133,99 €
Now in its third edition, the text covers all aspects of biopharmaceutical manufacturing. Structured like a textbook, it is aimed at a wide audience in industry and academia and can be used as a reference as well as for training purposes.
Sie lesen das E-Book in den Legimi-Apps auf:
Seitenzahl: 711
Veröffentlichungsjahr: 2021
Cover
Title Page
Copyright
Medical disclaimer
Preface to Third Edition
Preface to First Edition
List of Abbreviations
Part I: Introduction
1 Biopharmaceutical Production: Value Creation, Product Types, and Biological Basics Introduction
1.1 Role of Production in Pharmaceutical Biotechnology
1.2 Product Groups
1.3 Basics of Biology
Part II: Technology
2 Manufacturing Process
2.1 Role of the Manufacturing Process in Biotechnology
2.2 Process Schematic and Evaluation
2.3 Cell Bank
2.4 Fermentation
2.5 Purification
2.6 Formulation and Filling
2.7 Labeling and Packaging
3 Analytics
3.1 Role of Analytics in Biotechnology
3.2 Product Analytics
3.3 Process Analytics
3.4 Environmental Monitoring
3.5 Raw Material Testing
3.6 Product Comparability
Part III: Pharmacy
4 Pharmacology and Drug Safety
4.1 Action of Drugs in Humans
4.2 Routes and Forms of Administration
4.3 Drug Study
4.4 Path of the Drug from the Manufacturer to Patients
4.5 Drug Safety
Part IV: Quality Assurance
5 Fundamentals of Quality Assurance
5.1 Basic Principles
5.2 Benefit of Quality Assurance Activities
5.3 Quality Management According to ISO 9000
5.4 Structure of Quality Management Systems
5.5 Quality Management System Components in the Pharmaceutical Area
5.6 Quality Assurance in Development
6 Quality Assurance in Manufacturing
6.1 GMP
6.2 Operative Workflows under GMP Conditions
6.3 Production of Investigational Drugs
Appendix A: Case Study Part IV: Warning Letters by FDA
Part V: Pharmaceutical Law
7 Pharmaceutical Law and Regulatory Authorities
7.1 Fields of Pharmaceutical Law
7.2 Bindingness of Regulations
7.3 Authorities, Institutions, and Their Regulations
7.4 Official Enforcement of Regulations
7.5 Drug Approval
Appendix B: Case Study Part V: Clinical Trials for Protein Products
B.1. Mabthera®/Rituxan®
B.2. Enbrel®
B.3. Remicade® Infliximab
B.4. Humira® 40 mg
B.5. Lucentis®
B.6. Zaltrap®
Part VI: Production Facilities
8 Facility Design
8.1 Basic Principles
8.2 GMP-Compliant Plant Design
8.3 Basic Concepts for Production Plants
8.4 Clean and Plant Utilities
8.5 Equipment Cleaning
8.6 Clean Rooms
8.7 Automation
8.8 QC Laboratories
8.9 Location Factors
9 Planning, Construction, and Commissioning of a Manufacturing Plant
9.1 Steps of the Engineering Project
9.2 Project Schedules
9.3 Cost Estimates
9.4 Organization of an Engineering Project
9.5 Successful Execution of an Engineering Project
9.6 Legal Aspects of Facility Engineering
Part VII: Economy
10 Production Costs
10.1 Drug Life Cycle
10.2 Position of the Manufacturing Costs in the Overall Cost Framework
10.3 Basic Principles of Cost Calculation
10.4 Costs of Biotechnological Manufacturing Processes
10.5 Accounting Methods
11 Investments
11.1 Basic Principles
11.2 Value–Benefit Analysis
11.3 Investment Appraisal
11.4 Dynamic Payback Time
12 Production Concept
12.1 Capacity Planning
12.2 Dilemma of In-House Manufacturing
12.3 Aspects of Manufacturing Outsourcing
12.4 Make-or-Buy Analysis
12.5 Process Optimization
12.6 Supply-Chain Management
Appendix C: Examples Part VII: Manufacturing Cost Calculation
C.1. Introduction
C.2. Basic Assumptions for Both Production Processes
C.3. Step 1: Production of Product 1 in Dedicated Facility
C.4. Step 2: Addition of a Second Product
Part VIII: Production Organization and Digitalization
13 Organization of a Manufacturing Facility
13.1 Functional Setup of a Manufacturing Plant
13.2 Development of a Plant Organization
13.3 Organizational Charts and Cooperation Pathways
13.4 Cultural Aspects: The Human Factor
14 Digitalization
14.1 Operational and Digital Perspective
14.2 Digital Maturity
14.3 Integration and IT Architecture
14.4 Digital Transformation
14.5 Digital Applications in the GMP Environment
References
Further Reading
Fermentation
Purification
Aseptic Filling and Lyophilization
Bioanalytics
Regulatory
Pharmacy and Clinical Development
Quality and Validation
Good Manufacturing Practice
Facility Design
Clean Rooms
Project Management
Engineering
Economy
Weblinks
Index
End User License Agreement
Chapter 1
Table 1.1 Examples for recombinant proteins.
