU/mL when

using 10 μL of the sample lysate without any prior enrichment . Consequently, this level

of detection has not been previously reported in pharmaceutical samples.

The PCR disadvantage lack of discrimination between viable and non-viable cells

solve by; the researchers, integrating a preenrichment step into their study. After a 24 hr

preenrichment, the number of cells increased and the MDL was lowered to 1 CFU/mL.

The positive PCR results obtained for the bacterial concentration of about 101 and 100

CFU/mL and intensification of other amplicon following enrichment were expected,

because bacteria could multiply exponentially within 24 hr. Standard identification

methods usually require several days; with quick lysis, specific primers, and PCR protocol,

S. aureus contaminations of about 1-2 CFU/mL in pharmaceutical products were identified

to the species level. This study indicated that the components contained in the final

product were not inhibited by adverse effects and did not limit detection by PCR.

Jiménez et al. (2000) compared the method of rapid analysis of PCR with the

standard method for quality evaluation of pharmaceutical raw materials and finished

products with low levels of microbial contaminants. In their study they developed and

analyzed PCR assays for detecting low levels of bacterial and mold contamination in

pharmaceutical samples containing less than 10 CFU and to compare the assays to

standard conventional methods.

The samples were artificially contaminated with less than 10 CFU of E. coli, S.

aureus, P. aeruginosa, and A. niger. The sample was evaluated for low levels of microbial

contaminants in raw material, including carboxymethylcellulose, lactose, and semiticone,

and for finished product such as: denture adhesives, liquids, anti-flatulents and medicated

ointments. The bacterial DNA was extracted from each enrichment broth by mild lysis in

25

Tris–EDTA–Tween 20 buffer containing proteinase K; DNA mold was obtained by boiling

samples in Tris–EDTA–SDS buffer for 1 hr. A 10-mL aliquot of extracted DNA was added

to Ready-To-Go PCR beads and specific primers for E. coli, S. aureus, and P. aeruginosa.

A 50-mL aliquot of extracted mold DNA was used for amplification of specific sequences

of A. niger DNA.

With the PCR, they detected low levels of microbial contamination in all the

samples of raw material and finished product. Indicating that 100% interrelation exists

between both methods (standard method vs. PCR). The standard methods required 6–8

days, while PCR detection of all microorganisms was completed within 27 hr. Low levels

of microbial contamination were detected in all raw materials and products using PCR

assays and rapid quality evaluation of pharmaceutical samples resulted in optimization of

product fabrication, quality control, and release of finished products.

The barriers to the acceptance of rapid methods in pharmaceutical microbiology

gradually have been overcome. For full acceptance, minimal sample preparation must be

combined with better resolution that does not compromise the effectiveness of testing.

Bosshard et al. (2006) performed a study to compare phenotypic versus molecular

identification. The results of molecular analyses were compared with two commercially

available identification systems (API® 20 NE, VITEK™ 2 fluorescent card). Using 16S

rRNA gene sequence analyses, 92% of the isolates were assigned to species level and

8% to genus level. Using API® 20 NE, 54% of the isolates were assigned to species and

7% to genus level, and 39% of the isolates could not be discriminated at any taxonomic

level. VITEK™ 2 obtained 53%, 1%, and 46%, respectively, with 15% and 43% of the

isolates corresponding to species not included in the API® 20 NE and VITEK™ 2

databases, respectively. Bosshard et al. concluded that 16S rRNA gene sequencing was

26

an effective means for the identification of clinically relevant nonfermenting gram-negative

bacilli.

They proposed an algorithm for proper identification of nonfermenting gram-

negative bacilli and concluded that the majority of strains could not be accurately identified

by phenotypic profiling because species assignment was found to be reliable only when

excellent (or good) species identification was achieved. This was the case in 35% of the

isolates. Molecular identification was more laborious than phenotypic identification; results

of sequencing were usually available within one to two working days. Given these

considerations, they developed an algorithm for the effective and proper identification of

gram-negative nonfermenters in the diagnostic laboratory (Fig. 2.01). Thus, if API® 20 NE

or VITEK™ 2 do not yield an excellent (or good) species identification, the subsequent

assays shoul target nonfermenters and subjected to 16S rRNA gene sequencing if an

adequate species assignment is problematic.

27

Figure 2.01. Algorithm for the identification of nonfermenting gram-negative bacilli.

(Bosshard et al 2006).

In accordance with Bosshard et al. (2006), the 16S rRNA sequenced gene has

several benefits, compared with phenotypical identification, including that it was not

restricted to a specific group of bacteria and information from the public GenBank®

database was available that covers the whole spectrum of phylogenetic diversity; it was

new, so, other systems currently did not have species assigned to a group or related

bacteria. However, the results were precise and do not depend on the change of the strain

or individual interpretation.

An objectionable organism in accordance with the USP38-NF33 (2015) for the

TSM test was any Salmonella spp. Salmonelloses was responsible for many infections in

humans and animals, but are not detectable in certain clinical samples that contain a small

28

number of the organisms. Pathmanathan et al. (2003) studied the possibility of applying

the PCR procedure using a pair of primers targeting the hilA gene to detect Salmonella

spp. A total of 33 Salmonella strains from 27 serovars and 15 non-Salmonella strains from

eight different genera were included. The PCR with all the Salmonella strains produced a

784 bp DNA fragment that was absent from all the non-Salmonella strains tested in the

study. The detection limit of the PCR was 100 pg with genomic DNA and 3 × 104 CFU/mL

with serial dilutions of bacterial culture. Subsequently, an enrichment-PCR method was

developed to test the sensitivity of the hilA primers for the detection of Salmonella in faecal

samples contaminated with different concentrations of Salmonella choleraesuis subsp.

choleraesuis serovar Typhimurium.

The method described by the authors allowed the detection of Salmonella

Typhimurium in faecal samples at a concentration of 3 × 102 CFU/ml−1. They concluded

that the hilA primers were specific for Salmonella spp. and the PCR method presented by

them may be suitable for the detection of Salmonella spp.in feces better than the

conventional method of analysis. The conventional method of isolating strains of

Salmonella spp. takes four to seven days to complete, in addition to being laborious and

needing adequate personnel. Substances such as bilirubin and biliary salts in human fecal

matter contains inhibit PCR use, making organisms detection difficult.

Among the principal monitoring and repeated approach of many investigations of

different industrial pharmaceutical classifications, most of their efforts are applied to the

study and implementation of microbiological methods that includes: organism’s isolation,

early detection of pathogenic organisms, characterization, and enumeration of

microorganisms of environmental samples, raw materials, and finished products. These

are categorized in accordance with the type of alternate technology used. The alternate

technology can include the growth of microorganisms, organism viability, presence or

absence of the microorganisms, cellular components, or “artifacts” present, nucleic acid

29

methods, and traditional methods combined with computerized images (Moldenhauer

(2006). One notable question when audits are made in the pharmaceutical industries is,

What specific outlying organisms within samples analyzed in programs of environmental

monitoring programs need to be characterized and identified to genus, species or serotype

of strains? This key question directs the pharmaceutical industry towards developing

strategies that could be applied effectively to identify pathogenic or objectionable

microorganisms.

Quality control monitoring will include information concerning the excipients,

content of the pharmaceutical ingredients, behavior in manufacture environments, and the

integrity of the finished product (Cundell 2006). These strategies will determine the

specifications of flawed product on not complying with the required specifications. The

specifications are contained under the requests of “International Compendia

Harmonization” (ICH) a harmonized standard for Japanese, American, and European

product specification. With the harmonization changes occurred in 2007 relating to

specifications and methods of tests for pharmaceutical applicable preparations. Involving

the evaluation and adaptation of the limits of values in the monographs principally for the

European pharmacopeia, in accordance with ICH (2007).

The current information from 1997 until 2006 stemmed from an evaluation of 40

different pharmaceuticals to establish limits for microbiological quality testing not

exceeding 93% of the examined raw material. A total of 5.5% of the analyses were

supported to be within the specifications after applying it to the factor of validate of

tolerance of 5 of the ancient method. Only 1.5% of the analyses resulted as out of

specifications (OOS), generally from raw material of vegetable origin (Bomblies et al.

2007).

The FDA has stated that research priorities should be directed towards creating

investigative fields and laboratories with sensitive, effective hardware for analyses which

30

are cost effective (Avallone 1986; Cundell 2006; Miller 1982). Microbiology laboratories

perform tests for the detection of objectionable pathogenic microorganisms from the raw

material up to the finished product, guarantee that the product expires within

specifications. The USP38 Chapters <61> (Microbiological examination of non-sterile

products: Microbial Enumeration Tests (Met)) and USP <62> (Microbiological examination

of non-sterile products: Test for specified microorganisms) establishes the determination

of entire microbiological content of bacteria, yeasts, and fungi in pharmaceutical samples

for non-sterile products. In spite of the regulations, microbial contamination continues as

a major problem on a global scale.

Lee et al. (2008) demonstrated the effectiveness of Real-time PCR technique

based on the determination of the gene rRNA in terms of quantity of the number of copies

analyzed for the absolute and relative quantification with E. coli. The rapid methodology

was cost effective without presenting safety problems, compared to traditional methods

such as Southern Blot Analysis. Detection of medically-related highly-pathogenic

objectionable organisms in the pharmaceutical industry focused on S. aureus and P.

aeruginosa. A modification of Real-time PCR was based on "LightCycler® hybridization

probes" that originally was developed for analysis of clinical samples.

In a study by Skof et al. (2007), optimization of Real-time PCR was achieved by

analyzing 34 different pharmaceutical topical-use application samples compared in

parallel form with the standard protocol for European pharmacopeia. The tests made with

the modified PCR could detect 1-10 CFU of both bacteria in gram or milliliter units of

pharmaceutical product in 26 hr., including 24 hr. of enrichment. However, microbiological

standard methodologies require 5-7 days. They concluded that the Real-time PCR was

an efficient tool for the rapid detection of S. aureus and P aeruginosa in topical-application

pharmaceutical products .

31

The Real-time PCR method used in the analyses of the TSM test in

pharmaceutical microbiology laboratories are not found commonly in literature; articles

usually refer to the chemical product, in medical and biotechnological devices, and clinical

areas concerning objectionable organisms such as: E. coli, P. aeruginosa, Salmonella

spp., S. aureus, A. niger and C. albicans. The methodology of uniplex PCR amplicons

were effectively sequenced, corroborating the conservation of used primers. Other

validation parameters such as specificity, sensitivity, and robustness were evaluated. The

method provides a high performance screening methods to test different pharmaceutical

preparations for specified microorganisms for the detection of microbiological

contamination (Ragheb et al. 2012).

Real-time quantitative PCR (qPCR) technology offers fast and reliable

quantification of any target sequence in a sample (Burgos et al. 2002). They analyzed the

effect of manufacturing lots of PCR reagents on two main PCR parameters, specificity and

sensitivity. It was used four different amplicons, incorporating either viral DNA or mouse

genomic DNA. Even though a PCR product could be obtained among a variety of

parameters, they observed relevant variations in sensitivity relating to the reagents’

formulation. They concluded that different lots of reagents may determine the analytical

performance of PCR assays, indicating that reagents testing import when the PCR

protocol was used for quantitative purposes (Burgos et al. 2002). There were many

methods available for quantification of nucleic acids; real-time -qPCR was the most

sensitive and precise method (Ferre 1992; Klein 2002). Changsoo et al. (2005)

demonstrated the feasibility of two real-time qPCR methods to determine plasmid copy

number. The relative and absolute quantification methods required separate detection of

the plasmid and the host chromosomal DNA. Detection was performed using a primer set,

specific to a single-copy gene in each target molecule. They used SYBR Green I dye in

32

real-time -qPCR analyses because it has several advantages over sequence-specific

probes when singleplex PCRs are performed.

A hybridized test using Real-time PCR really was developed by Kearns et al.

(2002). The test was developed for the rapid detection in less than one hour of the

susceptibility to Penicillin by the bacterium Streptococcus pneumoniae. Real-time PCR

was sensitive to culture methods, microscopy or detection of antigen, in addition to

providing information of susceptibility even in the cases of negative cultures.

Jaffe et al. (2001) evaluated in his study that the methods of Real-time PCR, in

addition to being cost effective for each sample, also provided precision and rapidity of

DNA extraction. In their study he mentions that he pathogenic gram-negative bacteria, P.

aeruginosa, has emerged as one of the most problematic nosocomial organisms. This

objectionable organism is problematic for the pharmaceutical industry because it is

clinically indistinguishable from other gram negative bacterial infections. The percentage

of mortality of P. aeruginosa is high, as well as this bacteria is inherently resistant to

common antibiotics. The detection at clinical laboratory levels is performed by the

standard methodology of identification and susceptibility, taking 48 hr to complete the

analysis.

McArthur and Bibb (2008) developed a method of fingerprinting using DNasae IT7

exonucleous applied to actII-orf4 from f Streptomyces coelicolor. The A3 encodes the

activation of the specific route for the production of the antibiotic actinorhodin. In the

development of a live map, they created oligonucleotides incorporating the regulatory

elements S. coelicolor affected antibiotic production. In the determination of the

oligonucleotide decoys (mimicking fragments of ADN inhibiting specific transcription

factors containing specific sequences of recognition), they made a protocol of transfection

process which was developed for liquid cultivation that support an efficient entry of the

decoys, with RT-qPCR to demonstrate the persistence of the decoy for >70 hr. The

33

measurements in the effect of the growth, expression of, and the production of antibiotic

demonstrated that one of the decoys in concordance with the plate test was more effective

at increasing the production of actinorhodin.

The development of a novel Real-time PCR for the detection and differentiation of

eleven medically important Aspergillus spp. and Candida spp. in clinical specimens was

performed by Schabereiter-Gurtner et al. (2007). The application of the Real-timePCR

targeting the region ITS2 of the fungi was developed for the detection and differentiation

of the species, including Aspergillus flavus, A. fumigatus, A. nidulans, A. niger and A.

terreus, and C. albicans, C. dubliniensis, C. glabrata, C krusei, C. parapsilosis, and C.

tropicalis using the LightCycler® instrument. The results of Real-time PCR were compared

with results of culture, histology, or ELISA system. The results suggest that the tests for

Aspergillus spp. and Candida spp. can be adapted for clinical laboratories as simple

techniques, especially for early scrutiny the objectionable of Candida and Aspergillus spp.

Once again, Real-time PCR methodology was potentially an important tool in the detection

of stages of early diagnosis in infections caused by fungi.

In a retrospective analysis of how the pharmaceutical industry has evolved in the

latter decade, the incorporation of molecular techniques was significantly demonstrated

for analyses for the control of the quality of the pharmaceutical products, especially to

determine the diversity of microorganisms in products utilized by the pharmaceutical

industries. The identification of microbial contamination within products provides valuable

information which can be used to discover the possible source of contamination and

distribution of microbial species in the pharmaceutical environments (Jiménez 2007). The

FDA listed 134 non-sterile products from 1998 until 2006, which represented 48% of

contamination from Burkholderia cepacia, Pseudomonas spp. and Ralstonia picketti.

Contamination by yeasts and fungi represented 23% gathered from products; 60% from

not-sterile product belonging to gram negative bacteria, and 4% associated with gram-

34

positive bacteria (Jiménez. 2007). Of 193 contaminated products, 78% of the

contamination stemmed from the lack of sterility of the product, and 7% from yeast and

fungi contamination. Data revealed that an 6% belonged to gram-negative bacteria, and

1% from gram-positive bacteria.

2.2. Health Risks from Objectionable Organisms

The question, “What was an objectionable organism?” was pertinent among

industrial microbiologists and regulators. This discussion should be based on science,

combined with consideration for reasoned risk analysis (Sutton 2012). The presence of

the objectionable organisms in pharmaceutical preparations represents an imminent risk

to the health of the consumer. The term “risk” within the pharmaceutical industry can be

described from three principals: These can be: (1) in an approach known as the current

good manufacturing practices (GMPs), (2) directed to nonperformance of the raw material

and finished product, and (3) compliance with the USP for the laboratory analyses needed

in accordance with the type of product that was manufactured. This investigation presents

its approach to fulfill analyses of raw material and finished product in accordance with the

requests of the TSM testing in chapters <61> and <62> detail in the USP (2015).

The use of contaminated pharmaceutical preparations has proven hazardous to

the health of users (Cooker 2005). There have been reports of drug-borne human

infections worldwide. Contamination of pharmaceuticals with microorganisms can also

bring about changes in their physical characteristics, including breaking of emulsions,

changes in odor and color, fermentation of syrups, thinning of creams, turbidity, or

deposits, (Shaikh et al. 1988). The incidence of microflora in non-sterile preparations

generally was influenced by the nature of the ingredients (whether natural or synthetic),

and the quality of the vehicle and the care and attitude of personnel involved in their

handling (Parker 2000). Limits for objectionable microorganisms in oral products intended

for use by immunocompromised patient populations, such as children and cancer

35

sufferers, who are more at risk of microbial infections, should be more stringent than limits

for oral products intended for treating patients that are not immunocompromised (Gad et

al. 2011).

With the harmonization of the compendia tests, additional guidance was added to

the informational chapter USP <1111> which states: “…the significance of other

microorganisms recovered should be evaluated in terms of the following:

• Use of the product, with it’s hazard varying according to the route of

administration (eye, nose, respiratory tract).

• Nature of the product, such as does the product support growth? Does it have

adequate antimicrobial preservation?

• The method of application.

• Intended recipient, including risks which may vary for neonates, infants, or the

debilitated.

• The use of immunosuppressive agents, corticosteroids.

• Presence of disease, wounds, or organ damage”.

Where warranted, a risk-based assessment of the relevant factors was conducted by

personnel with specialized training in microbiology and in the interpretation of

microbiological data. Assessment for raw materials processing the product, the current

testing technology, and availability of materials of the desired quality (USP 2015).

According to Sutton and Jiménez (2012) regarding enforcement reports for non-

sterile products, about 75% of the recalls are from either OTC products or personal-care

products from 2004 to 2011. The causes for the non-sterile recalls are presented in

Figure 2.02.

36

Figure 2.02. Causes for non-sterile recalls (Sutton and Jiménez 2012).

Of the 142 non-sterile recalls during the period from 2004 to 2011, a total of 103

were tested positive for objectionable organisms and 22 for microbial contamination in

general. Of those 103 objectable organisms cited in recalls included: 77 gram-negative

bacilli, 3 gram-positive cocci and 23 yeast/molds recalls.

The compendium guidance provides the basis for an evaluation of potentially

objectionable organisms by a competent, trained, professional microbiologist. Risk

assessments are encouraged (Sutton and Jiménez 2012). The first consideration should

be total numbers of microorganisms present. Immunocompromised patients are at

increased risk for oral drugs, so low total aerobic counts (10 CFU/g for tablets) were

established for the specification (Manu-Tawiah 2001). The source or the cause of primary

contamination must be identified for appropriate action to be taken. The primary

contamination may be intrinsic with contaminated raw materials, or extrinsic-contaminated

37

during or after processing (Kushwha 2009). The severity of the effects that

microorganisms may have on any particular drug product is a function of the nature of the

product with its intended use, and the nature of the contaminant microorganisms

(Kushwaha 2010).

At one end of the spectrum, microbial contamination of a sterile parenteral product

may, on injection into a incapacitated patient, result in fatality; or patients may refuse to

begin or continue medication because of aromas, discolorations, or off-flavors with

microbial origin. Thus, the presence of microorganisms should to be avoided in drug

products (Nigel 2002).

2.2.1. From GMP’s Perspective

The main importance of verifying that raw material and finished products are free

of the presence of microorganisms is that contamination could represent a risk to the

health of the consumer; thus, it ismportant to follow Good Manufacturing Practices (GMPs

2015). According to Title 21 of the Federal Code of Regulations, Part 211 (21CFR211

2014), following the norms of GMP applying to food, cosmetic, and drugs industries will

provide quality standards for manufacture of the product, with non-compliance products

removed from the market. This prevents health risks to human beings. In the United

States, current Good Manufacturing Practice (cGMP) regulations are issued by the U.S.

Food and Drug Administration (FDA) as the minimum requirements for quality systems for

FDA-regulated products such as biologics, devices, drugs and food. One requirement of

cGMP regulations was the monitoring of microbiological contamination (Kushwaha 2009).