Chapter 2
Table 2.1 Comparison between batch and continuous culture.
Table 2.2 Differences between suspension fermenters for cell culture and mic...
Table 2.3 Examples for matrix materials and their brand names.
Table 2.4 Labeling of chromatography gels.
Chapter 3
Table 3.1 Methods for structural analysis of proteins.
Table 3.2 Methods for physical data of protein solutions.
Table 3.3 Methods for identification of impurities.
Table 3.4 Methods for identification of contaminants.
Chapter 4
Table 4.1 Phases of clinical testing.
Table 4.2 Sources for post-marketing data collection of safety-relevant info...
Chapter 5
Table 5.1 Examples of “Good Practice” rules.
Table 5.2 Categories of quality costs (Schneppe and Müller 2003).
Table 5.3 Examples of documentation in the pharmaceutical area.
Chapter 6
Table 6.1 Structure of the EU GMP Guideline (EudraLex, Vol. 4).
Table 6.2 Clean-room gowning for different clean-room classes.
Table 6.3 Different storage areas have to be set up for different substance ...
Table 6.4 Regulatory classification of changes to the manufacturing process.
Chapter 7
Table 7.1 EudraLex – the European comprehensive body of legislation for the ...
Table 7.2 List of the ICH Q series (quality documents).
Table 7.3 ISPE baseline pharmaceutical engineering guides.
Chapter 8
Table 8.1 Chances and risks for realization of high technical complexity and...
Table 8.2 Impact of basic design concepts (rows) on optimization parameters ...
Table 8.3 Water qualities and their areas of use.
Table 8.4 Phases of the qualification of a WFI system.
Table 8.5 Utility users and suppliers.
Table 8.6 Generation of utilities.
Table 8.7 Overview of types of waste and usual disposal routes.
Chapter 9
Table 9.1 Phases of facility engineering.
Chapter 10
Table 10.1 Cost positions of fermentation.
Table 10.2 Cost positions of purification.
Table 10.3 Cost positions of filling and packaging.
Table 10.4 Types of calculations.
Chapter 11
Table 11.1 Classification of investments according to the investment object.
Table 11.2 Effect of investments.
Table 11.3 Other features of investments.
Chapter 12
Table 12.1 Calculation scheme and examples for processing times in fermentat...
Table 12.2 Types and features of cooperation.
Table 12.3 Items to be regulated in manufacturing and supply agreements.
Table 12.4 Overview of in-house (“make”) and external (“buy”) manufacturing.
Appendix C
Table C.1 Cost structure of example step 1.
Table C.2 Cost structure of example step 2.
Table C.3 Costs of products.
Table C.4 Costs comparison between purely internal and mixed option.
Chapter 13
Table 13.1 Typical functional split in a manufacturing facility.
Chapter 14
Table 14.1 Typical IT systems in a pharmaceutical manufacturing plant with m...
Chapter 1
Figure 1.1 Role and tasks of production.
Figure 1.2 Subject areas in production. Inner circle...
Figure 1.3 Relationship between production and development.