The critical nature of the issue was reflected by its presence in three separate citations in

the cGMP (Sutton 2012):

21 CFR 211.84(d)(6) “Each lot of a component, drug product container, or closure

with potential for microbiological contamination that was objectionable in view of

its intended use shall be subjected to microbiological tests before use.”

38

21 CFR 211.113(a) “Appropriate written procedures, designed to prevent

objectionable microorganisms in drug products not required to be sterile, shall be

established and followed.”

21 CFR 211.165(b) “There shall be appropriate laboratory testing, as necessary,

of each batch of drug product required to be free of objectionable microorganisms.”

The GMPs include all the aspects in the manufacture of a drug, from the receipt of

the raw material, the facilities, equipment, training, and hygiene of the personnel to the

completed product. The GMPs require a detailed, written standard operating procedure

(SOP) for process, analysis, and handling that involves the quality of the final product.

Compliance with the GMPs protects the consumer by not allowing the sale of drugs or

products not fulfilling quality standards, such as non-intentionally added toxic content, for

non-compliance of a formula, or problems with processing, packaging, analysis, or

microbial or particulate contamination during its product manufacture (21CFR211. 2014).

Pharmaceutical manufacturing generally includes a complex, multi-step handling and

processing system risks from microbial contamination from diverse sources including

container-closures, equipment, personnel, the environment, the facility, and raw materials

(Guilfoyle et al. 2013).

The analyzed raw material and finished product must demonstrate that it comply

within the standard of GMPs named as a pure, safe, and effective product. If the company

was not complying with cGMP regulations, any drug made by them was considered

adulterated under the law. This means that the drug was not manufactured under

conditions that comply with cGMP (Rodríguez-Pérez 2014). It does not mean that there is

necessarily something wrong with the drug. The product can be perfectly safe for use, but

as established in the FD&C Act, a drug that was not manufactured following cGMP

requirements was considered adulterated. If failure to meet cGMP results in the

39

distribution of a defective drug, the company may subsequently recall that product. Once

the company follows cGMP, its label was assured of efficacy of the product.

The FDA trains its inspectors so that they conduct GMP examinations to ensure

compliance with its regulations. Thus, it guarantees the consumer a healthy, safe product.

The GMP is a system that helps ensure that the medicines are prepared and controlled in

a consistent way with specified quality standards and are manufactured as stipulated

when submitted to the FDA for commercial approval. Consequently, every industry

designs, in accordance with the fulfillment of the regulations of GMPs, the target to

minimize the risks of contamination by objectionable, indicators and/or pathogenic

organisms in accordance with the distinctions that the FDA authorizes. The principal risks

mainly found by the US FDA for the year 2013 and 2014 are described in Table 2.01.

Table 2.01. Major risks for non GMP compliance.

FDA US. 2014. Product recall.

Type of Incident Risk

Contamination in the final product Contamination in raw material

Damage to health Death

Presence of particulate matter Local inflammation, phlebitis, mechanical disruption of tissue, or immune response to the particulate lead to granulomatous formation, etc.

Incorrect labeling of containers

Incorrect medicine Secondary effects Damage to health Death

Lack of active ingredient Active ingredient higher than established

Ineffective treatment Harmful effects

40

Without GMPs it would be difficult to assure the quality, homogeneity, and

reproducibility of a lot of production, not only with regard to other lots, but inside the same

batch of the product. The federal regulations for the current good manufacturing practices

(cGMP’s) of drugs states quite clearly in 21 CFR 211.84(d)(6) that “each lot of a

component, closure, or drug product container, that has potential for microbiological

contamination, that is objectionable in view of its intended use, shall be subjected to

microbiological tests before use.(Torbeck et al. 2011).

2.2.2. Non-compliance of Product

The microbial diversity present in the pharmaceutical environment can provoke the

immediate withdrawal of the product from the market with the imminent alert of

safeguarding the health and life of the consumers because each product may support

microorganisms. Microbial growth potential can have a significant effect on the

development and design of the drug manufacturing process (Lolas and Metcalfe 2011).

The distribution and growth of the microorganisms in the pharmaceutical ambience

is limited by the environmental gradient where microorganisms may survive and grow with

various gradients of factors such as: temperature, water availability, pH, and concentration

of organic available components. Each gradient factor has a threshold value according to

each organism. Consequently, the optimization of the system is needed in terms of

environmental control to eliminate fluctuations, and to minimize or eliminate the survival

of the organisms. The presence of objectionable microorganisms in non-sterile products

is indicative of poor quality during the process, and the absence of optimization of the

system (Jiménez 2007).

The identification of microbial contamination in retired products (for recalls

definition, refer to Table 2.02) from the market provides information of the possible sources

of distribution of microbial species in the pharmaceutical industry environment (Torbeck

et al. 2011).

41

Table 2.02. Product recall definition.

A “recall” is a firm’s removal or correction of a marketed product that the FDA considers to be in violation of the laws it administers, and against which the FDA would initiate legal action (e.g., seizure). Recalls do not include market withdrawals. FDA assigns a numerical designation (I, II, or III) to a particular product recall to indicate the relative degree of healthhazard presented by the product.

A Class I recall: use of or exposure to a violate product may cause temporary or medically reversible adverse health consequences or where the probability of serious adverse health consequences is remote.

A Class II recall: the use of, or exposure to, a violate product may cause temporary or medically reversible adverse health consequences or where the probability of serious adverse health consequences is remote.

A Class III recall: a situation in which use of or exposure to a violate product is not likely to cause adverse health consequences.

Market withdrawal: occurs when a product has a minor violation that would not be subject to FDA legal action. A firm should remove the product from the marketplace or correct the violation. A product could be removed from the market due to tampering, without evidence of manufacturing or distribution problems.

Medical device safety alert: issued in situations (which may also be considere as recalls) where a medical device may present an unreasonable risk of significant injury.

Data obtained from U.S. FDA 2014.

An analysis of information of pharmaceutical products withdrawn by the FDA,

concludes, that of 134 non-sterile products from 1998 to September 2006, 48% of the

retired products were contaminated by B. cepacia, spp., or Ralstonia picketti (Jiménez

2007). Of these drugs, 23% were caused by yeasts and fungi. Sixty percent (60%) of the

outlying retired products contained gram-negative bacteria, and 4% were identified as

gram-positive bacteria. Otherwise, in the classification of sterile products,they indicated

that of 193 withdrawn products, 78% of the products lacked sterility and 7% were

contaminated with yeasts and fungi. About, 6% of the products were from gram-negative

bacteria, and a 1% were from gram-positive bacteria. Among the non-sterile and sterile

42

products, B. cepacia was the most frequent isolated organism, with 22% and 2.5% of the

products withdrawn from the market, respectively. Based on the literature, B. cepacia,

Pseudomonas spp. and Ralstonia picketti can be associated with water contamination,

whereas the yeasts and fungi and gram-positive bacteria can indicate poor environmental

controls (Jiménez 2007).

One of the principal causes of recalls reported by Torbeck et al. (2011) may have

been attributed to the bacterium B. cepacia complex (Bcc) because of inadequate testing

and specification (e.g., inadequate microbiological analysis, contaminated raw materials,

and incomplete/incorrect testing for antimicrobial effectiveness). The product recalls

included occurred during the years 2000 and 2008. A total of eight recalls were Class I,

six were Class II, and two were Class III. Of the recalls, six recalls were initiated voluntarily,

following FDA judgments, and the firms initiated an additional 10 voluntary recalls. The

product types included anticavity rinse, baby and adult washcloths, electrolyte solution,

eyewash, mouthwash, nasal spray, radiopaque preparations, skin cream, surgical prep

cloth. Burkholderia cepacia complex had contaminated each of these products, even in

the presence of one or more antimicrobial preservatives. This species consists of 17

closely related species of the ß -proteobacteria subdivision, which continues to be

principal cause for contamination within the pharmaceutical industry (Torbeck et al. 2011).

Table 2.03 lists retired products from the market for the year 2013 as a consequence of

contamination by microorganisms (FDA US 2013).

43

Table 2.03. Products withdrawn from market in 2013 due to microbial contamination.

Date Industry Product Contaminant

Mar 20, 2013

Medprep Consulting Inc. All Lots of all compounded products

Potential mold contamination

Mar 20, 2013

Clinical Specialties Compounding Pharmacy

All lots of sterile Products repackaged and distributed by Clinical Specialties Compounding

Lack of sterility Assurance

Mar 17, 2013

Medprep Consulting Inc. All lots of all Compounded Products

Potential mold contamination

Mar 16, 2013

Medprep Consulting Inc. All ots of Magnesium Sulfate, 2g in Dextrose, 5% in water, 50mL for injection

Mold contamination

Retrieved from U.S. Food and Drug Administration. Guidance Documents. 2015.

Verifying compliance with quality standards of the articles, raw material and

finished product was critical to avoid major consequences as demonstrated in Table 2.04.

The FDA and the Center for Disease Control and Prevention (CDC) identified the

presence of contamination by bacteria and/or fungi in vials without opening the products

Betamethasone, Cardioplegia, and Triamcinolone (FDA U.S. 2014, CDC 2014). Among

the included specified organisms, there were bacteria such as Bacillus spp. and fungi such

as Aspergillus fumigatus, species that can be pathogenic to the human beings.

Particularly, A. fumigatus was a risk for immunocompromised patients.

44

Table 2.04. CDC and FDA Laboratory-Confirmed organisms from product samples.

Laboratory-confirmed Organisms from Product Samples Associated with NECC Recalled Lots of Betamethasone, Cardioplegia, and

Triamcinolone Solutions

Medication Lot Number Microbial Contamination

Betamethasone 6 mg/mL injectable – 5 mL per vial

08202012@141 Paenibacillus pabuli/amolyticus; Bacillus idriensis; Bacillus flexus; Bacillus simplex; Lysinibacillus sp., Bacillus niacini, Kocuria rosea, Bacillus lentus

Betamethasone 6 mg/mL injectable – 5 mL per vial

07032012@22 Bacillus niabensis; Bacillus circulans

Betamethasone 12 mg/mL injectable – 5 mL per vial

07302012@52 Bacillus lentus, Bacillus circulans, Bacillus niabensis, Paenibacillus barengoltzii/timonensis

Betamethasone 6mg/mL injectable – 5 mL per vial

08202012@44 Bacillus lentus, Bacillus firmus, Bacillus pumilus

Betamethasone Betamethasone 6 mg/mL injectable – 5 mL per vial

08152012@84 Penicillium sp., Cladosporium sp.

Triamcinolone 40mg/mL injectable – 1 mL per vial

06062012@6 Bacillus lentus, Bacillus circulans, Bacillus niabensis, Bacillus nealsonii, Bacillus subtilis group, Bacillus firmus

Triamcinolone 40 mg/mL injectable – 2 mL per vial

08172012@60 Aspergillus tubingensis, Penicillium sp.

Triamcinolone 40mg/mL injectable – 10 mL per vial

08242012@2 Aspergillus fumigatus

Cardioplegia solution 265.5 mL per bag

09242012@55 Bacillus halmapalus/horikoshii, Brevibacillus choshinensis

Data obtained directly from (CDC) Centers of Disease Control and Prevention 2014.

45

When we do an interrelation of the outlying and identified organisms quoted in

Jiménez’ 2007 study, and the presented organisms of the products withdrawn from the

market by the FDA and identified by the CDC we see that these organisms do not belong

to the USP list of objectionable organisms. Nevertheless, it was important to highlight that

, if we refer to the USP in Chapter <1111> microbiological examination of non-sterile

product: criteria of acceptance for pharmaceutical preparations and substances for

pharmaceutical use it establishes that the presence of certain microorganisms in non-

sterile preparations can have the potential of reducing or deactivating the therapeutic

activity of the product. These organisms may provoke adverse effects to the health of the

patient. Thus, pharmaceutical industries are entrusted with assuring a decrease in the

bioburden of the different dosages of the finished product that they manufacture. The most

important and determinant aspect was that they must adhere to Chapter <1111> where a

new stipulation indicates that, in addition to the microorganisms listed as specific

organisms that must be absent in non-sterile products, the industry has the responsibility

for determining the significance of the presence of other organisms recovered, in terms of

risk to the public health.

The isolated organisms clearly described in the released investigations previously

presented in this study belong to the division of other organisms, because these were not

a part of the specific objectionable organisms. Otherwise, these organisms appear to be

isolated cases among the retired products. This indicates that the industry cannot limit its

identification to objectionable organisms, indicators and/or pathogenic organisms that

appear in the USP lists. For this reason, the industry must evaluate other organisms

isolated under the following parameters (USP38 2015):

Use of the product; dangers vary according to the route of administration (eyes,

nose, respiratory tract, etc.).

46

Nature of the product; does not product support microorganisms growth and

maintains sufficient antimicrobial preservation.

Application of standardized product-analysis methods.

Target population (newborns, infants, etc.).

Uses immunosuppressive cortisteroid agents.

Presence of diseases, wounds, organ damage.

In the event that any of these situations present, it was important that an evaluation

of the risks of pertinent factors be performed by personnel specialized in microbiological

protocols and capable of the interpretation of the microbiological data USP38-NF33,

Chapter <1111> (2015).

2.2.3. From the United States Pharmacopoeia (USP) Perspective

Pharmaceutical products are exposed to microbiological contamination that can

represent risks to the health of the consumer (FDA 2015). The presence of certain

organisms in non-sterile preparations can have the potential of reducing or inactivating the

therapeutic capacity or efficacy of the pharmaceutical product and having the potential to

adversely affect the health of patients, in accordance with the USP38-NF33 <1111>

(2015). The presence of these organisms may causes degradation of the product,

changes in the aesthetics, and the efficacy of the product. If these organisms are

potentially harmful or may produce toxins, there would not be in compliance with USP

(2015). The FDA identifies the microorganisms as: harmful, objectionable, and

opportunists (FDA 2009).

An organism is assigned a harmful designation if it contained or liberated toxins

and is responsible for infections and illnesses in human beings. Examples of these

organisms are: Salmonella spp., S. aureus, E. coli, P. aeruginosa, C. albicans and A.

niger. These well-known organisms designated as objectionable can cause illnesses or

47

deterioration of the product. Some strains of the genus Pseudomonas, like P. putida and

P. maltophilia represents this type of organism. Opportunistic organisms can cause

illnesses in immunocompromised patients and includes the majority of the microbial flora

in raw material, excipients, and those from pharmaceutical manufacturing environments

(Jiménez 2007; Clontz 2009).

The USP establishes methods for determining the microbiological load known as

a bioburden test and the microbial limits test designed to determine aerobic or facultative

anaerobic heterotrophic mesophilic organisms because these are the most common

contaminants in pharmaceutical industry facilities. Table 5 summarizes the organisms

identified by the FDA as organisms that should not be present in pharmaceutical

preparations and present a risk to consumer health (Jiménez 2007).

48

Table 2.05. Summary of organisms that are more frequently a health threat.

Organism Morphology

Effect

Clostridium spp. Gram positive bacteria bacillus

Causes toxic effects. Possible virulence like gas gangrene, necrotic enteritis.

Candida spp. Yeast Pathogenic to humans Aspergillus niger Fungi Aspergilliosis Fusarium spp. Penicillium spp. Alternaria spp. Trichosporum spp.

Fungi Opportunistic pathogens to humans

Trychophyton spp. Microspora spp.

Fungi Potential source of infection in keratinized tissues

Staphylococcus spp. Micrococcus spp. Streptococcus spp.

Gram positive cocci bacteria

Skin infection Toxins and enzymes produced destroy the white and red cells Considered a highly opportunistic pathogen Resistant to antibiotics.

Staplylococcus epidermidis S. saprophyticus S. haemolytycus

Gram positive cocci bacteria

Opportunistic pathogen. Less virulent than S. aureus

P. aeruginosa Gram negative bacteria bacillus

Pathogen in humans. Causes infections on burns, cuts, urinary tract and lower respiratory tract.

2.3. Contribution to Procedures, Costs and Benefits for Pharmaceutical Industries

The first rapid methods of microbiological analysis by clinical microbiologists began

in the 1960 (Fung 2002). Rapid methods and automation in microbiology was a dynamic

area in applied microbiology dealing with the study of improved methods in the

characterization, early detection, isolation and enumeration of microorganisms in clinical,

food, industrial, and environmental samples. From 1960 to 1970, rapid methods of

identification of microorganisms used by the pharmaceutical industry was based on a few

"kits" for miniature tests. This miniaturized methodology was used mainly as an alternative

technology to traditional biochemical tests. Figure 1.01 estimated the trends of rapid

49

methods and automation in microbiology by medical microbiologists and food

microbiologists from 1965 to 2000. The age of miniaturization and diagnostic kit

development occurred from 1965 to 1975; the age of immunological test kits from 1975 to

1985; and the age of genetic probes, molecular testing systems, and polymerase chain

reaction (PCR) from 1985 to 1995 (Fung 2002).

The PCR method marked an important stage in microbiological analysis, both in

clinical samples and in the field of pharmaceutical investigation. The challenge to the

clinical microbiology laboratory was how to respond to problem, for example in the case

of a worldwide pandemic (such as MRS- methicillin-resistant S. aureus). This includes the

implementation of protocols for mandated legislation that will require an active screening

program. Although more expensive molecular techniques have the potential of providing

sensitive and rapid results, they may not be the most appropriate for some institutions.

Culture requires a longer processing time, but can achieve comparable sensitivity,

depending on the culture method employed (Marlowe et al. 2011). The Table 2.06 shows

the advantages and disadvantages of culture and molecular methods for screening of

MRSA according to Marlow et al. (2011).

Table 2.06. Advantages and disadvantages of culture and molecular methods for

screening of MRSA.

Method Sensitivity Specificity (%)

Time to results (h)

Cost Technologist Skill Level

Culture

Molecular

Lowª

High

100

<100

18-48

<24

Low

High

Moderate

Moderate to high

ª Improved sensitivity with chromogenic agars and broth enrichment.

In the industrial field the rapid microbiological systems are grouped together in four

(4) principal platforms (Clontz 2009).

50

1. Methods based in the growth of microorganisms: This test measures

chemical reactions and physiological changes of the organism as a result of

microbial growth under specific conditions, where technicians can measure

such changes as electrical impedance / conductivity of the test solution,

biochemical reactions, or ATP bioluminesce.

2. Methods based on the artifact: This method involves the analysis of the

component of the microbial cell, including fatty acids. These methods include

gas chromatography, ELISA, and MALDI-TOF mass spectrophotometry

cellular components such as nucleic acids and proteins.

3. Methods based on nucleic acids: This method targets the amplification of

the DNA of microorganisms using PCR and the riboprinting technique. This

methodology was used to identify microorganisms and differentiate between

species and was useful for the identification of organisms involved in

contamination during the investigation process for non-compliance with the

quality control standard.

4. Methods based on viability: This methodology involves the use of viable

strains or biological bookmarks capable of detecting and of enumerating

microorganisms without the need to incubate them to increase their cellular

density. An example of this technology includes the fluorescent labeling

methods such as: fluorescent flow cytometry, immunofluorescence and

fluorescent coloring of nucleic acids used as viable labels.

The technique of real-time quantitative PCR, provides advantages for the detection

of indicator and objectionable organisms for the pharmaceutical industry. Of the four

platforms, the number three: “methods based on detection of nucleic acids” should be the

main approach utilized by the Pharmaceutical Industries (Clontz 2009).

51

The principal contribution of this investigation for the industries was through the

study of minimal trace detection of E. coli ATCC®8739™ and Aspergillus brasiliensis

ATCC®16404™ in pharmaceutical preparations using the Real-time PCR technique. Real-

time PCR can detect objectionable organisms with sensitivity, specificity, efficacy for the

characterization and rapid identification during the analyses of raw material, excipients,

environmental monitoring and finished product.

These four parameters can be defined as follows:

Sensitivity: indicates the minimum quantity of DNA required for the

amplification to be produced.

Specificity: obtains an amplified sole product, such as the detection of a

specific sequence for E. coli ATCC®8739™ y A. brasiliensis ATCC®16404™

strains.

Efficiency: achieves the maximum amplification in a determined number of

cycles validated.

The present study will help to set the bases for the development of written

procedures, protocols, and validations for the TSM, using the specific molecular

methodology of Real-time PCR. Through this investigation, another contribution will detail

a step-by-step protocol for microbiological molecular analyses applied directly to samples

of raw material and pharmaceutical OTC product. In terms of this point it important mention

that there was little or no literature regarding the application of Real-time PCR directed to

the test TSM in the pharmaceutical industry. The majority of the performed studies are

directed to the PCR methodology. Currently, in Puerto Rico, through an interview with

personnel, there are eight (8) industries that are using MicroSeq® molecular system. No

industry in the Island uses the methodology for microbiological analysis in

pharmaceuticals or biopharma.

52

The MicroSeq® method was directed to the genetic sequence of 16S at the

ribosomal level. Currently in Puerto Rico (in 2013) the UPR Medical Sciences Campus,

Forensic Sciences program and other Universities on the Island utilize Real-time PCR

(Personal communication: Ana Lliteras Field Application Specialist, Molecular Biology

Bioanalytical Instrument. 2013). The prevailing techniques used in most of the

pharmaceutical industries, and described in the compendium were conventional

techniques, with culture transfer methods, phenotypic observation of the colony, and

biochemical tests to obtain final identification (USP38-NF33 <61> 2015). These

conventional techniques consume a lot of time, are not specific, and, lack accuracy and

precision to demonstrate the entire content of the presence of viable organisms in a

sample (Jiménez 2011).

A Real-time PCR system to universally detect microbes at a limit of 10 to 50 CFU

within 5-6 hr was developed by Walzer et al. (2007). Their system was useful to replace

conventional microbial plating techniques for the analysis of microbial contamination in

liquids such as water. Advantages of using rapid systems include reducing the product

release cycle time, reducing raw material, decreasing simultaneous work-in-progress,

increasing finished product inventories, improving the quality of the microbial testing,

automating microbial testing, collecting electronic test data and information

creation,starting investigations earlier in response to the out of specifications (OOS)

results, potentially reducing risk of microbial product contamination, and refining the

manufacturing processes within the laboratory (Cundell 2006).

Table 2.07 breaks down the principal contributions of this investigation to the

established SOPs in the pharmaceutical industries and the principal benefits of adopting

this molecular technique.

53

Table 2.07. Contribution and benefits to SOPs.

Contribution to Established SOPs Benefits of the Technique

Addition of molecular methodology to established SOP. Add the analysis of TSM and bioburden under technique Real-time PCR.

The validation of Real-time PCR facilitates the securing of the results of rapid identification in a minimum of 30 min for contaminants, objectionable organisms, and pathogenic contaminants. Detected as the reaction was occurring.

Alternate or principal technique

As an alternate it suggests using identification of detected objectionable organisms and detection of contamination of lots on hold because they are in a process of investigation. Facilitates decision-making due to promptness in securing results.

Restructuring flow of analysis tests in the laboratory.

Real-time PCR facilitates a practical distribution in the execution of the analyses because it decreases work load. It diminishes the preparation of means, solutions, wash teams, sterilization cycles and personnel required by conventional method.

Decrease of test execution time. Releasing batches of products in less time. The execution of the test by Real-time PCR can take from 30 min with the StepOne™ Plus

equipment. Conventional methods require 2 days for bacterial growth and 7 days for fungi and yeasts, then organisms are characterized and identified after the incubation time.

Decrease time to analyses results

Accelerates speed of the analyses and/or its response. Real-time PCR is obtained in fewer than 50 min, whereas traditional methods takes 5 to 14 days.

54

The traditional technique for the identification of fungi and yeasts requires too

much time and effort, and requires analysts with experience in mycology, whereas the

Real-time qPCR involves only one step based on the sequence of the preserved DNA.

The Real-time qPCR has demonstrated, in clinical studies, the advantages of an effective

system, including effective, precise, and rapid analyses that traditional techniques of

culture and identification do not provide. While standard identification methods require a

mean of several days, the virtue of quick analysis, specific primers, and PCR protocol with

a S. aureus contamination of about 1-2 CFU mL¯¹ in pharmaceutical products were

identified to species level by Samadi et al. (2007).

The Real-time PCR technique contributes to the mandate of the GMPs to assure

that the final product was pure, safe, and effective, while providing a mechanism of

minimal detection at trace levels of the presence of any microbiological contamination.

Table 2.07., continued. Accuracy, sensitivity and speed of analysis

Facilitates rapid execution of assays. Precision was based on determining DNA concentration and the sensitivity to detect the presence in the sample. The traditional method determines growth in terms of CFU of the organism through pour-plate or filtration method.

Analysts' training with the technique (multi-faceted analysts)

Positively affects distribution of human resource The technique is not complicated and with adequate certification training, analysts can gain knowledge of events within in the termocycler during the analysis.

Decreased biomedical waste Decreased materials utilized and as such saving space on deposits of biological waste. Facilitates removal of biological waste Avoid media preparations, solutions and equipment used in traditional method was.

55

This technique helps safeguard the health and life of human beings and animals, and to

distribute medicines that expire with the quality standards required by regulatory agencies.

Currently, the microbiological tests performed in the pharmaceutical and

biopharmaceutical industries are divided into three principal categories, including (1)

detection that implies a qualitative aspect; (2) quantitative enumeration; and (3)

characterization and identification of the viable specified organisms (USP <61>, <62>

2015).

Traditionally the microbiological methods described in the compendium (USP

2015) are based on microbial growth. These require a lot of work and manipulation, in

addition to being time consuming. These types of tests need days of incubation,

obstructing and preventing management from taking proactive measures to correct

contamination caused by viable microorganisms that may represent an imminent risk to

the health of the consumer and contribute to nonperformance of the product.

Consequently, microbiological growth techniques limit operations, thus directly impacting

the sensitivity, accuracy and reproducibility of the test because it involves extended

manipulation during test execution. The conventional methodology, based on microbial

growth lacks flexibility and relies on colony counts which require mathematical models to

achieve the best interpretation of the results (Kai 2004).

Most RMM are inherently expensive and the justification of its introduction into a

system requires a thorough evaluation of the method itself. Table 2.08 present an example

of a generic and a practical approach to its measurement in a real laboratory world (Gandhi

2006).

56

Table 2.08. Generic and practical approach to microbial mesurements methods.

Input variables Output variables

Material cost (media, kits, reagents,

glassware, etc.) (Mc)

Number of samples processed

(Sn)

Labor cost (employee, training, etc.) (Lc) Number of samples released or

reported results . (Sp)

Fixed cost (instrument, lab space,

validation, maintenance, etc.) (Fc)

Number of samples re-tested

(Sr)

Number of materials (media, kits,

reagents, ect) (Mn)

Process time (St)

Number of man hours (preparation,

testing, cleaning and maintenance, etc.) (Hn)

Number of working hours (Hn)

According to Gandhi the technique for measuring the efficiency presented in his

study was broad and its application was not limited to RMM; rather, it can be applied to

laboratory instruments as well (2006). However, Table 2.09 presents the principal

contributions of this investigation in terms of saving time and money, compared to the

Real-time PCR technique. The costs were presented in terms of the decrease or

elimination of efforts during the achievement of the test. Table 2.10 presents the savings

in terms of materials, personnel, effort and time invested during the tests of the

conventional microbiology that describes the compendium of the USP versus the method

of Real-time PCR.

57

Table 2.09. Contribution in terms of cost for the pharmaceutical industry.

Contribution

Mechanism

Decrease in costs

From a conventional methodology of microbiology to the molecular Real-time PCR

System change from growth in Petri dish to micro tube.

Elimination of use of large quantities of Petri dishes and culture media.

From a system of huge handling to one of less intervention

Elimination of a continuous system of transfer to a Petri dish for one of the filling wells. Elimination of manipulation to then apply to automated systems.

Minimizing costs and time avoiding time repetition of samples or "resamples" from contamination during the manipulation of the sample. Savings in investigations when the test fails due to microbiological handling during sampling.

From a system limited by the type culture media and conditions of incubation to one that identifies any live or dead sample according to molecular protocol designed.

Elimination of the integrity factor from the media in terms of nutrients, water content, temperature of pouring technique and manipulation.

Time saving in the preparation of the medias and solutions. No need to wait for days of incubation.

From a system where wait times are days and weeks to obtain results to a system where results are obtained at the moment of the reaction.

Eliminates the incubation process and posterior testing. Results are obtained in 30 min to 1 hr.

Time saved in materials, better use of personnel. Results are obtained immediately, easing the decision making in OOS cases.

From a system based in numbering, detection in CFU, and characterization to a system that identifies the organisms genome base on DNA concentration.

Conventional system uses multiple equipment while Real-time PCR uses only one equipment.

Time savings and better distribution in the work flow in the lab.

From a system based in phenotypic characteristics and biochemical tests to a sensitive, accurate and DNA detection.

Conventional system uses selective and differential media. Molecular test detects genetic expression.

Money saving in early detection of minimal contamination, thus avoiding non-compliance with quality standards.

58

In contrast, Table 2.10 presents the costs per reaction based on the costs of the

reagents that were used in this investigation. This cost does not include miscellaneous

items such as micropipettes, micropipette tips, gloves, trays, or biosafety bag. The

intention of Table 2.10 is to provide an estimate of the investment in the reagents needed

for the execution of the test of TSM for the method of Real-time PCR.

Table 2.10. Costs of molecular reactives translated to costs per reaction.

Description Price Items (quantity

Total Cost per reaction

Molecular reagents and primers

Sufficient reactive to perform 200 reactions 3 reactions (triplicates) = 1 sample 66 Samples can be processed with this data the cost per sample would be approximately $6. This does not include energy, labor and miscellaneous.

TaqMan MGB Probe 6,000pmoles

$258.00 2 $516.00

Sequence Detection Primers Aspergillus niger, Escherichia coli

$30.00 4 $120.00

PrepMan® Ultra

Sample

$103.00 1 $103.00

TaqMan® Fast

Universal PCR Master Mix

$208.00 1 $208.00

Water Molecular Biology Grade, Ultrapure

$13.39 1 $14.00

Micro tubes 2mL $155.40 1 $156.00

TOTAL 10 $1,117.00

2.4. Advantages and Disadvantages of the Techniques Described in the

Pharmacopeia vs. Molecular Method

The methodology described in the pharmacopeia for the analysis of the

microbiological quality for non-sterile products comprises two general methodologies.

These are: (1) methods for the enumeration, which represents the quantitative aspect of

the analysis, and (2) methods of determination of specific organisms, qualitative aspect of

the test. In pharmacopeia these two methodologies are contained in Chapters <61> and

59

<62> of the USP38-NF33 (2015). The following was the description of the conventional

method used by the pharmaceutical industry, as described in the compendium of the

USP38-NF33. (2015).

2.4.1. USP38-NF 33 Chapter <61> Microbiological Examination of Non-sterile

Products:Test of Microbiological Enumeration

USP Chapter <61> details the tests for the quantitative estimate of bacteria and

mesophilic fungi present in various pharmaceutical articles, from raw material to the

finished product. These tests are mainly designed to verify that the test expires within the

specifications for microbial quality. The alternate methods such as automated methods

can be used instead of the tests established in the compendium whenever there was

evidence of the studies of qualification, and these demonstrate its equivalence (USP38-

NF33 <61>. 2015). The preparations of the samples can also be modified as needed,

whenever they are based on the obtained results of the qualification tests. Any

antimicrobial property present in the product has to be removed and neutralized before

the routine tests are performed. This chapter contains a guide for the procedures in

estimating of the total count of aerobic microorganisms (TAMC – Total Aerobics Microbial

Count) and the entire count combination of yeast and fungi (TYMC-Total Yeast Mold

Count).

2.4.1.1. Preparation of the Sample

According with the USP, the samples must be prepared using the method that fits

the type of product to be analyzed. The validation of the analysis method for the product

ensures that the preparation of the sample does not alter any microbiological component

present in the sample and that no antimicrobial or inhibitory property supports their growth

(USP38-NF33 <61>. 2015).

When the sample are prepared for TSM, all the steps in the analysis have to be

made using sterile materials and aseptic techniques. The USP provides the

60

recommendations to dissolve or to suspend the product to obtain a homogeneous mix in

the event the product shows insolubility characteristics. The preparation of the sample

involves dissolving or suspending 10mL or 10g from the specimen to be analyzed in a

sodium chloride peptone buffer with a pH value of 7.0 or a phosphate buffer of 7.2pH, or

broth of Soy Casein Digest Broth (SCDB).

The prepared dilution was usually 1:10mL. Chapter <61> provides the guide for

the preparation of the sample when the sample was insoluble in water; the product was in

aerosol and for transdermal patches.

2.4.1.2. Total Aerobic Microorganisms Count (TAMC)

The TAMC test for the estimation of aerobic mesophilic microorganisms for

bacteria, as well as for fungi, utilizes the culture media Soy Casein Digest (SCD). This test

was performed using aseptic techniques with Laminar Flow Hood (LFH) conditions or in a

Biological Safety Laminar Flow Hood cabinet to prevent contamination during the test. It

was applied to non-sterile samples, or from the raw material, finished product, and process

formulations. This analysis was performed using the following techniques, depending on

the type of product and the confirmation of suitability during the validation of the method:

(1) Membrane filtration

(2) Pour-plate method

(3) Spread-plate method

(4) MPN – most-probable-number method

Table 2.11 presents four methods used in the pharmaceutical industry to analyze

the raw material, finished product, and process formulations. It summarize the steps for

the process of the samples to the incubation process (USP <61>. 2015).

61

Table 2.11. Products examination techniques.

Technique Procedure

Filtration by membrane

Transfer an appropriate amount to two filters with membrane.

Immediately filter the sample. Rinse by flushing each membrane.

Transfer a membrane over the SCD agar surface for TAMC. Incubate at 30-35°C between 3 to 5 days.

Transfer a membrane over the SDA surface for TYMC. Incubate at 20-25ºC for 5 to 7 days.

Calculate the number of cfu per g or mL of product.

Pour plate

Transfer 1mL of the sample to two sterile Petri dishes, in duplicate for each medium.

Add 20mL of SCD agar to one of the plates that contains the sample.

Homogenize

Add 20mL of the SDA agar to another plate containing the sample.

Homogenize

Incubate at 30-35ºC 3 to 5 days for SCD.

Incubate at 20-25ºC 5 to 7 days for SDA.

Select the corresponding plate corresponding to a given dilution and that shows highest number of colonies less than 250 for TAMC and 50 for TYMC.

Take the average of the counts by means of culture and calculate the cfu number per g or per mL.

Surface-Spread

Transfer 1mL of the simple over the surface of the culture medium.

Perform duplicates per each culture medium.

Disperse in an even form.

Incubate at 30-35ºC 3 to 5 days for SCD.

Incubate at 20-25ºC 5 a 7 days for SDA.

Select the corresponding plates to a given dilution that it shows the highest number of less than 250CFU for TAMC and 50CFU for TYMC.

Take the average of the counts by means of the culture medium and calculate the number of CFU per g or mL.

62

Table 2.11. continued.

Most-Probable-Number

The results by this method are unreliable. These are obtained especially for mold counts.

Thus, it was kept for the TAMC enumeration in situations where there was no other available method (USP38-NF33 <61> 2015.

Table 2.11 Reflects the stipulated steps for each method according to USP38-NF33

Chapter <62> 2014.

2.4.1.3. USP Results Interpretation

TAMC was considered to be equal to the number of CFU found using SCDA agar.

If fungal colonies were detected in the media, these were counted as part of the

TAMC.

The total count of yeast and fungi (TYMC) was considered part of the number of

CFU using agar SDA.

If bacteria colonies were detected in the media, these were counted as part of the

TYMC.

When TYMC was expected to exceed the acceptance criteria due to bacteria

growth, use Sabouraud dextrose agar containing antibiotic.

If the count was performed through the MPN method, the resulting calculated value

was TAMC.

When an acceptance criteria for microbiological quality was recommended, it was

explained as follows

10¹ CFU: maximum aceptable count = 20 CFU

10² CFU: maximum aceptable count = 200 CFU

10³ CFU: maximum aceptable count = 2000 CFU

63

2.5. USP 38-NF33 Chapter <62> Microbiological Examination of Non-sterile

Product: Tests for Specified Microorganisms

USP38-NF33 Chapter <62> describes the test for the detection of specific

organisms. The test was designed primarily to determine when a substance or preparation

meets the established specifications of microbiological quality. This chapter presents

alternative microbiological methods, including automated methods which may be used

and will always be accepted as demonstrably equivalent to the stipulated method by

pharmacopoeia. It further notes specific selective media to be used for determining the

following target organisms: E. coli, Salmonella spp., P. aeruginosa, S. aureus, C. albicans

and Clostridia spp.

2.5.1. Product Analysis

Table 2.12 presents the specific organisms and the culture technique utilized for

each of the organisms using selected culture media. It also shows the parameters for the

results interpretation.

64

Table 2.12. Test for the Absence of Specified Microorganisms.

Organism Test/Culture Medium Results Interpretation

Escherichia coli

MacConkey broth – incubate at 42ºC 24-48 hr Subculture in MacConkey agar– incubate 30-35ºC 18- 72 hr

Growth-can indicate positive Confirm with its identification

Salmonella spp Rappaport Vassiliadis Salmonella enrichment Broth – incubate at 30-35ºC 18-24 hr Subculture in xylose Lysine deoxychocolate Agar – incubate at 30-35ºC 18-48 hr

Well-developed colonies, color red or without black center-can indicate positive Confirm its identification

Pseudomonas aeruginosa

Subculture in Centrimide agar– incubate at 30-35ºC 18-72 hr

Growth – can indicate positive Confirm its identification

Staphylococcus aureus

Subculture in Mannitol Salt agar – incubate at 30-35ºC 18-72 hr

Growth with White or yellow colonies with a yellow zone – can indicate positive Confirm its identification

Clostridia spp. Reinforced medium for Clostridia – incubate in anaerobic conditions at 30-35ºC 48 hr Subculture in Colombia agar - incubate under anaerobic conditions at 30-35ºC 48-72 hr

Growth of low bacillus under anaerobic conditions with or without spores with negative catalase can indicate positive. Confirm its identifiction

Candida albicans Saburoud dextrose agar – incubate at 30-35ºC 24-48 hr

Colonies are large, whitish , round and moist can indicate positive Confirm its identification

65

2.6. Advantages and Disadvantages of the Techniques Described by

USP38-NF33 <61>

The pharmaceutical industries for non-sterile products must ensure the decrease

in the microbiological load of final dosages of product, raw material, and process

formulation. The criteria of acceptance established for non-sterile product Chapter <1111>

(USP 2015) is based on the technique of counting the total number of aerobic

microorganisms (TMAC) and the combination of yeasts and fungi counts (TYMC) as the

USP38-NF33 (2015) establishes. These specifications change in accordance with the

presentation and use of the product. The rapidity and alternatives for the "screening" in

the microbiological analyses will depend on the technique used, the target organism, and

the economic factor, which can determine what methodology appears to be viable for

routine use in the pharmaceutical environment (van der Zee and Huis in’t Veld 1997).

Cundel (2006) suggested that the pharmaceutical industry justify microbial

identification programs, including, selection of rapid, easy, and effective methods for

species identification. This would successfully benefit investigations of failure products.

He also suggested evaluating methods used in microbiological laboratories with precise

methods, such as the methodology of molecular genetics for bacteria and fungi. Samadi

et al. (2007) agreed that conventional techniques comprise the use of selective means,

examination of microscopic colonies, and biochemical tests, but that the industry should

move to molecular techniques for the optimization of the laboratory analyses.

Counting methods in Petri dishes such as the technique of pour and spread-plate

technique following USP38 <61> are methods that lack flexibility to be useful during any

moment or situation. It was necessary to evaluate these skills and apply a mathematical

model to achieve a better interpretation of results (Kai 2004). This involves an increase

in the percent of errors because this technique involves a lot of manipulation, growth

factors, incubation, and interpretation using mathematical conversion, each which can

66

affect negatively the test results. These methods consume a lot of time during, previous

and post preparation, to obtain a final result. Molecular methodology provides much more

rapid and accurate identification and helps the understanding of the type of organisms in

the industry’s laboratory (Jiménez 2011). This makes the process more efficient because

it provides a rapid evaluation of the product and its process. Table 2.13 presents the

summary of the advantages and disadvantages of the techniques described in the USP38

<61>.

67

Table 2.13. Advantage and disadvantages of the techniques described in the USP38<61>.

Technique Advantage Disadvantage

Filtration per membrane

Easy to perform

There are less steps of preparation than other methods.