Figure 1.4 Relationship between production and marketing.
Figure 1.5 Schematic production workflows of important product groups. Produ...
Figure 1.6 Schematic comparison of traditional therapy and cell and gene the...
Figure 1.7 Schematic of a prokaryotic cell.
Figure 1.8 Schematic of a eukaryotic cell.
Figure 1.9 Schematic of energy metabolism.
Figure 1.10 Schematic of cell division.
Figure 1.11 Intentional and unintentional addition of viruses (or phages) in...
Figure 1.12 Basic principles of protein biosynthesis.
Figure 1.13 Protein structures.
Figure 1.14 Molecular structure of human insulin. S–S disulfide bridges betw...
Figure 1.15 Schematic of antibody IgG.
Figure 1.16 Amphoteric behavior of proteins. The ambient pH value determines...
Figure 1.17 Schematic of a cytoplasmic membrane with lipids.
Chapter 2
Figure 2.1 Variability of the process generates variable product quality.
Figure 2.2 Basic terms of manufacturing of biopharmaceutical products.
Figure 2.3 Schematic of the manufacturing process of the drug substance. Chr...
Figure 2.4 Schematic of the manufacturing process of the final product (phar...
Figure 2.5 Typical path for the generation and use of the cell bank.
Figure 2.6 Development of the cell mass and the target protein concentration...
Figure 2.7 Schematic of fermentation modes: batch, fed-batch, and continuous...
Figure 2.8 Sterilization kinetics. The achievable germ reduction depends on ...
Figure 2.9 Typical fermenter for suspension culture with microorganisms or a...
Figure 2.10 Principles of reactors for adherently growing cell cultures....
Figure 2.11 Typical installation for a batch or fed-batch process for mammal...
Figure 2.12 The path from harvest to purified target protein. (a) Host cell ...
Figure 2.13 Interaction between overall yield, step yield, and number of sep...
Figure 2.14 Product purity and volume from harvest through active agent. The...
Figure 2.15 Sources of process-related impurities.
Figure 2.16 Methods for cell separation (centrifuge, chamber-filter press) a...
Figure 2.17 Primary treatment of proteins expressed intracellularly.
Figure 2.18 Adsorption and desorption mechanism.
Figure 2.19 Adsorption in batch and expanded-bed mode.
Figure 2.20 Normal-flow filtration and TFF. Both can be operated as surface ...
Figure 2.21 Schematic of micro- and ultrafiltration (UF) in the TFF mode and...
Figure 2.22 Membrane processes and their potential applications for the sepa...
Figure 2.23 Schematic of the chromatography operation in the binding mode (I...
Figure 2.24 Schematic of chromatography operation in the flow-through mode....
Figure 2.25 Mode of operation of an IEC (here anion exchanger).
Figure 2.26 Mode of operation of affinity chromatography.
Figure 2.27 Mode of operation of SEC and HIC.
Figure 2.28 Design of a chromatography installation. Enlargement shows the g...
Figure 2.29 Principles of precipitation and extraction.
Figure 2.30 Separation characteristic of an ultrafiltration membrane (Rauten...
Figure 2.31 Typical separation process for an extracellularly expressed prot...
Figure 2.32 Typical separation process for an extracellularly expressed prot...
Figure 2.33 Schematic of formulation and filling.
DS
=
drug substance
.
Figure 2.34 Position of freeze-drying in the formulation process.
Figure 2.35 Labeling and secondary packaging.
Chapter 3
Figure 3.1 The four areas of quality control.
Figure 3.2 Concept of measure for activity. The effect achieved with a defin...
Figure 3.3 Schematic of amino acid analysis.
AA
=
amino acid
.
Figure 3.4 Schematic of Edman protein sequencing. AA = amino acid.
Figure 3.5 Schematic of peptide mapping.
m
/
z
= mass-to-charge ratio.
Figure 3.6 Principle of SDS–PAGE. Left...
Figure 3.7 Isoelectric focusing.
Figure 3.8 Schematic of capillary gel electrophoresis.