Membrane can break.

Possible loss of sample when rinsing membrane.

Difficult to count the colonies.

Colonies usually appear colorless

Method of platecount

pour plate

Surface spread plate

Requires mastery of skills in basic microbiological techniques for analysis execution.

It was the traditional approach currently microbiology curriculum in the academic area.

Currently the most widely used technique for TSM.

To perform the analysis various equipment are required.

Takes much preparation of sterilization of material and glassware.

Takes excessive preparation of culture medium and solutions.

Involves much manipulation before, during, and after the test.

Takes days and weeks to see results.

Requires post tests after incubation period.

For the identification and determination of other species that are not listed among the specific organisms, highly qualified personnel are required for its identification.

Most-Probable-Number (MPN)

Its interpretation is confusing.

Not frequently used.

It is not dependable.

The accuracy of the method is lower than the filtration technique or plate count.

No reliable results are obtained especially for mold count.

The MPN is reserved for counting TAMC in situations where no other method available (USP38 <61>).

The choice of method was based on factors such as the nature of the product and the

required microorganisms (USP38-NF33 <61>. 2015).

68

2.7. Applications Present and Future

In the Real-time qPCR method, the advantage was that the processes of

amplification and detection take place simultaneously in a closed microtube without the

need for later action (Livak and Schmittgen 2001). It combines the amplification of PCR

and detection in only one step, thus eliminating the need to detect the product of PCR in

an electrophoresis gel. The product increases exponentially with the number of cycles.

Using fluorescence for detection, measures the quantity of DNA synthesized at the time

the reaction happens. For this reason, it was referred to as Real-time because it was at

that time when synthesis occurs. This was possible because the fluorescence emission

was proportional to the quantity of DNA formed. Consequently, this allows detection and

registration of the kinetics of the amplification (Bustin et al. 2009). The Real-time PCR

equipment has a thermocycler to changes temperature, depending on the stage of the

process. The thermocyclers perform PCR during real-time by incorporating a fluorescent

reader designed to measure changes at any time and to emit fluorescence for each of the

wells where the amplification was performed. The Real-time PCR fluorescent detection

systems can be of two types; intercalating agents or specific probes marked with

fluorochromes, designed in a specific way.

The fluorescence detection system was by hydrolysis probe, which were marked

with two types of fluorochromes. One does the donor's function and other the acceptor.

This process was based on the fluorescent transference across the resonance (FRET)

between two molecules (Valasek 2005). The fluorescence detection most used are

hydrolysis probes, also called TaqMan probes, molecular beacon probes, and FRET

probes. The pilot phase of the Real-time PCR runs for this research used hydrolysis and,

TaqMan® probes. The hydrolysis probes were oligonucleotides marked with a donating

fluorochrome in the 5' end of the molecule that expresses fluorescence, having been lit

and an acceptor in the 3' that absorbs the fluorescence liberated by the donor, with the

69

donating molecules and acceptors located spatially close. The emission spectrum of the

first overlaps the absorption spectrum of the second. As long as the probe was intact, the

fluorescence emitted by the donor is absorbed by the acceptor. However, during the

amplification of target DNA, the probe hybridizes its complementary strand. When moving

along the chain during, synthesis, Thermus aquaticus DNA polymerase having 5'

exonuclease activity hydrolyzes the free 5' end of the probe, and thus causes release of

the donor fluorophore. Because the donor and acceptor were spatially far apart, the

fluorescence emitted from the first was collected by the reader (Kubista et al. 2006).

The Real-time qPCR comprises a thermocycler and fluorescence reader. These were

designed to perform the reading of the fluorescence emitted by each of the wells used at

any time of the reaction (Vinuza-Burgos 2009). In this research, the molecular runs were

performed using the StepOne™ Real-time PCR System from Applied Biosystem through

Bioanalytical Instrument. The differences between the devices lie in the rapidity of the

production capacity and results in the number of samples that can be processed

simultaneously and the number of channels that detect the emission of both

fluorochromes. Consequently, one can use several probes labeled with different

fluorophores, to identify different types of target DNA in the same reaction (multiplex PCR)

or internal controls to incorporate reaction to detect the presence of certain inhibitors

(Dorak 2006).

2.8. Advantages of the Real-time PCR

The advantages provided by the Real-time PCR methodology in this research were

directed towards two (2) areas: options presented by the equipment and advantages in

the technique.

70

2.8.1. Options of the Real-time PCR Equipment’s

The Real-time PCR equipment software equipment provides options, such as: (1)

amplifying and detecting the DNA or RNA in the sample, (2) performing multiple PCR runs,

(3) quantifying the target DNA or RNA sample, and (4) conducting analysis of dissociation

curves.

2.8.2. Advantages of the Real-time PCR Technique

The main advantage of the Real-time PCR equipment is its ability to rapidly monitor

the progress of the PCR reaction as it occurs in real time (Bustin 2009). This methodology

does not require any additional process for the detection of DNA or RNA. If the computer

is Light Cycler, it can complete the process ranging from 30 to 40 min, thus saving time

and allowing a greater flow of samples and test runs. Its ability to precisely measure the

amount of amplicons (DNA fragments) at each cycle, allows highly accurate quantification

of the amount of starting materials in samples. Another advantage is that when testing in

closed systems, the risk of contamination is minimized. Amplification and detection occurs

in a single tube, eliminating post PCR manipulations. The Real-time PCR quantifies the

initial concentration of DNA present in the samples by being more practical, easy, and

accurate than conventional methods. Real-time PCR equipment has a high capacity to

perform multiple qualitative, quantitative determinations of mutations, and PCR tests,

among others using the same equipment. However, under conventional methodologies,

many types of equipment are required to perform the test (Valasek 2005).

71

Table 2.14. Advantages and disadvantages of the Real-time PCR.

Advantages

Disadvantages

Product detection occurs

in each reaction cycle

allowing analysis of the

reaction kinetics. Ease of

optimization.

High sensitivity and

specificity

Faster results

High reproducibility

Lower contamination risk

Facilitates workflow

No post-PCR handling:

reduces the risk of

contamination.

Precise quantification of

DNA or RNA

Requires small entry

amounts of DNA or RNA

Improved performance

Detection of more than

one specific product from

the same reaction.

Using specific probes is

more expensive at first,

but it involves long-term

savings.

It has technical limitations

inherent in any trial.

There are various

conditions that affect the

process of binding or

hybridization between the

probe and target

segment, such as

temperature, salt

concentration and pH of

the reaction.

Non-specific products

(artifacts) can increase

the value of fluorescence

obtained during the

reaction. This was more

common when using

SYBR-green dyes.

72

Table 2.14. continued

Minimizes experimental

variations.

Cost effective

Decreases biological

waste.

Probe should be designed

differently for each target

to study.

Specific probes are more

expensive.

2.9. Real-time PCR Comparison versus Microbiological Methods

The advantages and disadvantages of the Real-time PCR method versus the

traditional microbiological method have been evaluated from the technical, economic (in

terms of time/cost) and the regulatory aspects. Microbiological analysis methods usually

fall into three broad categories, Including molecular biology, biochemistry, and

microbiology. Molecular methods in general cover a broad range of techniques based on

DNA analysis and discrimination of the microorganism. This technique eliminates the time

required to grow and the intervals associated with the growth and development of the

culture.

Conventional microbiological methods are those that have more variety and are

less useful for characterizing consortium of communities (Spiegelman et al. 2005). Such

methods are based on the traditional tools of cell count, use of selective media, and

microscopic examination to provide the general characteristics of a community of

microorganisms as a whole. Many of microbiological tests reflect fast and inexpensive

procedures to create a profile of the organism under study. Conventional microbiological

methods are useful in making a screening of the organism (de Boer and Beumer 1999).

73

Table 2.15 presents a summary of the advantages and disadvantages of

conventional microbiology methods.

Table 2.15. Advantages and disadvantages of conventional microbiology methods.

Advantages Disadvantages

Uses basic

microbiology

skills

Phenotypic properties (biochemical, carbon sources) can be

variable, subjective, and dependent on the growth parameters and

body health.

Method most

used

The cellular fatty acid profile changes with temperature, age of the

culture, and growth medium.

Some identification systems require subjective tests as gram

staining, oxidase, and coagulase before the final determination or

determination of the appropriate card or miniaturized system.

Generates large amounts of biological waste.

2.10. Taxonomic Change of Aspergillus niger to Aspergillus brasiliensis

2.10.1. Morphologic Characteristics of Genus Aspergillus

The mitosporic genus Aspergillus was characterized by production of specialized

hyphae called conidiophores located on the conidiogenous cells which generate

asexualspores or conidia (Figure 2.03) (Abarca 2000).

74

Figure 2.03. Morphological structures of the genus Aspergillus. A-B: conidiophores,C-D:

conidial heads. (Retrieved from Abarca 2000).

The name A. brasiliensis refers to the locality where the culture was originally

isolated. Colony diameters at 7 days: Czapeck Yeast Agar (CYA) at 25⁰ and 37⁰C, and

Czapek Yeast Autolysate with 5% NaCl (CYAS) at 25C: 71–76 mm; Malt Extract Agar

(MEA) 52–70 mm; Yeast Extract Agar (YES) 75–80 mm; Oatmeal Agar (OA) 32–36 mm;

Creatine Agar (CREA) 32–44 mm, poor growth, strong acid production. Colony first growth

white then dark brown to black (Fig. 2.01). Exudates absent, reverse cream-colored to

light brown color. Conidial heads usually globose at first and later radiate, occasionally

developing into several conidial columns; stipes 700–170068 – x13 μm, walls thick,

smooth, pale brown; vesicles 30–45 μm wide, almost globose; biseriate; with metulae

covering virtually the entire surface of the vesicle, measuring 22–3063–6 μm; phialides

flask-shaped, 7–963–4 μm; conidia are subglobose, 3.5–4.8 μm in diameter, and

75

echinulate, with no sclerotia observed in the culture extype. All isolates produced several

naphtho-c-pyrones (including aurasperone B), tensidol A and B and pyrophen (Vargas et

al. 2007).

Figure 2.04. Aspergillus brasiliensis sp. nov. CBS 101740T. (a) Colonies on CYA; (b)

colonies on OA; (c) colonies on MEA; (d–g)conidiophores; (h) conidia under light

microscope; (i) conidia as seen using SEM. Bars, 10 mm (d–h) and 5 mm (i). (Adapted

from Vargas et al. 2007).

76

Figure 2.05. Colony morphologies of type strains of species assigned to Aspergillus

section nigri grown on CYA and MEA plates at 25 °C for 7 d. (A–B) A. aculeatinus, (C–D)

A. aculeatus, (E–F) A. brasiliensis, (G–H) A. carbonarius, (I–J) A. costaricaensis, (K–L) A.

ellipticus, (M–N) A. foetidus, (O–P) A. japonicus, (Q–R) A. heteromorphus, (S–T) A.

homomorphus, (U–V) A. ibericus, and (W–X) A. lacticoffeatus. (Adapted from Samson RA,

Noonim P, Meijer M, Houbraken J, Frisvad JC and Varga J. 2007).

77

2.10.2. Aspergillus: Importance in Industry, Agriculture and Medicine

Different Aspergillus spp. are abundantly distributed in nature. The genus

Aspergillus consists of a large number of species, including several opportunistic

pathogens (e.g. A. fumigatus, A. terreus), toxin producers (e.g. A. flavus, A. parasiticus)

and industrial species (A. niger, A. aculeatus, A. oryzae). The genus was separated into

several sections, such as the yellow and the black aspergilli. The black aspergilli

(Aspergillus section nigri) are cosmopolitan, and contain species commonly found in

industrial contamination (Meijer et al. 2011). These can be isolated from a variety of

substrates. The simple dispersion of conidia and their small size, facilitates suspension

in air for extended periods. Because of this, humans are frequently exposed to this mold

by inhalation.

Different Aspergillus spp. are the most common cause of invasive fungal

infections. These frequently turn out to be fatal in immunocompromised patients (Fridkin

and Jarvis 1996; Trick and Jarvis 1998). Although Aspergillus fumigatus was the most

common etiologic agent in this type of fungal infection, other species of the genus such

as A. flavus, A. terreus, A. niger, and A. nidulans (Emericella nidulans) are also considered

responsible for invasive infections (Abarca 2000). In addition to its role as an opportunistic

pathogen, this genus is important because it is an organism used in fermentation

processes for the production of citric acid. This genus contains several species of positive

or negative economic importance in industries, such as agriculture and medicine. Most

aspergilli, including species of economic importance, reproduced only by asexual spores.

Recently, genome projects have been completed for the following Aspergillus species: A.

fumigatus, A. nidulans, A. niger and A. oryzae. Several other species are also being

sequenced. Information from these genome projects will continue to be useful in

interpreting aspects of the evolution of sexuality, phylogeny, and the extent of secondary

metabolite diversity (Bennett 2009). The black aspergilli are an important group of species

78

in food mycology, medical mycology and biotechnology (Gams et al. 1985). Many species

cause food spoilage. Others are used in the fermentation industry to produce amylases or

lipases, which are hydrolytic enzymes and citric acid and gluconic acid, which are organic

acids (Jurjević et al. 2000). They are also candidates for genetic manipulation in the

biotechnology industries. Aspergillus niger used under certain industrial conditions has

been granted the status, :“generally recognized as safe” (GRAS), by the Food and Drug

Administration of the US government (Vargas et al. 2007).

Aspergillus can be detrimental in two ways: (1) directly as an opportunistic

pathogen causing aspergillosis, and (2) indirectly due to aflatoxin production generated in

food products that can lead to aflatoxicosis.

2.10.3. Taxonomy: Approaches to Distinguish A. niger from A. brasiliensis

The taxonomy of fungal species, similar to many other microorganisms, suffers

frequent revisions due to the discovery of new species and to the development and

gathering of characterization data and morphological information. The morpho-taxonomy

helps in the identification of many species. The macro and micro-morphological, including

spectral mass analyses for the phenotypic characterization of 13 Aspergillus section Nigri,

characterization of conidia (spores) is done with an scanning electron microscopy. It was

useful to discriminate the key morphological characteristics of the conidia and to separate

closely related fungi (Simões et al. 2013). A variety of methods have been proposed by

the literature to detect fungi in minimal concentrations in early stages of the laboratory

analysis.

These molecular methods include PCR, and/or a combination thereof, with certain

techniques which are useful at detecting Aspergillus. Methods such as RSIC, C-probe and

probe with pyro sequencing or investment DirectSS/dsDNA detection have been used to

identify both pathogenic bacteria and fungi. This would help develop an older standard for

the detection of Aspergillus spp. (Abdin et al. 2010). Invasive aspergillosis was a major

79

cause of morbidity and mortality in immunocompromised and critically ill patients.

Standard culture methods for the diagnosis of Aspergillus infections have limited

sensitivity and specificity and are time consuming. The recent availability of novel

molecular based diagnostic techniques offers the potential of rapid, sensitive and specific

pathogen detection (Faber et al. 2009). With the increasing incidence and mortality of

fungal infection, the requirements for strict diagnostic approaches was now an urgent

issue.

Traditional screening techniques, such as cultivation, provide a poor input of

diagnosis (Faber et al. 2009). Therefore, expectations are set that molecular biologically

techniques have the potential to develop a diagnostic approach in the pathogenicity of the

fungus (Li et al. 2004). The definite and rapid diagnosis of invasive aspergilosis is

necessary because of the high mortality caused. The real-time PCR assay provides a high

sensitivity and specificity for detection of fungal DNA and rapidly identifies most of clinically

relevant Aspergillus species (Ramírez et al. 2009).

2.10.4. Reclassification of Strains ATCC® 16404™ from Aspergillus niger to

Aspergillus brasiliensis

Since 1965 the taxonomy of the Aspergillus spp. has been mainly governed by

description by Raper and Fennell (1965). There have been major changes in the taxonomy

of Aspergillus species including their telemorph states (Abarca 2000; Geiser et al. 2007).

Black aspergilli was one of the most difficult groups concerning its classification and

identification. New molecular approaches have shown that there was a high biodiversity,

but that species are occasionally difficult to recognize based solely on their phenotypic

characters (Samson et al. 2007). During a study of the genetic relationships among black

aspergilli collected worldwide, four isolates have been identified which did not fit into any

of the currently accepted 19 species of Aspergillus section Nigri (Samson et al. 2007;

Noonim et al. 2008, Perrone et al. 2008). Any proposed new species should show

80

evidence for evolutionary divergence from other taxa, particularly unique DNA characters

at multiple loci. The polyphasic approach was suggested as the ‘gold standard’ for species

delimitation using a combination of multilocus sequence data, combining morphological

physiological characteristics with ecological data (Samson et al. 2009).

The ATCC®16404™ strain, known as A. niger, has been designated as a reference

strain for quality control in a number of fungal reference strains widely used in clinical

applications and for industrial testing. These fungal isolates are cited in a number of official

methods (eg, Harmonized Microbial Limits USP <61> and Sterility Tests) and manuals, as

well as the Code of Federal Regulations. The taxonomic and phylogenetic report clear

placement of these two black aspergilli strains, ATCC® 9642™ and ATCC ®16404™, as A.

brasiliensis, were with previously classified as A. niger (Houseknecht et al. 2008).

Furthermore, the ATCC® 9642™, deposited as Aspergillus niger, also being renamed as

A. brasiliensis. Sequence alignment has revealed that all lots of 9642 and 16404 are

identical, but 16888 differ from 9642 and 16404 in five positions (Table 2.16).

Table 2.16. The difference in ITS sequences.

Position* 140 166 172 173 384

Aspergillus niger type strain ATCC® 16888™ A T A A Deleted

ATCC® 16404™ C C T T T

ATCC®9642™ C C T T T

Aspergillus brasiliensis type strain IMI 381727 C C T T Deleted

Note: * The first C in the border sequence (TTACCG) of 18S and ITS1 was here defined as position 1.

Table 2.14 indicates that there was approximately 1.0% difference base pair-wise

within the ITS region between the Aspergillus niger type strain and the other two black

aspergilli ATCC®16404™ and ATCC®9642™. Differences of this magnitude typically

81

raise the question whether ATCC®16404™ and ATCC®9642™ can be adequately

considered as A. niger (Houseknecht et al. 2008).

These results demonstrated that ATCC®16404™ and 9642™ are distinct from A.

niger ATCC®16888™ and form a separate clade with A. brasiliensis strain IMI381727

(Figure 2). Therefore, the commonly used fungal barcoding sequences indicate that

ATCC®16404™ and 9642™ are A. brasiliensis, not A. niger, indicating substantial

differences, genotypically and phenotypically, between the type strain of A. niger

(ATCC®16888™) and the other two widely used fungal reference strains, ATCC®9642™

and 16404™.

Figure 2.06. A Neighbor Joining Tree of Black Aspergilli based on Their ITS DNA

Sequences. (Adapted from: Houseknecht et al. 2008).

These results demonstrated that ATCC®16404™ and 9642™ are distinct from A.

niger ATCC®16888™ and form a separate clade with A. brasiliensis strain IMI381727

(Figure 2.06). Therefore, the commonly used fungal barcoding sequences indicate that

82

ATCC®16404™ and 9642™ are A. brasiliensis, not A. niger (Houseknecht et al. 2008). The

sequence data for A. brasiliensis ATCC®16404™ was 18S ribosomal RNA gene, partial

sequence; internal transcribed spacer 1), 5.8S ribosomal RNA gene, and ITS 2, complete

sequence; and 28S ribosomal RNA gene, partial sequence (Figure 2.07).

Aspergillus brasiliensis ATCC®16404™ 18S partial gene sequence

TGATATGCTTAAGTTCAGCGGGTATCCCTACCTGATCCGAGGTCAACCTGGAAAG

AATGGTTGGAAAACGTCGGCAGGCGCCGGCCAATCCTACAGAGCATGTGACAAA

GCCCCATACGCTCGAGGATCGGACGCGGTGCCGCCGCTGCCTTTCGGGCCCGTC

CCCCCGGAGAGAGGGGACGGCGACCCAACACACAAGCCGGGCTTGAGGGCAGC

AATGACGCTCGGACAGGCATGCCCCCCGGAATACCAGGGGGCGCAATGTGCGTT

CAAAGACTCGATGATTCACTGAATTCTGCAATTCACATTAGTTATCGCATTTCGCTG

CGTTCTTCATCGATGCCGGAACCAAGAGATCCATTGTTGAAAGTTTTAACTGATTG

CAAACAATCGACTCAGACTGCACGCTTTCAGACAGGGTTCGTGTTGGGGTCTCCG

GCGGGCACGGGCCCGGGGGGCAGAGGCGCCCCCCCGGCGGCCGACAAGCGGC

GGGCCCGCCGAAGCAACAGGGTACAATAGACACGGATGGGAGGTTGGGCCCAAA

GGACCCGCACTCGGTAATGATCCTTCCGCA

Figure 2.07. The sequence data for A. brasiliensis ATCC®16404™ was 18S ribosomal

RNA gene, partial sequence.

The pharmacopoeia list a number of microorganisms to be used in the compendia

of microbiological tests for confirming the growth-promoting, indicative, and inhibitory

properties of the media and demonstrating the suitability of the test for a specific test

article. Major national culture collections are specified as the sources for these test strains

83

based on their history of deposition and maintenance and use in the compendia tests.

Using these microorganisms, it has long been assumed that these strains are

interchangeable and that sourcing the strains from different culture collections has no

impact on the result of the media quality control and method qualification tests (Cundell et

al. 2010). The black aspergilli are one of the more difficult groups concerning classification

and identification, and several taxonomic schemes have been proposed. New molecular

approaches have shown that there was a high biodiversity, but that species are

occasionally difficult to recognize based solely on their phenotypic characters (Samson et

al. 2007). For species descriptions it was recommended to examine several gene

sequences (e.g., ITS, calmodulin, β-tubulin, and actin) and submit them to recognized

sequence databases (Samson et al. 2009).

Significant advances have been made to identify clinically important molds.

Although molds have historically been identified using observations of colonial and

microscopic morphology, this time-consuming procedure requires expertise and can be

problematics for determining species identification. But especially in the case of the

management of high-risk patients, DNA-based identification are increasingly employed in

clinical laboratories. Methods such as the GenProbe® assay, are based on the polymerase

chain reaction such as single-step PCR, RAPD-PCR, rep-PCR, nested PCR, PCR-RFLP,

PCR-EIA, and microarray-based, Luminex technology-based, and Real-time PCR-based

methods. Because of the complexity and variation of target organisms, many methods are

slowly being validated. However, DNA sequencing chemistry using comparative DNA

sequence analysis was an attractive tool for fungal identification. This methodology can

be developed within the laboratory or purchased commercially. These methods can be

useful, but depend largely in the reliability and breadth of databases used for comparison

(Balajee et al. 2007). Molecular biological techniques can be used to investigate the

biological properties and mechanisms of pathogenic fungal infections and antifungal

84

studies (Li et al. 2004). The Real-time PCR assay provides a high sensitivity and

specificity for detection of fungal DNA and rapidly identifies most of clinically relevant

Aspergillus species (Ramírez et al. 2009).

85

Chapter Three

Materials and Methods

This research study focused on testing for microbiological examination OTC

pharmaceutical products in oral and ectopic areas. Two different methodologies for test

specified microorganisms (TSM) were compared. Microbiological conventional pour- plate

methodology established by the USP38 with molecular Real-time PCR method was

applied. The comparison between the two methods was based on the detection in terms

of sensitivity and high specificity in the presence and absence of specific organisms in a

pharmaceutical sample. The organisms used as contaminants to pharmaceutical samples

were the strains of Escherichia coli ATCC®8739™ and Aspergillus brasiliensis

ATCC®16404™. The samples analyzed were composed of three raw materials and three

OTC pharmaceuticals products. This study focused on demonstrating the capacity of each

method to demonstrate the minimum detection in the presence of contamination to levels

of traces of the organisms.

The test was carried out under controlled conditions using aseptic techniques,

laminar flow hood (LFH), use of sterile materials, equipment sanitation, and environmental

monitoring during the test and control group in each experimental run. All analyzes were

performed on a LFH to prevent microbial contamination. Environmental monitoring was

performed with exposure of plates containing culture media SCDA and SDA during

handling and testing. Precautions were taken to avoid contamination using a control group

during the test to prove that microorganisms were revealed in the tests were not affected.

3.1. Microbiological Analysis

3.1.1. Identification Method

Pour-plate technique was used as enumeration method on plate count (TEM).

Thepetri dish used as exposure plate technique, and for the test (TSM) was 100 x 20mm.

86

The conventional method of selection was based on the factors of the nature of the

product, analyzed raw material required, and the acceptance limits of microorganisms

(USP 2015).

Pharmaceutical products used for analysis were finished products representations

of OTC tablet, liquid, and cream. The raw materials analyzed were: Corn Starch, Sodium

Chloride and Sucrose.

3.1.2. Negative Control

To verify the testing condition, a negative control was performed using the chosen

diluent phosphate buffer solution in place of the tested article. There must be no growth.

3.1.3. Growth Promotion Test

A growth promotion test for each culture medium batch and buffer solution used in

the research was conducted. With this test, the ability of the media and solutions used to

grow and regain the inoculated organisms was demonstrated. The batch used Soy Casein

Digest Agar (SCDA), Soy Casein Digest Broth (SCDB), and phosphate buffer solution that

were inoculated with NTM (not more than) 100 CFU of E. coli strain ATCC®8739™ and

incubated at 30-35°C for three days. The batch of Saboraud Dextrose Agar (SDA),

Saboraud Dextrose Broth (SDB), and Phosphate Buffer Solution were inoculated with

NMT 100 CFU of A. brasiliensis strain ATCC®16404™ and incubated 20-25°C for 5 days.

At the end of the incubation time for each organism, plate count was taken, and the results

were scored. The results of growth on the plates according to USP <62> did not differ from

the original inoculum. Solid growth medium results obtained did not differ by a factor

greater than 2 from the calculated value for standardized inoculum.

3.1.4. Suitability of the Counting Method in the Presence of Product

A total of 10g or 10mL of each finished product and raw material were individually

dissolved and transferred to 90mL of phosphate buffer solution at pH 7.2. Triplicate

87

samples were used in duplicate for the three OTC products and the three raw materials.

Each tube containing the finished product and raw materials was contaminated with 1mL

of not more than 100 CFU E. coli and A. brasiliensis respectively. The E. coli-contaminated

tubes were incubated at 30-35°C for three days. The tubes contaminated with A.

brasiliensis were incubated at 20-25°C for five days. Once the incubation time for each

organism was completed, the tubes were transferred to a Petri plate using the pour-plate

method and counting plate method. This was done in triplicate for each OTC and raw

material for each organism studied. The OTC pharmaceutical products and raw aterials

used in this research fell into the classification of water soluble products.

3.1.5. Specified Microorganisms

The specific organisms used were the strains of E. coli ATCC®8739™ and A.

brasiliensis ATCC®16404™ for the quantitative enumeration as a representation of

mesophilic bacterial and fungal growth under aerobic conditions. The experimental phase

was conducted concurrently using the USP pour-plate conventional methodology and

Real-time PCR Step One Plus™ System from Applied Biosystems® as a molecular

method.

3.1.6. Preparation of Test Strains

The viable microorganisms used for inoculation were not more than five transfers

or passages removed from original master seed-lot (USP38 2015). The microorganisms’

test-strain growth was prepared with a suspended colony sample of each organism in

enriched culture medium SCDB for E. coli and SDB for A. brasiliensis. The test strain for

A. brasiliensis was incubated separately on SDB at 20-25ºC for 3 days. Once developed,

both inoculum mother culture vigorous vortexes were applied to each test tube containing

their corresponding mother inoculum. A quantity of 1mL of E. coli mother inoculum was

transferred to each of two duplicate test tubes, each containing 9 mL of phosphate buffer

88

solution at pH 7.2 and incubated for 24 hr at 30-35°C. Then A. brasiliensis was transferred

to 1 mL of inoculum mother to two test tubes (duplicate) each containing 9 ml of phosphate

buffer solution at pH 7.2 with 0.05% polysorbate 80 and incubated for 24 hr at 20-25°C.

These inoculums in the study represent the 10º dilution.

3.1.7. Inoculum Preparation - Determination of the Population Known to

Contaminate Pharmaceutical Sample

From the original inoculum of 100 E. coli and A. brasiliensis, a dilution 1:10 was

performed with a total of 15 test tubes. The 15 tubes comprised from 10-¹ to 10-15 for each

organism with each tube containing 9 mL of sterile phosphate buffer was placed in a rack

for E. coli and A. brasiliensis. An additional tube was labeled as negative control, only

containing 9 mL of phosphate buffer solution at pH7.2. The 100 seed inoculum containing

E. coli ATCC®8739™ was vigorously vortexed. Using a sterile pipette, 1 mL of the inoculum

was removed from tube 100 and transferred to a test tube labeled 10-¹ mL. The used

pipette was discarded. The dilution of 10¯¹ was vortexed and immediately processed. With

another sterile pipette, 1 mL of tube 10-¹ was transferred to tube 10-2. The used pipette

was discarded and transferred for serial dilution; this procedure was continued until tube

10-¹5 mL. The same process was followed in serial dilution for A. brasiliensis

ATCC®16404™.

After the serial dilution series of each organism, a plate-count was performed.

Fifteen sterile Petri dishes were labeled from 10ˉ¹ to 10ˉ15 in triplicate for each dilution. An

additional sterile Petri dish was labeled as a negative control in triplicate. The negative

control contained only culture medium. The tube labeled 10-1 was thoroughly vortexed, 1

mL solution containing E. coli extracted, and transferred to a sterile dish labeled 10-1. The

pour-plate method was immediately performed. A total of 20 mL of the culture medium

plates were added, each containing SCDA for E. coli and for A. brasiliensis dishes

89

containing SDA. A circular motion was applied to homogenize the 1 mL that was added to

the culture medium. The 15 steps to triplicate tubes in both E. coli to A. brasiliensis were

repeated. The pour-plate method was also performed in triplicate for the negative control

and inoculum 100 of each specific organism. Escherichia coli triplicates were incubated at

30-35°C for 3 days. A. brasiliensis triplicates were incubated at 20-25°C for 5 days.

Reading of the count of E. coli colonies for 24 and 48 hr and for A. brasiliensis at 24, 48

and 96 hr were performed. Known populations were chosen for each organism ranging

from 10 to 100 CFU/plate.

3.1.8. Preparation of the Pharmaceutical Sample

The method that was used for the preparation of the sample depended on the

physical characteristics of the type of product that was analyzed for the test for specified

organism (TSM test). Selected samples of raw materials (corn starch, sodium chloride

and sucrose) and finished oral product types (tablets, liquid, and cream) of were each

diluted into duplicate bottles containing 90 mL of phosphate buffer solution for each

labeled material, and performed in triplicate for each objectionable organism. For each E.

coli 10-⁷ bottle another, bottle was labeled as 10-15. The bottle 10-⁷ represented the dilution

chosen in E. coli representing the population from which the sample was contaminated.

The bottle labeled as 10-15 represented the last tube in the serial dilution process,

specificity analysis method. With 10-15 dilution for each specific organism, the sensitivity

of each method was shown for detecting the minimum level of the presence of traces of

the specific organism (E. coli ATCC®8739™ and A. brasiliensis ATCC®16404™)

concentration in a pharmaceutical sample.

For A. brasiliensis, two bottles containing 90 mL of phosphate buffer solution were

also labeled in triplicate for three OTC (over the counter) and three raw materials. A bottle

was labeled as 10¯³ and another bottle as 10¯15. The 10¯³ dilution bottle was chosen for A.

90

brasiliensis as the known population that OTC samples (tablets, liquid and cream) and

samples from contaminated raw materials (corn starch, sodium chloride, and sucrose).

The bottle labeled 10-15 represented the last series of dilutions during the sensitivity test

of the pour-plate method analysis and Real-time PCR. With the 10-15 dilution containing

the objectionable organisms, the sensitivity of each method was demonstrated for each

method to detect minimum levels contaminant traces (E. coli and A. brasiliensis) in each

sample analyzed for raw materials and pharmaceutical products.

Table 3.01. Selected dilutions for test for specified microorganisms (TSM).

Organisms Selected dilution known Population

Last dilution

Escherichia coli ATCC®8739™ 10-⁷ 10-15

Aspergillus brasiliensis ATCC®16404™ 10-³ 10-15

Table 3.01 shows the selected dilutions per objectionable organism to contaminate the

samples of the final product and raw materials.

During the preparation of the pharmaceutical sample, a sterile mortar was used to

pulverize, dissolve, and homogenize tablets into 90 mL phosphate buffer solution with a

7.2 pH. Ten grams or milliliters of the pharmaceuticals samples (solid, liquid and cream)

were added to 90mL phosphate buffer solution. The content was mixed using a analog

vortex mixer for homogenization of the solution. Once reached the solubility in the buffer

solution containing 10% w / v of the product, we artificially contaminated each sample of

raw materials and OTC product with E. coli (ATCC®8739™) and A. brasiliensis

(ATCC®16404™), respectively.

Escherichia coli was added to each bottle labeled 10ˉ⁷ 1 mL of the population

known as 10¯⁷ of E. coli and in the bottles labeled 10¯15 1mL of the dilution of the organism

91

corresponding to tube 15 for the test for specified organisms phase. The same was done

for A. brasiliensis. To the bottles labeled as 10¯³ runs of A. brasiliensis 1 mL of the

population known organism 10-³ and 10-15 labeled bottles was added 1 mL of the dilution

of the organisms belonging to the tube 15 of the stage specified organisms test of the

method. All runs were performed in triplicate. This made a total of three pharmaceutical

known populations contaminated by dilution tube 15 in triplicates. This does 2 bottles of

90 mL phosphate buffer solution containing 10g or mL of product + 1 mL contaminant in

triplicate to equal a total of 18 bottles per run with E. coli or A. brasiliensis per TSM test

for pharmaceutical product. There were also a total of 18 bottles of specific organisms

TSM run phase for raw materials. Table 3.02, shows the samples made by the described

organism.

Table 3.02. Amount of samples per objectionable organism.

Organisms Known population Dilution 10-15

Escherichia coli

ATCC®8739™

Known population 10-⁷

Tablet

Bottle 1

Bottle 2

Bottle 3

Liquid

Bottle 1

Bottle 2

Bottle 3

Cream

Bottle 1

Bottle 2

Bottle 3

Tablet

Bottle 1

Bottle 2

Bottle 3

Líquid

Bottle 1

Bottle 2

Bottle 3

Cream

Bottle 1

Bottle 2

Bottle 3

Aspergillus

brasiliensis

ATCC®16404™

Known population 10-³

Bottle 1

Bottle 2

Bottle 3

Bottle 1

Bottle 2

Bottle 3

Bottle 1

Bottle 2

Bottle 3

Bottle 1

Bottle 2

Bottle 3

Bottle 1

Bottle 2

Bottle 3

Bottle 1

Bottle 2

Bottle 3

Table 3.02 describes the contaminated samples for the TSM test according to

theobjectabionable organisms. These samples containing product which were artificially

92

inoculated with the organisms were incubated for 24 hr at 30-35ºC for bacteria and 20-

25ºC for fungi for 5-7 days.

3.1.9. TBC and TYMC Test: Plate Method

A quantity of 1.0 mL of the dilution of the product artificially contaminated with E.

coli and A. niger were individually transferred to each sterile Petri dish. An additional batch

was utilized without sample for negative control. The pour-plate method 20 mL of SCDA

for E. coli and SDA for A. brasiliensis (a warmer temperature to 45°C) was added to every

two Petri dishes. The plates were covered, performing a rotary movement to homogenize

the way with the inoculum, allowing it to harden at room temperature. As soon as the

media solidified, plates were inverted and incubated at 30-35°C for 3 days for bacteria,

and 20-25°C for 5-7 days of fungi. Continued to the period of incubation, the colonies of

every pour-plate method were counted, and the number of CFU was calculated for g or

mL of product. Colonies obtained in plates to confirm the identification of E. coli and A.

brasiliensis strains were identified.

3.1.10. Identification of the Preparation of the Inoculum

After the period of incubation, the broth of enriched SCDB containing E. coli was

streaked on MacConkey agar (MCA) and Eosine Methylene Blue (EMB). All these

selective media were incubated to 30-35⁰C for 48hr After 48hrs of incubation, the colonies

were streaked on the SCDA. Gram stain does to the isolated colonies to confirm bacteria

morphology. Traditional identification is performed using the biochemical manual system.

In samples for fungi, following the period of incubation of Sabouraud Dextrose

Broth (SDB) containing the inoculum of the product and the organism A. brasiliensis, a

transfer was made on SDA and incubated at 20-25ºC for a minimum of 7 days. After the

incubation period, the number of colonies were counted and identified to genus and

93

species levels using macroscopic and microscopic characteristics evaluation of the

organism applying the morphological characteristics and taxonomic manuals.

3.2. Molecular Method Real-time PCR

For the molecular phase Applied Biosystems Step One Plus™ Real-time PCR

System equipment was used. The StepOnePlus™ systems used fluorescent-based

polymerase chain reaction (PCR) reagents to provide: (1) Quantitative detection of target

nucleic acid sequences (targets) using Real-time analysis. (2) Qualitative detection of

targets using post-PCR (endpoint) analysis, and (3) Qualitative analysis of the PCR

product (achieved by melt curve analysis ocurring post-PCR) (Applied Biosystems 2014).

The equipment used was designed to work using 48 wells. The program used was the

Primer Express® Software version 3.0 for using TaqMan primers and probes for

performing the method of Real-time PCR.

Through this method, the ability to detect the presence or absence of specific

nucleic acid sequences for E. coli and A. brasiliensis was established using TaqMan®

Fast Universal PCR Master Mix 2X which comprised the steps of polymerization,

cleavage, strand displacement, and selected polymerization. The components in the

experiment included the nucleotide sequence of the target A. brasiliensis and E. coli

organisms. The method compared the samples under study in each run. The samples

analyzed were run on par with conventional microbiological runs. These were the

Sensitivity Test methods where dilutions ranged from 100 to 10-15 containing A. brasiliensis

and E. coli separately. The detection of low concentration of trace level objectionable

organisms with the TSM test organism were also presented. All runs with its endogenous

control were run to eliminate variations in the efficiency of the retro transcription. The

samples in the wells’ plates were performed in triplicate for each sample with its

endogenous per plate.

94

The method of analysis to interpret the test was based on the Comparative Ct

Method for Relative Quantification (ΔΔCt). The CT value is the threshold cycle, the cycle

number when the flourescence for the reaction crosses a threshold line. The CT values

are logarithmic and are used either directly as in the comparative CT method or indirectly

as in interpolation to standard curves to create linear values for quantitative analysis. The

software collected raw fluorescence data at different points after each extension step of

PCR. Each reading was composed of a three phases throughout the system:

Excitation – The instrument illuminated all wells of the reaction plate within the

instrument, exciting the fluorophores in each reaction.

Emission – The instrument optics collected the residual fluorescence emitted from

the wells of the reaction plate. The resulting image collected by the device

consisted only of light that corresponded to the range of emission wavelengths.

Collection – The instrument assembleed a digital representation of the residual

fluorescence collected over a fixed time interval. The StepOne™ software stored

the raw fluorescent image for analysis.

After a run, the StepOne™ System software used calibration data (spatial, dye, and

background) to determine the location and intensity of the fluorescent signals of each

measurement, the dye associated with each fluorescent signal, and the significance of

the signal (Applied Biosystem 2015).

3.2.1. Preparing a PCR Reagent

The reactions were prepared with the FG, TaqMan® Fast Universal PCR Master

Mix (supplied at a 2X concentration, p/n 4304437). The PCR Master Mix provides the

entire necessary reagent for the 5’ nuclease PCR process, with the exception of the

primers, the TaqMan® probe and DNA template. The preparation of the reaction mix is

shown in Table 3.03. The working stock concentration of the primers was 50µM and the

95

working stock concentration of probe was 10 µM. The primers and probes were designed

by Applied Biosystems® Custom Oligo Synthesis Services. The forward and reverse

primer and Probe used are described in Table 3.04. The reconstitution of the Forward and

Reverse primers to 10,000 pmol are shown on Figure 3.02. The dilutions of the TaqMan®

probe to 100 µM in 50 µL are indicated in Figure 3.03.

The DNA templates that were later added to both the primers and the probe to

provide a working stock concentration of 10 µL. For this research the total reaction

volumes were 50µl.