Figure 3.9 Western blot (immunoelectroblot). GE = gel electrophoresis.
Figure 3.10 Schematic of a HCP ELISA.
AB
=
antibody
; FERM...
Figure 3.11 Typical chromatogram.
Figure 3.12 The essential elements of a mass spectrometer.
m
/
z
...
Figure 3.13 Principle of nucleic acid sequencing. A, C, G, T...
Figure 3.14 Typical plot of DSC.
Figure 3.15 Analytics around the fermenter.
Figure 3.16 Process analytics in purification.
Figure 3.17 Process analytics in formulation and packaging.
Figure 3.18 Sources of contamination of the process.
Figure 3.19 Workflow for evaluating comparability. Comparison of product qua...
Chapter 4
Figure 4.1 Correlation between drug quantity, drug effect, mechanism of acti...
Figure 4.2 Pathways of drugs in the body at systemic application.
Figure 4.3 Time–concentration curve for single dosing.
C
0
...
Figure 4.4 Time–concentration curve for multiple dosing.
Figure 4.5 (a) Quantification of PD parameters by analog data collection and...
Figure 4.6 Times, costs, and risks during drug development.
Figure 4.7 Path of the drug from the manufacturer to the patient (example fo...
Figure 4.8 Avoidable and non-avoidable causes for detrimental side-effects (...
Chapter 5
Figure 5.1 Fields of activity of quality assurance according to ISO 9001.
Figure 5.2 Documentation inside a
quality management
(
QM
) system (Modified f...
Figure 5.3 Flow diagram for the control of defective goods before delivery. ...
Figure 5.4 Sources of errors and preventive measures in manufacturing.
Chapter 6
Figure 6.1 GMP requirements increase with increasing market supply proximity...
Figure 6.2 Clean rooms in the process chain of biotechnological manufacturin...
Figure 6.3 Prevention of cross-contamination in different plant concepts. Ty...
Figure 6.4 V-model of qualification.
Figure 6.5 Documentation in manufacturing and testing.
MBR
=
master batch re
...
Figure 6.6 Process of product release under GMP. QC = quality control;
QA
=
Figure 6.7 Management of changes to the manufacturing process. QC...
Chapter 7
Figure 7.1 Bindingness of regulations. See text for abbreviations.
Figure 7.2 Organization of the FDA (excerpt).
Chapter 8
Figure 8.1 Functional areas of a biotechnological production facility.
Figure 8.2 Principal functional areas and flows of the process core and supp...
Figure 8.3 Process flow diagram for purification. Chrom...
Figure 8.4 Example of a conceptual design for a plant with mobile equipment....
Figure 8.5 (a) Product flow in the plant with mobile equipment. (b) Personne...
Figure 8.6 Room classifications in the production plant. White...
Figure 8.7 Air pressure steps and mapping of ventilation systems. Colored fr...
Figure 8.8 Conflicts of interest when optimizing a production plant for the ...
Figure 8.9 Integrated and fractal construction. Expansion areas are dashed....
Figure 8.10 Different configurations of flexible piping. (a) Transfer panel ...
Figure 8.11 Typical process steps for generating pharma-grade water from dri...
Figure 8.12 Elements of a water system, AP system, CIP system, WFI system, a...
Figure 8.13 Heating and cooling loops in a production plant.
Figure 8.14 Typical configuration of a CIP/SIP system. NaOH...
Figure 8.15 (a) Typical personnel air lock (side view) and (b) procedure for...
Figure 8.16 Example of a room-in-room concept.
Figure 8.17 Typical configuration of an HVAC system for clean rooms.
Chapter 9
Figure 9.1 Planning costs and failure cost risk in the planning phases.
Figure 9.2 Typical project schedule for the planning and construction of a p...
Figure 9.3 Recommended agreement structure (Modified from Braganz (2001).)
Chapter 10
Figure 10.1 Life cycle curve of a drug. Dashed line...
Figure 10.2 Overview over the profit/loss situation of a drug during develop...
Figure 10.3 Relations between investment, manufacturing costs, sales, and pr...
Figure 10.4 Basic scheme of costs (overhead calculation).