Table 3.03. Preparation of the reaction mix.

16 x 3 + 2 NTC = 50 samples per assay: 50 samples per assay (in μL)

TaqMan® Advanced Fast Master Mix 10.0 μL x 50 = 500

First forward 2.0 μL x 50 = 100

First reverse 2.0 μL x 50 = 100

Probe 2.0 μL x 50 = 100

cDNA template 4.0 μL x 50 = 200

Final Volume 20.0 μL

96

Table 3.04. Primers and probe per organisms.

Aspergillus brasiliensis

Forward Primer Reverse Primer Probe

GGGCCGCTGGCTTCTTA TGTTATTGCCGCGCACTTC 6 – FAM

ACTATCGGCTCAA

GCC MGBNFQ

Escherichia coli

CCGCGTGGTATGAAGAAAG CTTCCTCCCCCGCTGAA VICTTCGGGTTGTA

AAAGTACMGBNFQ

Table 3.04 presents the sequence detection primer 10.000 pmoles and the TaqMan®

MGM probes of 6.000 pmoles from Applied Biosystems used for A. brasiliensis and E.

coli.

3.2.2. Preparation and Extraction Samples for Escherichia coli

ATCC®8739™ and Aspergillus brasiliensis ATCC®16404™ Testing

To prepare the bacteria and fungi from the culture dilution, the cultures were

shaken by the PrepMan® Ultra Sample Preparation Reagent Well and the reagent settled

until any remaining bubbles had disappeared. We then transferred each appropriate

quantity of PrepMan® Ultra Sample Preparation Reagent into a 50 mL sterile conical tube.

Table 3.03 summarizes the reagents by reaction. The tubes were labeled and pipette filled

with 1 mL of culture broth.

This broth contained the bacteria or fungi in new 2 mL microcentrifuge screw-cap

tubes which were microcentrifuged at the highest speed for 2 min, aspired; the

supernatant was discarded using a VWR® Scientific disposable transfer micropipette with

the pipette 100-1000 µL. Using a 2 mL sterile microcentrifuge screw-cap tube, 100 µL of

97

the PrepMan® Ultra Sample Preparation Reagent were aseptically added into each tube.

Pipette tips were changed between tubes. After tightly screw-capping the microcentrifuge

tubes, the sample was vigorously vortexed and placed in a heat block and set to 100ºC

for 10 min, removed allowed to cool at room temperature for 2, and microcentrifuged at

the highest speed for 2 minutes. An 50 μL aliquot of the supernatant from the spun tubes

was transferred into a second set of microcentrifuge, screw-cap tubes. The remaining

supernatant was discarded. A 5 µL aliquot of supernatant was used for each assay

reaction. Finally, the samples were amplified using the thermal cycling protocol described

in Figure 3.01.

Table 3.05. Steps performed to extract the sample with PrepMan®.

Procedure

1. Centrifuged 1 mL of the sample 2 min at full speed.

2. Discarded supernatant.

3. Added 100 μl of PrepMan® Ultra.

4. Heated sample to 100⁰C for 10 min.

5. Removed 50 µL and stored at -20⁰C if the sample were not

analyzed immediately.

98

Figure 3.01. Computer screen template – Real-time PCR run method parameters.

3.2.3. Real-time PCR Amplification Process

For the preparation of the PCR amplification process, the DNA polymerase was

hot-satarted in the TaqMan® Universal PCR Master Mix (2X), No AmpErase UNG. This

amplified the target cDNA using sequence-specific primers and cleaved the TaqMan®

probed hybridized to the target sequence. The cleavage of TaqMan® probes caused by

the hot-start DNA polymerase generated the fluorescent signal. The plate document on

the StepOne™ Real-time PCR System was set up and the PCR reaction plate in triplicate

was prepared for each reaction with the recommended reaction volume of 20 µL for the

optical 48 well fast plate. Table 3.05 shows amounts of reagents used for each reaction.

Software for the Real-time PCR System showed the plate document that corresponded to

the reaction plate and the reaction plate was placed into the StepOne™ System, which

was started for the run. Figure 3.01 shows the run method parameters.

99

Table 3.06. Preparation of the 48-wells plate.

Procedure

Added 16μL reaction volume in each of the wells in the plate.

Added 4μL of each sample corresponding to each well.

Table 3.07. Reaction of Real-time PCR - Quantities of reagents by reaction.

Component Volume (μL)

TaqMan® Advanced Fast Master Mix

Primer Forward

Primer Reverse

Probe

cDNA template

Final Volume

10.0

2.0

2.0

2.0

4.0

20.0

Reconstitution volume

(10,000 pmol) (1 µmol / 1,000,000 pmol) = 0.01 µmol

0.01 µmol / X = 50 µmol / L = 0.0002 L = 200 µL

It was added 200 µL of nuclease free water to each test tube of lyophilized primer

to obtain a concentration of 50 µM in each tube.

Figure 3.02. Real-time PCR protocol used - Reconstitution of the forward and reverse

primers to 10,000 pmol.

100

Dilution performed

C1V1 = C2V2

100 µM = 100 µmol / L

100 µmol / 1,000,000 µL (50 µL) = 10 µmol / 1,000,000 µL (V2)

we cleared for V2 = 500 µL

500 µL - 50 µL = 450 µL

It was added 450 µL of 1X TE buffer to the tube of TaqMan® probe to dilute it to

10µm.

Figure 3.03. Protocol used for dilution of the TaqMan® probe to 100 µM in 50 µL.

Table 3.08. Quantities for reaction.

Reactive µL per

reaction

Reaction mix

multiply

# de Reactions + 1

Final Concentration

Universal Master Mix II 10 multiply by 1X

Primer F 2.0 number of 900nM

Primer R 2.0 samples 900nM

TAqMan® Probe 2.0 250nM

DNA template 4.0 ---

Water ---- ---

Final Volume 20.0

101

Chapter Four

Results and Discussion

The minimal detection of Escherichia coli ATCC®8739™ and Aspergillus

brasiliensis ATCC®16404™ in three different raw materials (corn starch, sucrose, sodium

chloride) and three pharmaceutical products (solid, liquid, cream) contaminated with a

specific known population was detected at minimum level of dilution10¯¹⁵ by Real-time

PCR (Table 4.09). Results of the study for two phases included. Phase I (sensitivity test)

for minimal detection of microorganism’s present in the sample for E. coli indicated that

the detection in culture medium for bacteria was from the dilution 10⁰ to 10¯⁹, with 10¯⁹ as

the minimum amount that was detected under conventional pour-plate method with a

number of colonies on average of 1CFU / plate (Table 4.01). The chosen dilution in the

serial dilution was 10¯⁷ for E. coli, which consistently presented the parameter from 10 to

100 CFU of the required known population of cells to contaminate the raw materials and

the OTC products .

Table 4.01 contains the results in CFU/plate by conventional pour-plate method

for bacteria. From the dilution of 10¯¹⁰ to 10¯¹⁵, there was no colony growth, indicating no

presence of bacteria was detected with the conventional microbiological method. Figure

4.01 shows the results of E. coli in CFU by the pour-plate method. To confirm the presence

of contamination with bacteria in each sample, the microorganisms were isolated and

biochemically identified (Table 4.02).

102

Figure 4.01. Sensitivity test for minimal detection of E.coli ATCC®8739™ using the CFU

conventional method.

103

Table 4.01. Sensitivity test for minimal detection of Escherichia coli ATCC®8739™ by

conventional pour-plate method. Technique used: Serial dilution/counting on plate

(CFU/plate).

Escherichia coli ATCC®8739™

Mean of Triplicates by Runs

Sample Run 1 Run 2 Run 3

Negative Control 0 0 0

10⁰ TNTC TNTC TNTC

10¯¹ TNTC TNTC TNTC

10¯² TNTC TNTC TNTC

10¯³ TNTC TNTC TNTC

10¯⁴ TNTC TNTC TNTC

10¯⁵ TNTC TNTC TNTC

10¯⁶ 293 299 299

10¯⁷ 66 67 67

10¯⁸ 9 8 9

10¯⁹ 2 1 1

10¯¹⁰ 0 0 0

10¯¹¹ 0 0 0

10¯¹² 0 0 0

10¯¹³ 0 0 0

10¯¹⁴ 0 0 0

10¯¹⁵ 0 0 0

104

Table 4.01 presents the results of the averages of triplicates for each run in triplicate

repeated five times. The bold type letter shows the population selected as known

population.

The known population was the dilution selected to artificially contaminated

samples of raw materials and finished products for the second phase of the study (phase

II - TSM). Also, table 4.01 shows in bold letters the dilution selected as the most diluted

to demonstrate the sensitivity effect of both methods.

Table 4.02. Confirmatory Test for E. coli ATCC®8739™ identification detected in USP

conventional pour-plate and Real-time PCR methods.

Sample Gram Stain ID Real-time PCR

10⁰ Gram negative rod E. coli +

10ˉ¹ Gram negative rod E. coli +

10ˉ² Gram negative rod E.coli +

10ˉ³ Gram negative rod E.coli +

10ˉ⁴ Gram negative rod E.coli +

10ˉ⁵ Gram negative rod E.coli +

10ˉ⁶ Gram negative rod E.coli +

10ˉ⁷ Gram negative rod E.coli +

10ˉ⁸ Gram negative rod E.coli +

10ˉ⁹ Gram negative rod E.coli +

105

The Table 4.02 and Table 4.03 shows the results obtained in the identification of

bacteria and fungi from samples used for sensitivity test for both methodologies

(Conventional pour-plate and Real-time PCR).

Table 4.02., continued.

10ˉ¹⁰ No growth 0 CFU +

10ˉ¹¹ No growth 0 CFU +

10ˉ¹² No growth 0 CFU +

10ˉ¹³ No growth 0 CFU +

10ˉ¹⁴ No growth 0 CFU +

10ˉ¹⁵ No growth 0 CFU +

106

Table 4.03. Confirmatory Test for A. brasiliensis ATCC®16404™ identification detected

in USP conventional pour-plate and Real-time PCR methods.

Sample Meetsª

Macro/Micromorphology Observations

ID Real-time PCR

10⁰ + A. Brasiliensis +

10ˉ¹ + A. Brasiliensis +

10ˉ² + A. Brasiliensis +

10ˉ³ + A. brasiliensis +

10ˉ⁴ + A. brasiliensis +

10ˉ⁵ + A. brasiliensis +

10ˉ⁶ No growth 0 CFU +

10ˉ⁷ No growth 0 CFU +

10ˉ⁸ No growth 0 CFU +

10ˉ⁹ No growth 0 CFU +

10ˉ¹⁰ No growth 0 CFU +

10ˉ¹¹ No growth 0 CFU +

10ˉ¹² No growth 0 CFU +

10ˉ¹³ No growth 0 CFU +

10ˉ¹⁴ No growth 0 CFU +

10ˉ¹⁵ No growth 0 CFU +

ª Colony first white then dark brown to black. The reverse of the colony light brown.

Exudates absent. Conidial heads globose at first and later radiate,walls thick, smooth, pale

brown; vesicles 30–45 mm wide, nearly globose; biseriate; metulae covering the entire

surface of the vesicle with phialides flask-shaped with conidia subglobose, echinulate. No

sclerotia observed in the culture .

107

The fungal results indicated that A. brasiliensis was detected by conventional

microbiological method at the 10¯⁵ dilution with a minimum detection average of 3 x 10⁵

cells (Table 4.04). The table presents the results of the averages of triplicates for each

run in triplicates repeated five times. Figure 4.02 shows the results of A. brasiliensis in

CFU by the conventional pour-plate method. Based on the reproducibility of the results,

they showed that the dilution representing the range of 10 to 100 CFU for the fungus was

10¯³ dilutions (Table 4.04). The 10¯⁷ dilutions in E. coli and 10¯³ in A. brasiliensis were

chosen to contaminate the second phase of this study to test for specified microorganisms

(TSM).

108

Figure 4.02. A. brasiliensis results in CFU for sensitivity test by conventional pour-plate

method.

109

Table 4.04. Sensitivity test for minimal detection of Aspergillus brasiliensis

ATCC®16404™ by conventional pour-plate method. Technique used: Serial

dilution/Counting on plate (CFU).

Aspergillus brasiliensis ATCC®16404™

Mean of Triplicates by Runs

Dilution Run 1 Run 2 Run 3

Negative Control 0 0 0

10⁰ TNTC TNTC TNTC

10¯¹ TNTC TNTC TNTC

10¯² 210 44 218

10¯³ 40 43 43

10¯⁴ 7 6 11

10¯⁵ 1 7 1

10¯⁶ 0 0 0

10¯⁷ 0 0 0

10¯⁸ 0 0 0

10¯⁹ 0 0 0

10¯¹⁰ 0 0 0

10¯¹¹ 0 0 0

10¯¹² 0 0 0

10¯¹³ 0 0 0

10¯¹⁴ 0 0 0

10¯¹⁵ 0 0 0

110

The tables’ 4.01 and 4.04 indicated in bold the chosen population of bacteria and

fungi to contaminate the raw materials and finished products to the TSM test. Also, the

dilution of 10¯¹⁵ was chosen even when the conventional method did not detect the

presence of organisms by the sensitivity test.

Thus Real-time PCR demonstrated the sensitivity of the methodology to detect the

minimal trace of E. coli and A. brasiliensis in the presence of raw material and OTC

products for TSM by conventional and molecular methods. To directly detect the presence

of specified bacteria and fungi contamination in the raw material and finished product,

DNA was extracted using TaqMan® Advanced Fast Master Mix analyzed StepOne™

Real-time PCR with two different primers to amplify a conserved DNA sequence for E. coli

16S and A. brasiliensis ITS region. The number of copies of the DNA sequence for E. coli

and A. brasiliensis with specific sequence-primers designed for each organism was

detected. Tables 4.05 and 4.06 presents the threshold cycle (Ct) at which the fluorescent

signal of the reaction crosses the threshold. This Ct was used to calculate the initial DNA

copy number, because the Ct value was inversely related to the starting amount of target.

The results obtained from Ct by molecular Real-time PCR methodology indicated

amplification of the DNA target for samples from 10¯¹ to 10¯¹⁵ for E. coli and A. brasiliensis

in comparison with the control (Tables 4.05 and 4.06).

111

Table 4.05. Real-time PCR – Treshold Cycle (Ct) values of E.coli at minimum detection

(Sensitivity test).

Sample Triplicates Ct Ct mean Sample Triplicates Ct Ct mean

10¯¹ 1 15.36 25.76 10¯⁹ 1 32.50 34.87 2 34.02 25.76 2 37.09 34.87 3 27.92 25.76 3 35.03 34.87

10¯² 1 28.461 28.19 10¯¹⁰ 1 33.35 32.21 2 28.20 28.19 2 30.48 32.21 3 27.90 28.19 3 32.80 32.21

10¯³ 1 30.98 31.21 10¯¹¹ 1 33.89 33.74 2 31.09 31.21 2 33.69 33.74 3 31.57 31.21 3 33.65 33.74

10¯⁴ 1 31.88 32.65 10¯¹² 1 30.54 30.21 2 33.65 32.65 2 29.91 30.21 3 32.42 32.65 3 30.17 30.21

10¯⁵ 1 32.59 33.19 10¯¹³ 1 30.24 31.65 2 33.79 33.19 2 32.70 31.65 3

33.19 33.19

3 32.01 31.65

10¯⁶ 1 29.71 29.24 10¯¹⁴ 1 32.78 30.90 2 29.30 29.24 2 34.50 30.90 3 28.71 29.24 3 25.43 30.90

10¯⁷ 1 33.71 32.93 10¯¹⁵ 1 28.77 33.39 2 31.42 32.93 2 38.00 33.39 3 33.66 32.93 3 33.39 33.39

10¯⁸ 1 33.30 33.27 Control 1 10.90 15.89 2 33.25 33.27 Control 2 15.27 15.89 3 33.26 33.27 Control 3 21.50 15.89

Table 4.05 presents the Ct values to determine the ability of the method to detect

the presence of the bacteria even at dilutions 10¯¹⁵. The results show that the control of E.

coli amplified to a Ct of 15.89. The positive control E. coli DNA was from Life Technologies

product code 362251. All samples detected the presence of the objectionable bacteria

with DNA amplification above the Ct value of 25.76. The amplifications signals were

considered positive for E. coli when it was greater than the signal emitted by the control.

The result was negative when it was less than the signal of Ct of 15.89. These results

112

were compared with the number of CFU obtained by the conventional microbiological

pour-plate method. A comparison of the amplification curves for E.coli in comparison with

conventional method revealed significant differences, indicating that the amplification

precision for detection of Real-time PCR was more sensitive and accurate than

conventional method (Fig 4.03). No E. coli contamination in terms of CFU for sensitivity

test was observed in SCDA culture plate for conventional pour-plate method from dilution

10¯¹⁰ to 10¯¹⁵. The bacteria detection obtained by pour- plate method was lower in

comparison with the Real-time PCR, suggesting that the Real-time PCR was more

sensitive than the convetional pour-plate method. Results from bacteria are shown in

Table 4.04. Only 30 minutes was needed to perform the molecular technique Real-time

PCR detection by the StepOne™.

This suggested that the Real-time PCR assays with TaqMan® MGM probe

provided a sensitive and practical system for detection the objectable microorganisms.

These probes were more specific than primers at higher temperature. The primers were

designed according to standard PCR guidelines. These was specifics for the target

sequences for E. coli and A. brasiliensis (Table 3.04).The entire samples reflects that

contains the targets organisms during the cycle (Fig 4.03). The cycle indicated that enough

sample was accumulated for a cleaved probe to allow amplification signals to emerge from

the baseline. At this point, the visible amplification signal inversely related to the initial

target quantity.

113

Escherichia coli ATCC® 8739™

Aspergillus brasiliensis ATCC®16604™

Figure 4.03. Amplification plot for a 10 fold dilution for E.coli ATCC® 8739™ (above) and

A. brasiliensis ATCC®16404™ (below).

114

As shown in Figure 4.03, the Real-time PCR with the customs TaqMan® assay

designed primer demonstrated positive results with the specific target bacteria and fungal

species and did not detect other non-target microorganisms in the samples. The amount

of viable bacteria and fungi cells count in the serial dilutions for sensitivity test and TSM

samples were determined by Cellometer® Auto M10 (Nexcelom Bioscience) and

compared with the corresponding conventional CFU plate counts method (Table 4.07 and

Table 4.08).

In A. brasiliensis, the fungi colony growth was observed until 10¯⁵, no CFU were

reflected from 10¯⁶ to 10¯¹⁵. In all repetition and replicates, no contamination was observed

from dilution 10¯¹⁰ to 10¯¹⁵ for samples containing E. coli and also no contamination was

observed for A. brasiliensis from dilution 10¯⁶ to 10¯¹⁵ (Fig. 4.02). Both strains used to

contaminate the samples and recovered by the plate count were isolated and identified to

confirm that the organisms recovered were the strain used for the study.

115

Table 4.06. Real-time PCR Ct values of A. brasiliensis ATCC 16404 at minimum

detection sensitivity test.

Sample Triplicate Ct Ct mean Sample Triplicate Ct Ct mean

10¯¹ 1 23.61 28.17

10¯⁹ 1 32.37 32.44

2 28.17 28.17 2 32.49 32.44 3 32.73 28.17 3 32.45 32.44

10¯² 1 26.37 26.16 10¯¹⁰ 1 27.51 27.45 2 26.00 26.16 2 27.66 27.45 3 26.12 26.16 3 27.18 27.45

10¯³ 1 29.49 29.46 10¯¹¹ 1 25.42 25.39 2 29.47 29.46 2 25.37 25.39 3 29.41 29.46 3 25.38 25.39

10¯⁴ 1 32.97 32.74 10¯¹² 1 29.40 29.57 2 32.45 32.74 2 29.89 29.57 3 32.80 32.74 3 29.43 29.57

10¯⁵ 1 31.48 31.40 10¯¹³ 1 32.31 32.79 2 31.43 31.40 2 32.90 32.79 3 31.28 31.40 3 33.15 32.79

10¯⁶ 1 33.19 33.62 10¯¹⁴ 1 32.01 31.26 2 33.58 33.62 2 30.96 31.26 3 34.09 33.62 3 30.81 31.26

10¯⁷ 1 36.94 36.06 10¯¹⁵ 1 17.50 25.69 2 36.24 36.06 2 25.59 25.69

3 34.99 36.06 3 33.98 25.69

10¯⁸ 1 33.29 33.67 Control 1 21.77 21.71 2 34.69 33.67 Control 2 21.62 21.71 3 33.04 33.67 Control 3 21.74 21.71

The E. coli count obtained by CFU counting in contrast with the Real-time PCR Ct

value (Table 4.05) correlated to dilutions corresponding to bacterial counts ranging from

10⁰ to 10¯⁹ for E. coli in terms of detection the presence of organisms in the sample. The

fungi correlated in detection from 10⁰ to 10¯⁵ (Table 4.06). The Real-time PCR method

had amplification of E. coli DNA from Taq DNA polymerase in all dilutions from 10¯¹ to

10¯¹⁵ by the sensitivity test and detected the bacteria in the raw materials and OTC product

samples analyzed at 10¯⁷ and 10¯¹⁵ for TSM. Also, in A. brasiliensis was obtained

116

amplification in all samples from sample 10¯¹ to 10¯¹⁵ for sensitivity test and 10¯³ and 10¯¹⁵

for detection of the presence of the fungi in TSM for finished product and raw materials.

Table 4.07. Live and dead cells of Escherichia coli ATCC® 8739™ with Cellometer®Auto M10.

Dilution Live cell Dead cell Viability %

10⁰ 265 205 56.4

10¯¹ 225 201 52.8

10¯² 194 158 55.0

10¯³ 124 89 58.0

10¯⁴ 103 86 54.7

10¯⁵ 102 80 56.0

10¯⁶ 75 114 39.7

10¯⁷ 74 110 37.0

10¯⁸ 57 37 60.0

10¯⁹ 28 36 43.8

10¯¹⁰ 26 37 41.3

10¯¹¹ 21 32 40.4

10¯¹² 11 22 33.3

10¯¹³ 0 16 0

10¯¹⁴ 0 13 0

10¯¹⁵ 0 0 0

117

Table 4.08. Live and dead cells of Aspergillus brasiliensis ATCC® 16404™ with

Cellometer®Auto M10.

Dilution Live cell Dead cell Viability %

10⁰ 323 263 55.1

10¯¹ 284 214 57.0

10¯² 241 182 58.8

10¯³ 194 157 55.3

10¯⁴ 156 139 53.6

10¯⁵ 132 79 62.6

10¯⁶ 124 89 58.2

10¯⁷ 104 86 54.7

10¯⁸ 103 81 56.0

10¯⁹ 57 38 60.0

10¯¹⁰ 28 36 43.8

10¯¹¹ 13 24 35.1

10¯¹² 6 18 25.0

10¯¹³ 2 5 28.0

10¯¹⁴ 0 3 0

10¯¹⁵ 0 4 0

118

A comparison of Total Bacteria plate count with Ct detection in all analyzed

samples of raw material and OTC products revealed significant differences between the

microbiology pour-plate method versus Real-time PCR because the Real-time PCR

product amplified in all samples. The conventional method did not have the ability to detect

small traces of the 10¯¹⁵ sample dilutions while the Real-time PCR method was able to

detect at this dilution. Cell viability was 52% for the E. coli strain and 54% for A.

brasiliensis. The E. coli counts of more than 100 cells by the cytometry and plate count

correlated to dilutions from 10¯1 to 10¯⁶. Aspergillus correlated to dilution from 10¯¹ to 10¯²

(and from 10¯³ onwards there was no correlation). Bacteria and fungal cytometry with

dilutions from had cell viability at 10¯¹³ and from 10¯¹⁴ to 10¯¹⁵ reflected no viable cells.

Table 4.08 compares the data obtained for the detection performed by the plate count,

cytometry, and Real-time PCR methods.

119

Table 4.09. Presence in CFU, Cells and Ct value.

Escherichia coli ATCC® 8739™ Aspergillus brasiliensis ATCC® 16404™

Dilution Live cell Plate

CFU

Ct Live cell Plate CFU Ct

10¯¹ 284 TNTC 25.76 323 TNTC 28.17

10¯² 241 TNTC 28.19 284 210 26.16

10¯³ 194 TNTC 31.21 241 40 29.46

10¯⁴ 156 TNTC 32.65 194 7 32.74

10¯⁵ 132 TNTC 33.19 156 1 31.40

10¯⁶ 124 297 29.24 132 0 33.62

10¯⁷ 104 67 32.93 124 0 36.06

10¯⁸ 103 9 33.27 104 0 33.67

10¯⁹ 57 1 34.87 103 0 32.44

10¯¹⁰ 28 0 32.21 57 0 27.45

10¯¹¹ 13 0 33.74 28 0 25.39

10¯¹² 6 0 30.21 13 0 25.69

10¯¹³ 2 0 31.65 6 0 31.65

10¯¹⁴ 0 0 30.90 2 0 30.90

10¯¹⁵ 0 0 33.39 0 0 33.39

120

As shown in Table 4.09 the expected detection of E. coli and A. brasiliensis was

successfully amplified and detected with specific primers in each phosphate buffer

dilutions (sensitivity test).

The results obtained in the second phase of the study for test for specified

microorganisms (TSM) are shown in Table 4.10. Samples of finished products and raw

material contaminated with E. coli strain cells at concentrations of 10¯⁷ were recovered by

the conventional method like the molecular Real-time PCR method. However, the

conventional method failed to detect E. coli strain in samples of finished products and raw

materials to a minimum concentration of 10¯¹⁵ (Table 4.10), contrary to the Real-time PCR

that did detect the presence of bacteria in all samples of raw materials and finished

products analyzed at concentrations from 10¯⁷ to 10¯¹⁵. The TSM results obtained with the

strain of A. brasiliensis were similar to those with E.coli. The conventional methods did not

have the sensitivity to detect the contamination with A. brasiliensis in the samples of raw

material and finished product at a minimum concentration of 10¯¹⁵.

These results proved the sensitivity, acuracy and efficiency of primers and Real-

time PCR assay used in this study and, demonstrated that the raw material and

pharmaceutical finished product sample ingredients analyzed had no adverse effects on

Real-time PCR detection.

121

Table 4.10. Test for Specified Microorganisms detection by USP conventional pour-plate

and Real-time PCR 141.

TaqDNA fragment detection was amplified using specific primers for the target

bacterial and fungal DNA. The sensitivity of the Real-time PCR was constant, with 40

cycles performed in each experiments, indicatings the precision and effectiveness to

detect the strains of E. coli ATCC® 8739™ and A. brasiliensis ATCC® 16404™ in

pharmaceutical products and raw materials analyzed.

Escherichia coli

ATCC® 8739™

Aspergillus brasiliensis

ATCC® 16404™

Sample Plate count 10¯⁷ CFU

Ct 10¯⁷

Plate count 10¯¹⁵ CFU

Ct 10¯¹⁵

Plate count 10¯³ CFU

Ct 10¯³

Plate count 10¯¹⁵ CFU

Ct 10¯¹⁵

Finished product

Tablets 130 37.38 0 35.11 225 37.46 0 37.48

Liquid 10 37.46 0 37.48 24 37.70 0 36.89

Cream 5 37.70 0 37.89 26 38.28 0 37.97

Raw material

Sucrose 1 38.28 0 37.97 1 37.62 0 37.37

Corn

starch

1 37.62 0 37.37 1 35.20 0 34.71

Sodium

chloride

1 35.20 0 34.71 1 35.11 0 35.02

122

4.1. Data Analysis

4.1.1. Inferential Data Analysis

For nonparametric tests for two independent samples, the Mann-Whitney U test,

was used to determine statistical differences for the detection of Escherichia coli ATCC®

8739™ and Aspergillus brasiliensis ATCC® 16404™ for each of the two different methods:

conventional, and molecular Real-time PCR methods. There were no significantly

differences for the detection of the organisms among the two methods (Table 4.11). The

Mann-Whitney U test were not significant for the conventional method (74.000, n = 32, p

= .089); this value of p, when compared to a 0.01 significance indicates that the null

hypothesis (p > 0.05) is not rejected. Therefore, there was no statistically significant

difference between Escherichia coli ATCC® 8739™ and Aspergillus brasiliensis ATCC®

16404™ when using the conventional method.

Table 4.11. Mann-Whitney Test for detection of Escherichia coli ATCC® 8739™ and

Aspergillus brasiliensis ATCC® 16404™ for each of the two different methods.

_________________________________________________________________

Contrast Statistics

_________________________________________________________________

U de Mann-Whitney 74.000

Z -1.702

Asymptotic significance .089

________________________________________________________________ a. Grouping variable: Escherichia coli ATCC® 8739™ y Aspergillus brasiliensis

ATCC® 16404™.

The Mann-Whitney U test results were not significant for the Real-time PCR CT,

Mann-Whitney U (88.000 n = 30) =, p = .310. This value of p, when compared to a 0.01

significance indicates that the null hypothesis (p> 0.05) was not rejected. Therefore, there

is no statistically significant difference between Escherichia coli ATCC® 8739™ and

123

Aspergillus brasiliensis ATCC® 16404™ when using the Real-time PCR Ct method (see

Table 4.12).

Tabla 4.12. Mann-Whitney Test for Real-time PCR.

Contrast Statistics

U de Mann-Whitney

Z

Asymptotic significance

88.000

-1.016

.310

a. Grouping variable: Escherichia coli ATCC® 8739™

and Aspergillus brasiliensis ATCC®16404™

The Mann-Whitney U test results were not significant for the conventional and

molecular method (15.000, n = 12, p = 0.629). This value of p, when compared to a 0.01

significance indicates that the null hypothesis (p> 0.05) was not rejected. Therefore, there

is no statistically significant difference between Escherichia coli ATCC® 8739™ and

ATCC® 16404™ Aspergillus brasiliensis in different types of samples by conventional and

molecular method (see Table 4.13).

Table 4.13. Mann-Whitney Testª. _________________________________________________________________

Contrast Statistics

_________________________________________________________________

U de Mann-Whitney 15.000

Z -0.484

Asymptotic significance .629

_________________________________________________________________ a. Grouping variable: Escherichia coli ATCC® 8739™ y Aspergillus brasiliensis

ATCC® 16404™.

124

4.1.2. Inferential Analysis Methods

For nonparametric tests for two independent samples, the Kolmorgorov-Smirnov

test was used to determine statistical differences for the detection of Escherichia coli

ATCC® 8739™ and Aspergillus brasiliensis ATCC® 16404™ for the conventional and Real-

time PCR methods. This test can find differences when analyzing two methods with two

different measures (units).The Kolmogorov can analyze CFU (conventional method) with

Ct value (Real-time PCR).

The test results were significant, Kolmogorov Smirnov Z (N = 30) = 2.324, p = .000.

This value of p, when compared to a 0.01 significance indicates that the null hypothesis

(p <0.01) is rejected (Table 4.14 and Table 4.15). Therefore, there is a statistically

significant differences in the detection of Escherichia coli ATCC® 8739™ and ATCC®

16404™ Aspergillus brasiliensis in conventional microbiological pour plate and the Real-

time PCR methods.

Table 4.14. Frequencies.

Method N

Values

Conventional 30

Real-time PCR 30

Total 60

125

Table 4.15. Test statisticª.

Values

Most

Differences

Extreme

Absolute .600

Positive .600

Negative -.400

Z de Kolmogorov-Smirnov 2.324

Asymptotic significance (bilateral) .000

a. Grouping variable: Method.

126

Chapter 5

Conclusions and Recommendations

Real-time PCR testing using TaqMan® with specific primers and simple sample

protocol preparation procedure demonstrated increased sensitivity, effectiveness, and

replicability as compared to conventional pour-plate methods for detecting low levels of

bacteria and fungal contamination less than 1 CFU, in raw material and pharmaceutical

products. The Real-time PCR method detected the minimal trace of the presence of E.

coli ATCC® 8739™ at a dilution of 10ˉ¹ to 10ˉ¹⁵ for sensitivity test (minimal detection) and

of 10¯³ and 10¯¹⁵ in all samples analyzed for Test of specified Microorganisms (TSM) for

raw materials and OTC products. Also, in A. brasiliensis ATCC® 16404™ the molecular

method amplified in all samples from sample 10ˉ¹ to 10ˉ¹⁵ for sensitivity test and 10ˉ³ and

10ˉ¹⁵ for detection of the presence of the fungi in TSM for finished product (tablet, liquid

and cream) and raw materials (sucrose, corn starch and sodium chloride).

The Real-time PCR with TaqMan® was a sensitive, accurate and reproducibility

method targeting the E. coli ATCC® 8739™ and A. brasiliensis ATCC® 16404™ in raw

material and finished product. The Real-time PCR method had increased specificity at

trace levels with a concentration of 10ˉ¹⁵ of the organisms cells in comparison with the

conventional pour-plate method. It was found there is a statistically significant differences

in the terms of detection the presence of Escherichia coli ATCC® 8739™ and ATCC®

16404™ Aspergillus brasiliensis in conventional microbiological pour-plate and the Real-

time PCR methods. The RT-PCR is recommended for detecting contamination in raw

material because a minimum sensitivity of 10ˉ¹⁵ was obtained in all samples which might

not be possible for the conventional pour-plate method. This molecular methodology in

this study presented the benefits in terms of increased sensitivity, reduced the risk of

contamination and variability between runs.

127

The Real-time PCR method with TaqMan® proved be faster, and effective

methodology for the sensitive detection of objectionable microorganisms for the USP <62>

Microbiological Examination of Nonsterile Products Tests for Specified Microorganisms

and for microbial enumeration established in USP Chapter <61> Microbiological

Examination of Nonsterile Products Microbial Enumeration Tests.

Pharmaceutical industries should strive to incorporate Real-time PCR

methodologies into written standard operating procedures (SOPs), protocols, and

validation techniques. Thus, this investigation will contribute with the industry by detailing

a step-by-step protocol of microbiological molecular analyses applied directly to samples

of raw material and pharmaceutical OTC products.

This investigation will contribute in the rapid detection of the presence of

objectionable microorganisms in terms of sensitivity, specificity, and effectivity with a

minimal trace, which was not possible under conventional microbiology pour-plate

methodologies. This contribution will directly impact the traditional methodology

established in the pharmacopeia chapter <62> of the Compendium of the USP (2015).This

helps to reduce the risk of exposure of humans and animals to contaminated drugs that

do not meet the FDA and USP quality standards. The early and rapid detection of a

precise and effective methodology as the Real-time PCR helps in protecting public health

by avoiding outbreak of nosocomial infections.

128

Literature Cited

Abarca ML. 2000. Taxonomía e identificación de especies implicadas en la aspergilosis

nosocomial. Revista Iberoamericana Micología 17: 79-84.

Abdin MZ, Ahmad MM, Javed S. 2010. Advances in molecular detection of Aspergillus:

an update. Archives Microbiology 192(6): 409-425.

Abee T, Wounters JA. 1999. Microbial stress response in minimal processing.

International Journal Food Microbiology 50(1-2): 65-91.

Avallone HL. 1986. Laboratory controls – An FDA investigators viewpoint. PDA Journal

Pharmaceutical Science Technology 40(5): 178-182.

Baird RM. 1998. Pharmaceutical Microbiology 6th Ed. W.B. Hugo & A.D. Rusell (Eds)

Blackwell Science, Oxford, UK, pp 374-384.

Balajee SA, Sigler L, Brandt ME. 2007. DNA and the classical way: identification of

medically important molds in the 21st Century. Medical Mycology 45(6): 475-490.

Bennett JW. 2009. Aspergillus: a primer for the novice. Medical Mycology 47(1): 5-12.

Bomblies L, Weiss C, Bekman G. 2007. Examination of microbiological quality

of pharmaceutical raw material. Pharmaceutical Science Notes 1:1-7.

Bosch F, Rosich L. 2008. "The contributions of Paul Ehrlich to pharmacology: a

tribute on the occasion of the centenary of his Nobel Prize". Pharmacology 82 (3):

171–179.

Bosshard PP, Zbinden R, Abels S, Böddinghaus B, Altwegg M,

Böttger EC. 2006. 16S rRNA gen sequencing versus the API 20 NE

system and the VITEK 2 ID-GNB Card for identification of nonfermenting

Gram negative bacteria in the clinical laboratory. Journal Clinical Microbiology

44(4): 1359-1366

129

Brannan DK, Dille JC. 1990. Type of closure prevents microbial contamination of

cosmetic during consumer use. Applied Environmental Microbiology 56(5): 1476–

1479.

Burgos JS, Ramírez C, Tenorio R, Sastre I, Bullido MJ. 2002. Influence of reagents

formulation on real-time PCR parameters. Molecular and Cellular Probe 16: 257–

260.

Bustin SA, Benes V, Garson JA, Hellemans J, Kubista M, Mueller R, Nolan T, Pfaffl MW,

Shipley GL, Vandersompele J, Wittwer CT. 2009. The MIQE guidelines: minimum

information for publication of quantitative real-time PCR experiments. Clinical

Chemistry 55(4): 611-622.

Campana R, Scesa C, Patrone V, Vittoria E, Baffore W. 2006. Microbiological study of

cosmetic products during their use by cosmetics: health risk and efficacy of

preservative systems. Letters Applied Microbiology 42:301-306.

Casey W, Muth H, Kirby J, Allen P. 1998. Use of nonselective preenrichment media for

the recovery of enteric bacteria from pharmaceutical products. Pharmaceutical

Technology 22: 114-117.

[CDC] Centers of Dicease Control and Prevention. 2013. [Accessed 2014 December 09]

http://www.cdc.gov/medicationsafety/recalls/necc/#advice.

[CFR] Code of Federal Regulations. 2013. CFR Title 21. Vol 8: Part 210-211 current

Good Manufacturing Practice for Finished Pharmaceuticals. Available from:

http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/CFRSearch.cfm?CFRPar

t=211.

Changsoo L, Jaai K, Seung GS, Seokhwin H. 2005. Absolute and relative QPCR

quantification of plasmid copy number in Escherichia coli. Journal of

Biotechnology 123: 273-280.

130

Chaubron F, Martin-Minvielle A, Groulon S. 2006. One step real-time

PCR kits for the universal detection of organisms in industrial products. World

Intelectual Property Organization WO2004/044247. Available from:

"http://www.sumobrain.com/patents/WO2004044247.html",

Clontz L. 1998. Microbial limit and bioburden tests. Interpharm Press, Inc. IL,

USA. 230p.

Clontz L. 2009. Microbial limit and bioburden tests: validation approaches and global

requirements. CRC Press: Taylor & Francis Group. 311p.

Coker M. 2005. An assessment of microbial contamination during drug manufacturing in

Ibadan, Nigeria. European Journal of Scientific Research 7: 19-23.

Cundell AM. 2006. Microbial identification strategies in the pharmaceutical

industry. PDA Journal Pharmaceutical Science Technology 60: 111-123.

Cundell AM. 2006. Opportunities for rapid microbial methods American Pharmaceutical

Review 9(7): 50-56.

Cundell AM, Chatellier S, Schumann P, Lilischkis R. 2010. Equivalence of quality control

strains of microorganisms used in the compendial microbiological tests: are

national culture collection strains identical? PDA Journal Pharmaceutical Science

Technology 64(2): 137-155.

Davey HM. 2011. Life, death, and in-between: Meanings and methods in microbiology.

Applied and Environmental Microbiology 77(16): 5571-5576.

De Boer E, Beumer RR. 1999. Methodology for detection and typing of foodborne

microorganisms. International Journal Food Microbiology 50(1-2): 119-130.

Denoya, CD. 2009. Nucleic Acid Amplification -Based Rapid Microbiological Methods:

Are these Technologies Ready for Deployment in the Pharmaceutical Industry?

American Pharmaceutical Review. 12(9): 12-21.

131

Denoya C, Colgan S, du Moulin GC. 2006. Alternative microbiological methods in the

pharmaceutical industry: the need for a new microbiology curriculum. American

Pharmaceutical Review 9:10-18.

Dorak MT. 2006. Real-time PCR. Taylor & Francis Group. 333p.

Duguid J, Balkovic E, du Moulin GC. 2011. Rapid Microbiological methods: Where are

they now? American Pharmaceutical Review 14(7): 18-25.

Espy MJ, Uhl JR, Sloan LM, Buckwalter SP, Jones MF, Vetter EA, Yao JDC,

Wengenack NL, Rosenblatt JE, Cockerill III FR, Smith TF. 2006. Real-time PCR

in clinical microbiology: Applications for routine laboratory testing. Clinical

Microbiology Reviews 19(1): 165-256.