Figure 10.5 Steps of the decision-making process.
Figure 10.6 Influence of plant utilization on fixed, variable, and unit cost...
Figure 10.7 The areas of cost accounting.
Figure 10.8 Types of manufacturing costs.
Figure 10.9 Cost allocation in overhead calculation.
Figure 10.10 Options for parameters and calculation segments for the calcula...
Figure 10.11 Left = full costing; right = direct costing for the calculation...
Chapter 11
Figure 11.1 Life cycle of capital investment.
Figure 11.2 Decision process for investments with example for a facility con...
Figure 11.3 Different scaling methods in evaluation matrices of value–benefi...
Figure 11.4 Overview of static methods of investment appraisal. Comparison o...
Figure 11.5 Calculation of the NPV from the series of payments of an investm...
Chapter 12
Figure 12.1 Typical schedule for the implementation of market supply for a b...
Figure 12.2 Typical time line for technology transfer. Small-scale model...
Figure 12.3 Typical schedule for the implementation of market supply for a b...
Figure 12.4 DMAIC cycle.
Figure 12.5 Process capability. (a) Statistical process control. (b) Definit...
Figure 12.6 Process capability. (a) Process potential
C
p.
(b) Process capabi...
Figure 12.7 Supply chain for the market supply of a drug and typical values ...
Figure 12.8 Risk matrix with three risk examples, likely to trigger differen...
Appendix C
Figure C.1 Product unit costs dependency on facility usage (34 batches equal...
Figure C.2 Average costs per grams for two products plotted over external of...
Chapter 13
Figure 13.1 Production management and value flow. Up-/downstream: fermentati...
Figure 13.2 Swim lanes and exemplary RACI chart for the process: material de...
Figure 13.3 Typical organizational chart of a manufacturing plant. Up-/downs...
Chapter 14
Figure 14.1 Digital plant maturity model (Anttonen et al. 2020). ERP: enterp...
Figure 14.2 Horizontal and vertical integration of IT-based systems and comp...
Figure 14.3 IT-architecture models for integrating systems.
DSS
:
domain-spec
...
Figure 14.4 Digital strategy and functional layers connecting business and i...
Cover Page
Table of Contents
Title Page
Copyright
Medical disclaimer
Preface to Third Edition
Preface to First Edition
List of Abbreviations
Begin Reading
References
Index
End User License Agreement
iii
iv
v
xvii
xix
xx
xxi
xxii
xxiii
xxiv
xxv
1
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
33
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
61
62
63
64
65
66
67
68
69
70
71
72
73
74
75
76
77
78
79
80
81
82
83
84
85
86
87
88
89
90
91
92
93
94
95
96
97
98
99
100
101
103
104
105
106
107
108
109
110
111
112
113
114
115
116
117
118
119
120
121
122
123
124
125
126
127
128
129
130
131
132
133
134
135
136
137
138
139
141
143
144
145
146
147
148
149
150
151
152
153
154
155
156
157
158
159
160
161
162
163
164
165
166
167
168
169
171
173
174
175
176
177
178
179
180
181
182
183
184
185
186
187
188
189
190
191
192
193
194
195
196
197
198
199
200
201
202
203
204
205
206
207
208
209
210
211
212
213
214
215
216
217
219
220
221
223
225
226
227
228
229
230
231
232
233
234
235
236
237
238
239
240
241
242
243
244
245
246
247
248
249
251
252
253
254
255
256
257
258
259
260
261
262
263
264
265
266
267
268
269
270
271
272
273
274
275
276
277
278
279
280
281
282
283
284
285
286
287
288
289
290
291
292
293
294
295
296
297
298
299
300
301
302
303
304
305
306
307
308
309
310
311
312
313
314
315
316
317
318
319
321
322
323
324
325
326
327
328
329
330
331
332
333
334
335
336
337
338
339
340
341
342
343
344
345
346
347
348
349
351
352
353
354
355
356
357
358
359
360
361
362
363
364
365
366
367
368
369
370
371
372
373
374
375
376
377
378
379
380
381
382
383
384
385
386
387
388
389
390
391
392
393
394
395
396
397
398
399
400
401
402
403
405
406
407
408
409
410
411
413
415
416
417
418
419
420
421
422
423
424
425
427
428
429
430
431
432
433
434
435
436
437
438
439
440
441
442
443
444
445
446
447
448
449
451
452
453
454
455
457
458
459
460
461
462
463
464
465
466
467
468
469
Stefan Behme
Third Edition
Author
Dr.-Ing. Stefan Behme
Berlin, Germany
Chlumer Str. 3
12203 Berlin
Germany
Cover Design: Wiley
Cover Images: © Artisticco/Shutterstock;Reptile8488/Getty Images
All books published by WILEY-VCH are carefully produced. Nevertheless, authors, editors, and publisher do not warrant the information contained in these books, including this book, to be free of errors. Readers are advised to keep in mind that statements, data, illustrations, procedural details or other items may inadvertently be inaccurate.
Library of Congress Card No.: applied for
British Library Cataloguing-in-Publication Data
A catalogue record for this book is available from the British Library.
Bibliographic information published by the Deutsche Nationalbibliothek
The Deutsche Nationalbibliothek lists this publication in the Deutsche Nationalbibliografie; detailed bibliographic data are available on the Internet at <http://dnb.d-nb.de>.
© 2022 WILEY-VCH GmbH, Boschstr. 12, 69469 Weinheim, Germany
All rights reserved (including those of translation into other languages). No part of this book may be reproduced in any form – by photoprinting, microfilm, or any other means – nor transmitted or translated into a machine language without written permission from the publishers. Registered names, trademarks, etc. used in this book, even when not specifically marked as such, are not to be considered unprotected by law.
Print ISBN: 978-3-527-34947-0
ePDF ISBN: 978-3-527-83379-5
ePub ISBN: 978-3-527-83380-1
oBook ISBN: 978-3-527-83381-8
Medical disclaimer
The contents of this work are intended to further general scientific research, understanding, and discussion only and are not intended and should not be relied upon as recommending or promoting a specific method, diagnosis, or treatment by medical care providers for any particular patient. The publisher and the author make no representations or warranties with respect to the accuracy or completeness of the contents of this work and specifically disclaim all warranties, including without limitation, any implied warranties of fitness for a particular purpose. In view of ongoing research, equipment modifications, changes in governmental regulations, and the constant flow of information relating to the use of medicines, equipment, and devices, the reader is urged to review and evaluate the information provided in the package insert or instructions for each medicine, equipment, or device for, among other things, any changes in the instructions or indication of usage and for added warnings and precautions. Readers should consult with a specialist where appropriate. The fact that an organization or Website is referred to in this work as a citation and/or a potential source of further information does not mean that the author or the publisher endorses the information the organization or Website may provide or recommendations it may make. Further, readers should be aware that Internet Websites listed in this work may have changed or disappeared between when this work was written and when it is read. No warranty may be created or extended by any promotional statements of this work. Neither the publisher nor the author shall be liable for any damages arising herefrom.
What started out over ten years ago as being a book focused on protein manufacturing, in the meantime has evolved into a broad introduction of many different aspects of pharmaceutical operations. All but two sections – technology and regulatory – can be applied to any pharma production, which also gives testimony to the maturity and standardization that pharmaceutical protein manufacturing has achieved by now. The third edition of this book has been amended by two new chapters about plant organization and digitalization, broadly applicable to pharma. The general, conceptual, and simplifying approach makes the book a valuable source also for those working on transferring advanced pharmaceutical technologies like cell and gene therapies into economically feasible, large-scale solutions.
The original concept of the book – keep it simple and speak through pictures and examples – has been kept alive. The concept of strong simplification has been well received by many readers over the last years. Dipping into specific chapters of interest and quickly getting familiar with basic concepts and terminology has obviously addressed the needs of readers both in industry and academia. So, I am very happy to present the third edition of this book now and thank my editor Wiley-VCH for the ongoing support.