Fabe J, Moritz N, Henninger N, Zepp F, Knuf M. 2009. Rapid detection of common

pathogenic Aspergillus species by a novel real time PCR approach. Mycoses

52(3): 228-233.

Farrington JK, Martz EL, Wells SJ, Ennis CC, Holder J, Levchuk JW, Avis KE, Hoffman

PS, Hitchins AD, Madden JM. 1994. Ability of laboratory methods to predict in-use

efficacy of antimicrobial preservatives in an experimental cosmetic. Applied

Environmental Microbiology 60(12): 4553-4558.

[FDA] Food and Drug Administration (US). 2014. Products recall. [Accessed 2014

December 05. In: http://www.fda.gov/Drugs/DrugSafety/DrugRecalls/default.htm

[FDA] Food and Drug Administration (US). 2014. CDC Laboratory samples results.

Accessed 2014 December 05.

In:http://www.fda.gov/Drugs/DrugSafety/FungalMeningitis/default.htm

FDAGOV Approval. 2008. Clinical Trials.

Available from: http://www.fda.gov/cder/guidance/index.htm

FDA. Food and Drug Administration. 2014. Guide to inspection of microbiological

pharmaceutical quality control laboratories. Rockville, MD: Food and Drug

132

Administration. Available from:

http://www.fda.gov/ICECI/Inspections/InspectionGuides/ucm074914.htm.

FDA Press Release. Nov-Dec 2005 Issue. Available from: http://www.fda.gov

Ferre F. 1992. Quantitative or semi-quantitative PCR: reality versus myth. PCR Methods

and Applications 2: 1–9.

Fung DYC. 2002. Rapid methods and automation in microbiology. Comprehensive

Reviews Food Science and Food Safety 1: 1-20.

Gad GFM, Aly RAI, Ashour MSE. 2011. Microbial evaluation of some non-sterile

pharmaceutical preparations commonly used in the Egyptian market. Tropical

Journal of Pharmaceutical Research 10(4): 437-445.

Gams W, Christensen M, Onions AHS, Pitt JI, Samson RA. 1985. Infrageneric taxa of

Aspergillus. In: Samson RA, Pitt JI, eds. Advances in Aspergillus systematics. New

York: Plenum Press. p 55-64.

Ghandhi M. 2006. Efficiency with Rapid Microbiology Methods. American

Pharmaceutical Review 9 (5): 16-19.

Geiser DM, Klich MA, Frisvad JC, Peterson SW, Varga J, Samson RA. 2007. The

current status of species recognition and identification in Aspergillus. Studies

Mycology 59: 1-10.

Guilfoyle DE, Friedman RL, Hughes PF, Hussong D, Rosenberg AS, Brorson D. 2013.

Microbial risk in pharmaceutical manufacturing and ICHQ9. PDA Journal

Pharmaceutical science Technology 67(2): 79-80.

Houseknecht J, Stamenova E, Suh SO, Beck B, McKee M, Zhou J. 2008.

Reclassification of ATCC® 16404™ and ATCC® 9642™ as Aspergillus

brasiliensis. Pharmaceutical Microbiology Forum Newsletter 14: 2-8.

Hussong D, Mello R. 2006. Alternative microbiology methods and pharmaceutical

quality control. American Pharmaceutical Review 9(1): 62-68.

133

ICH. 2007. International Conference on Harmonization

Available from:

http://www.ec.europa.eu/Enterprise/pharmaceuticals/eudralex/index.htm.

Li RY, Li DM, Yu J, Liu W, Ji ZH, Wang DL. 2004. Application of molecular biology

techniques in the identification of pathogenic fungi and the diagnosis of fungal

infection. Journal of Peking University, Health Science 36(5): 536-539.

Jaffe RI, Lane JD, Bates CW. 2001. Real-time identification of

Pseudomonas aeruginosa direct from clinical samples using a rapid extraction

method and polymerase chain reaction. Journal Clinical Laboratory Analysis

15(3):131-137.

Jiménez L. 2001. Molecular diagnosis of microbial contamination in cosmetic

and pharmaceutical products: a review. Journal AOAC International 84: 671-

675.

Jiménez L. 2007. Microbial diversity in pharmaceutical product recalls and environments.

PDA Journal of Pharmaceutical Science and Technology 61(5): 383-399.

Jiménez L. 2011. Molecular applications to pharmaceutical processes and cleanroom

environments. PDA Journal of Pharamceutical Science Technology 65: 242-253.

Jiménez L, Smalls S, Ignar R. 2000. Use of PCR analysis for detecting

low levels of bacteria and mold contamination in pharmaceutical

samples. Journal Microbiology Method 41: 259-265.

Jurjević Ž, Peterson SW, Stea G, Solfrizzo P, Vargas J, Hubka V, Perrone G. 2012. Two

novel species of Aspergillus section Nigri from indoor air. IMA Fungus – The Global

Micological Journal 3(2): 159-173.

Kai FH. 2004. A generalized plate method for estimating total aerobic microbial count.

PDA Journal of Pharmaceutical Science and Technology 58(5): 261-265.

Karami A, Ahmadiz Z, Safiri Z, Khalilpour A, Morrovatis S. 2006. Development of and

134

rapid and simple multiplex polymerase chain reaction technique for detection of

Salmonella typhi. Saudi Medical Journal 27(8): 1134-1138.

Karanam VR, Reddy HP, Subba RBV, Rao JC, Kavikishore PB, Vijayalakshmi M. 2008.

Detection of indicator pathogens from pharmaceutical finished products and raw

materials using multiplex PCR and comparison with conventional microbiological

methods. Journal Industrial Microbiology Biotechnology 35(9): 1007-1018.

Kearns AM, Graham C, Burdess D, Haetherington J, Freeman R. 2002.

Rapid real-time PCR for determination of Penicillin susceptibility in

pneumococcal meningitis, including cultura-negative cases. Journal Clinical

Microbiology 40 (2): 682-684.

Klein D. 2002. Quantification using real-time PCR technology: applications and

limitations. Trends in Molecular Medicine 8: 257-260.

Klingspor L, Jalal S. 2006. Molecular detection and identification of Candida

and Aspergillus spp. From clinical samples using real-time PCR. Clinical

Microbiology Infection 12(8):745-753.

Kubista M, Andrade JM, Bengtsson M, Forootan A, Jonak, Link K, Sindelka R, Sjöback

R, Strömbom L, Ståhlberg A, Zoric N. 2006. The real-time polymerase chain

reaction. Molecular Aspects Medicine 27(2-3): 95-125.

Kushwaha P. 2010. Microbial contamination: A regulatory perspective. Journal of

Pharmacy Research 3 (1): 124-131.

Lanfear D, McLeod H. 2007. Pharmacogenetics: Using DNA to Optimize Drug

Therapy. American Family Physician 76:1179-1182.

Lee C, Lee S, Shin SG, Hwang S. 2008. Real-time PCR determination of rRNA gene

copy number: absolute and relative quantification assays with Escherichia coli.

Applied Microbiology Biotechnology 78(2): 371-376.

Linter K, Genet V. 1998. A physical method for the preservation of cosmetic products.

135

International Journal Cosmetic Science 20(2): 103-115.

Livak KJ, Schmittgen TD. 2001. Analysis of relative gene expression data using real-

time quantitative PCR and the 2(-Delta Delta C (T) method. Methods 25(4): 402-

408.

Lolas AG, Metcalfe JW. 2011. Evaluation of the microbial growth potential of

pharmaceutical drug products and quality by design. PDA Journal of

Pharmaceutical Science and Technology 65(1): 63-70.

Mackay IM, Arden KE, Nitsche A. 2002. Real-time pcr in virology. Nucleic Acids

Research. 30(6):1292-1305.

Madsen RE. 2001 “New Technologies for Microbiological Testing” In Microbiology in

Pharmaceutical Manufacturing. R. Prince, Ed, p. 147-156. Parenteral Drug Assoc

Baltimore.

Manu-Tawiah W, Brescia BA, Montgomery ER. 2001. Setting threshold limits for the

significance of objectionable microorganisms in oral pharmaceutical products.

PDA Journal of Pharmaceutical Science and Technology 55(33): 171-175.

Marlowe EM, Bankowki MJ. 2011. Conventional and molecular methods for the

detection of Methicillin-Resistant Staphylococcus aureus. Journal of Clinical

Microbiology 49(9): S55-S56.

Martínez-Bermudez A, Rodríguez de Lecea J, Soto-Esteras T, Vázquez-Estevez C,

Chena-Canete C. 1991. Tipos de contaminantes microbianos de materias primas

farmacéuticas. Revista Latinoamerica Microbiología 33:153-157.

Matsuda K, Tsuji H, Asahara T, Kado Y, Nomoto K. 2007.

Sensitive quantitative detection of comensal bacterial by rRNA-targeted

reverse transcription PCR. Applied Environmental Microbiology 73(1): 32-39.

Meijer M, Houbraker JAMP, Dalhuijsen S, Samson RA, de Vries RP. 2011. Growth and

136

hydrolase profiles can be used as characteristicas to distinguish Aspergillus niger

and other black aspergilli. Studies Micology 69(1): 19-30.

McArthur M, Bibb MJ. 2008. Manipulating and understanding antibiotic

production in Streptomyces coelicolor A3(2) with decoy oligonucleotides.

Proceedings National Academy Science United State America 22:105(3):1020-

1025.

Miller HI. 1982. Recombinant DNA as a paradigm of a new technology: Its impact on

regulation by the Food Drug Administration. PDA Journal Pharmaceutical Science

Technology 36(6): 248-250.

Miller MJ. 2005. Rapid microbiological methods and FDA’s initiatives for pharmaceutical

cGMO’s for the 21st Century, PAT, and sterile drug products produced by aseptic

processing. American Pharmaceutical Review 17(2): 65-67.

Miller MJ. 2009. It's Time to get rapid!. PDA Letter. April: 16-21.

Miller MJ. 2012. Case study of a new growth-based rapid microbiological method (RMM)

that detects the presence of specific organisms and provides an estimation of

viable cell count. American Pharmaceutical Review. 15(2):18-25.

Moldenhauer J. 2006. Viability based rapid microbiological methods for sterility

testing and the need for identification of contamination. PDA Pharmaceutical

Science & Technology 60(2): 81-88.

Nigel AH. 2002. Mirobial control of pharmaceutical. Encyclopedia of Pharamceutical

Technology. Third ed. Vol. 1: 1753-1764.

Noonim P, Mahakarachanakul W, Varga J, Frisvad JC, Samson RA. 2008. Two novel

species of Aspergillus section Nigri from Thal coffee beans. International Journal

Systematic Evolutionary Microbiology 58(7): 1727-1734.

Okeke IN, Lamikanra A. 2001. Bacteriological quality of skin-mosturizing creams and

137

lotions distributed in a tropical developing country. Journal Applied Bacteriology

91: 922-928.

Parker MS. 2000. Microbiological contamination and preservation of pharmaceutical

preparations. In: Pharmaceuticals: The science of dosage from design. 2nd ed.

China, Churchill Livingstone. P220.

Pathmanathan SG, Cardona-Castro N, Sanchez-Jiménez MM, Correa-Ochoa MM,

Puthucheary SD, Thong KL. 2003. Simple and rapid detection of Salmonella

strains by direct PCR amplification of the hilA gene. Journal Medical Microbiology

52(9): 773-776.

Perrone G, Varga J, Susca A, Frisvad JC, Stea G, Kocsube S, Toth B, Kozakiewicz Z,

Samson R A. 2008. Aspergillus uvarum sp. nov., an uniseriate black Aspergillus

species isolated from grapes in Europe. International Journal Systematic

Evolutionary Microbiology 58: 1032-1039.

Postgate JR. 1969. Viable counts and viability. Methods microbial 1: 611-628.

[PMM] Pharmaceutical Microbiology Manual. 2014. ORA.007, Version 1.1

In: http://www.fda.gov/downloads/scienceresearch/fieldscience/ucm397228.pdf.

Ragheb SM, Yassin AS, Amin MA. 2012. The application of uniplex, duplex and

multiplex PCR for the absence of specified microorganism testing of

pharmaceutical excipients and drug products. PDA Journal of Pharmaceutical

Science and Technology 66(4): 307-317.

Ragheb SM, Jiménez L. 2014. Polymerase Chain Reaction/Rapid method are gaining a

foothold in developing countries. PDA journal of Pharmaceutical Science and

Technology 68(3): 239-255.

Ramírez M, Castro C, Palomares JC, Torres MJ, Aller AL, Ruiz M, Aznar J, Martín-

Mazuelos E. 2009. Molecular detection and identification of Aspergillus spp. from

clinical samples using real time PCR. Mycoses 52(2): 129-134.

138

Rapper KB, Fennell DI. 1965. The genus Aspergillus. Baltimore: Williams & Wilkins.

Rinyu E, Varga J, Ferenczy L. 1995. Phenotypic and Genotypic Analysis of

Variability in Aspergillus fumigatus. Journal Clinical Microbiology 33(10): 2567-

2675.

Rodríguez-Pérez J. 2014. The FDA andwordlwide current good manufacturing practices

and quality system requirements guidebook for finished pharmaceuticals. ASQ

Quality Press Milwaukee 53203.

Sandle T. 2014. Approaching the selection of rapid microbiological method. Institute of

Validation Technology Network.

In:http://www.ivtnetwork.com/article/approaching-selection-rapid-microbiological-

methods.

Saghee MR, Sandle T, Tidswell EC. 2010. Microbiology and Sterility Assurance in

Pharmaceuticals and Medical Devices. Ed. New Delhi: Business Horizons.

In: Pharma Manufacturing. com.

http://www.pharmamanufacturing.com/articles/2011/004/?show=all

Samadi N, Alvandi M, Fazeli MR, Azizi E, Mehrgan H, Naseri M. 2007. PCR-

based detection of low levels of Staphylococcus aureus contamination in

pharmaceutical preparations. Journal Biology Science 7(2): 359-363.

Samsom RA, Noonim P, Meijer M, Houbraken J, Frisvad JC, Varga J. 2007. Diagnostic

tools to identify black aspergilli. Studies in Mycology 59: 129-145.

Samson RA, Houbraken J, Varga J, Frisvad JC. 2009. Polyphasic taxonomy of the heat

resistant ascomycete genus Byssochlamys and its Paecilomyces anamorphs.

Persoonia 22: 14-27.

Schabereiter GC, Selitsch B, Rotter ML, Hirschl AM, Willinger B. 2007. Development

139

of novel real-time PCR assays for detection and differentiation of eleven medically

important Aspergillus and Candida species in clinical specimens. Journal Clinical

Microbiology 10: 1128-1346.

Shaikh D, Jamshed TA, Shaikh R. 1988. Microbial contamination of pharmaceutical

preparations. Pakistan Journal of Pharmaceutical Science 1: 61-66.

Simões MF, Santos C, Lima N. 2013. Structural diversity of Aspergillus (section nigri)

spores. Microscopy Microanalysis 19(5): 1151-1158.

Ŝkof A, Poljak M, Krbavĉiĉ A. 2007. Real-time polymerase chain reaction for

detection of Staphylococcus aureus and Pseudomonas aeruginosa in

pharmaceutical products for topical use. Journal Rapid Method & Automation in

Microbiology 12(3): 169-183.

Spiegelman D, Whissell G, Greer W. 2005. A survey of the method’s for the

characteristization of microbial consortium and communities. Canadian Journal

Microbiology 51: 355-386.

Sutton SVW. 2012. What is an “Objectionable Organisms” The Shifting Perspective.

American Pharmaceutical Review. Posted Ocxtibr 12, 2012.

In: http://www.americanpharmaceuticalreview.com/Featured-Articles/122201-

What-is-an-Objectionable-Organism-Objectionable-Organisms-The-Shifting-

Perspective/

Sutton S. 2011. Accuracy of plate counts. Journal of Validation Technology 17(3): 42-

46.

Sutton S, Jiménez L. 2012. A review involving microbiological control 2004-2011 with

emphasis on FDA considerations of “Objectionable Organisms”. American

Pharmaceutical Review 15(1): 42-57.

Sutton SVW, Knapp JE, Dabbah R. 2001. Activities of the USP microbiology

subcommittee of revision during the 1995-2000 revision cycle. PDA Journal of

140

Pharmaceutical Science and Technology 55(1): 33-48.

Sutton SVW, Tirumalai R. 2011. Activities of the USP microbiology and sterility

assurance expert committee during the 2005-2010 revision cycle. RMM Review

July/August 12-30.

Tasara T, Schmacher S, Stephen R. 2005. Conventional and real time PCR based

approaches for molecular detection and quantitation of bovine species material in

edible glatine. Journal of Food Protection 68(11): 2420-2426.

Torbeck L, Raccasi D, Guilfoyle DE, Friedman RL, Hussong D. 2011. Burkhloderia

cepacia: This decision is overdue. PDA Journal Pharmaceutical Science

Technology 65(5): 535-543.

Trick WE, Jarvis WR. 1998. Epidemiology of nosocomial fungal infection in the 1990s.

Resvista Iberoamericana Micología 15: 2-6.

Treviňo M, Matínez-Lamas, Romero-Jung P, Varón C, Moldes L, García-Riestra C,

Regueiro BJ. 2009. Comparación entre las pruebas para la detección de beta-

lactamasas espectro extendido de los sistemas Vitek 2 y Phoenix.

Enferemedades Infecciosas y Microbiología Clínica 27(10): 566-570.

U.S. Food and Drug Administration. 2004. Safety, background and definitions. Recalls.

In: http://www.fda.gov/Safety/Recalls/ucm165546.htm

[USP] United States of Pharmacopea Compendium. 2015. Chapetr <61>. Microbial

examination of nonsterile products: microbial enumeration test. Vol. 38-NF33 pp.

57- 62.

[USP] United States of Pharmacopea Compendium. 2015. Chapetr <62>.

Microbiological examination of nonsterile products: Tests for specified

microorganisms. Vol. 38-NF33 pp. 62-67.

[USP] United States of Pharmacopea Compendium. 2015. Chapetr <1111>.

Microbiological examination of nonsterile products: Acceptance criteria for

141

pharmaceutical preparations and substances for pharmaceutical use. Vol. 38-

NF33 pp. 923-925.

[USP] United States of Pharmacopea Compendium. 2015. Chapter <1227>. Validation

of microbial recovery from pharmacopeial articles. Vol. 38-NF33 pp. 1163-1166.

[USP] United States of Pharmacopea Compendium. 2015. Chapter <1041> Biologics.

Vol. 30-NF 25 pp.1-3 pp 618-619.

[USP] United Sates of Pharmacopea Compendium. 2015. Chapter <1045>

Biotecnology. Vol 30-NF 25 pp. 626-640.

Valasek MA, Repa JJ. 2005. The power of real time PCR. Advances Physiology

Education 29(3): 151-159.

Van der Zee H, Huis in’t Veld JH. 1997. Rapid and alternative screening methods

for microbiological analysis. Journal AOAC International 80(4): 934-940.

Varga J, Kocsabé S, Tóth B, Frisvad JC, Perrone G, Susca A, Meijer M, Samson RA.

2007. Aspergillus brasiliensis sp. nov., a biseriate black Aspergillus species with

world-wide distribution. International Journal Systematic Evolutionary

Microbiology 57: 1925-1932.

Vickery MCL, Blackstone GM, DePaola A, Burkhardt B. 2005. Novel quantitative

PCR internal control system adaptable to any standard or real-time PCR assay.

FDA Science Forum. 04p.

Vinueza-Burgos C. 2009. PCR en Tiempo Real: la nueva era de la información genética

celular. REDVET Revista Electrónica de Veterinanria [Internet]. 10(2): 1-13.

Available from Redalyc.org:

http://www.veterinaria.org/revistas/redvet/n020209.html.

Walzer A, Tiemann F, Knaeblein J, Klaus PG, Mueller RH, Bunte T. 2007. Rapid

diagnostic method for quantitative testing of <100 microbes in water. PDA

Journal Pharmaceutical Science Technology. 61(5): 41-420.

142

Yengi LG, Leung L, Kao J. 2007. The evolving role of drug metabolism in drug

discovery and development. Pharmceutical Research 24 (5): 842-858.