Berlin, 23 May 2021
Stefan Behme
This book introduces the basic knowledge of industrial manufacturing of biopharmaceuticals. It is written for those wanting to understand the landscape, interfaces, and interactions between the different disciplines relevant for production as such; aspects of technology and analytics, pharmacy, quality assurance, regulatory affairs, facility technology, and economic efficiency are illustrated. The work shall serve as a textbook and reference at the same time, and is directed toward students as well as industry-experienced engineers, pharmacists, scientists, or economists wanting to acquire a basic knowledge of biotechnological production.
My daily industrial practice has inspired this book. Manufacturing advanced drugs under good manufacturing practice conditions can indeed be a critical factor for drug development and marketing. Being part of multidisciplinary teams, it became obvious to me that the technological and economic challenges of biopharmaceutical manufacturing and its interdependencies with adjacent disciplines are not understood everywhere. Decision making in interdisciplinary teams requires communication and appreciation of the constraints on the various counterparts in order to address them efficiently in the overall program. In contrast to this, particular disciplines become more and more specialized, using their language on a level difficult to understand for the counterparts foreign to the field, sometimes flavoring modern project work with a taste of the tale of the Tower of Babel.
Facilitating communication about manufacturing issues is the goal of this book. It does so by using numerous illustrations and simplifications, making the book easy to read. Correlations between disciplines are highlighted by cross-references, and a detailed keyword index facilitates the search for special topics. After having read this book, the reader should have a high-level understanding of the roles, correlations between terminologies of the different disciplines engaged in the production of biopharmaceutical proteins. For those wanting to dig deeper into the topics, literature recommendations and web links are provided for further reading.
I would like to thank Andrea Rothmaler and Andreas Janssen for their valuable input into the manuscript, my students at the Technical University of Dortmund for their instructive questions, and my company Bayer Schering Pharma AG for providing the opportunity to participate in exciting biotechnological projects.
I hope that my readers will enjoy reading this book as much as I have enjoyed writing it.
Berlin, October 2008
Stefan Behme
AA
Amino Acid (= AS)
ADR
Adverse Drug Reaction
AE
Adverse Event
AIEX
Anion Exchanger
AMG
Arzneimittelgesetz
AMWHV
Drug and drug manufacturing Regulation
AP
Aqua Purificata
API
Active Pharmaceutical Ingredient
APR
Annual Product Review
AR
Adverse Reaction (= ADR)
AR
Annual Report
ATP
Adenosine Triphosphate
AUC
Area Under the Curve
AVP
Aqua Valde Purificata
BAS
Building Automation System
BDS
Bulk Drug Substance
BLA
Biological License Application
BOD
Basis of Design
BP
Basen Pair
BPMN
Business process model and notation
BR
Batch Record
BRR
Batch Record Review
BSE
Bovine Spongiforme Encephalopathie
CAPA
Corrective Action Preventive Action
CBE30
Changes Being Effected in 30 days
CDW
Cell Dry Weight
CFR
Code of Federal Regulations
CFU
Colony Forming Unit
cGMP
Current Good Manufacturing Practice
CI
Chemical Ionization
CIEX
Cation Exchanger
CIP
Cleaning in Place
CJD
Creutzfeldt–Jakob Disease
CMC
Chemistry, Manufacturing, and Control
CMO
Contract Manufacturing Organization
CoA
Certificate of Analysis
CoC
Certificate of Compliance
COP
Cleaning out of Place
CRF
Case Report Form
CSV
Computerized system Validation
CTA
Clinical Trials Authorization
CTD
Common Technical Document, Clinical Trials Directive
CVMP
Committee for Medicinal Products for Veterinary Use
DIN
Deutsches Institut für Normung
DNA
Desoxyribonucleic Acid
DPPM
Digital plant maturity model
DQ
Design Qualification
DSC
Differential Scanning Calorimetry
EBR
Electronic Batch Record
ED
Effective Dose
EDQM
European Directorate for the Quality of Medicines
EIS
Electron Impact Spectroscopy
ELISA
Enzyme Linked Immunosorbent Assay
EMA
European Medicines Agency
EP
European Pharmacopoeia (PharmEur)
EPO
Erythropoietin
ERM
Enterprise recipe management
ERP
Enterprise resource planning
ETL
Extract–transform–load
FaaS
Function as a service
FAB
Fast Atom Bombardment
FBS
Fetal Bovine Serum
FCS
Fetal Calf Serum
FDA
Food and Drug Administration
FMEA
Failure Mode and Effect Analysis
FP
Final Product, Finished Product
HMI
Human machine interface
GAMP
Good Automated Manufacturing Practice
GCP
Good Clinical Practice
G-CSF
Granulocyte Colony Stimulating Factor
GEP
Good Engineering Practice
GFC
Gel Filtration Chromatography
GLP
Good Laboratory Practice
GM-CSF
Granulocyte Macrophage Colony Stimulating Factor
GMO
Genetically Modified Organism
GMP
Good Manufacturing Practice
GPC
Gel Permeation Chromatography
GSP
Good Storage Practice
GSS
Gerstmann–Sträussler Syndrom
GTP
Good Tissue Practice
HCP
Host Cell Protein
HIC
Hydrophobic Interaction Chromatography
HIV
Human Immunodeficiency Virus
HPLC
High Pressure Liquid Chromatography (also High Performance LC)
HPMC
Hydroxypropylmethyl-cellulose
HSA
Human Serum-Albumin
HVAC
Heat Ventilation Air Conditioning
IaaS
Infrastructure as a service
ICH
International Conference on Harmonization
IEF
Isoelectric Focusing
JEC
Jon Exchange Chromatography
IEX
Ion Exchanger
IF
Interferon
IGG
Immunoglobulin G
IIoT
Industrial internet of things
IL
Interleukin
IMP
Investigational Medicinal Product
IMPD
Investigational Medicinal Product Dossier
IND
Investigational New Drug
IOM
Investigations Operations Manual
IPC
In-Process Control
IQ
Installation Qualification
IR
Infrared
ISO
International Organization of Standardization
ISPE
International Society for Pharmaceutical Engineering
JP
Japanese Pharmacopoeia
KPI
Key Performance Indicator
LADME
Liberation, Absorption, Distribution, Metabolism, Excretion
LAL
Limulus Amebocyte Lysate
LD
Lethal Dose
LES
Laboratory execution system
LFH
Laminar Flow Hood
LIMS
Laboratory Information Management System
LOD
Limit of Detection
LOQ
Limit of Quantification
MALDI
Matrix Assisted Laser Desorption Ionization
MBR
Master Batch Record
MCB
Master Cell Bank
MCO
Molecular Cut Off (MWCO)
MES
Manufacturing execution system
MF
Microfiltration
MHLW
Ministry of Health, Labor, and Welfare
MSA
Manufacturing and Supply Agreement
MTD
Maximal Tolerated Dose
MWCO
Molecular Weight Cut Off
NDA
New Drug Application
NIST
National Institute of Standards and Technology
NPV
Net Present Value
OOS
Out of Specification (QC Context) or Out of Stock (Logistical Context)
OQ
Operational Qualification
PAB
Pharmaceutical Affairs Bureau
PAGE
Polyacrylamid Gel Elektrophoresis
PAS
Prior Approval Supplement
PAT
Process analytical technology
PCR
Polymerase Chain Reaction
PD
Pharmacodynamics
PD
Plasma Desorption
PDA
Parenteral Drug Association
PEG
Polyethylene glycol
PFBS
Pharmaceutical and Food Safety Bureau
PharmEur
European Pharmacopoeia
PIC/S
Pharmaceutical Inspection Convention/Scheme
PK
Pharmacokinetics
PLC
Programmable logic controller
PM
Posttranslational Modification
PMDA
Pharmaceutical and Medical Devices Agency (KIKO)
PoC
Proof of Concept (PoP)
PoP
Proof of Principle (PoC)
PQR
Product Quality Review
QA
Quality Assurance
QAA
Quality Assurance Agreement
QC
Quality Control
QM
Quality Management
rFVIII
Recombinant Factor VIII
RNA
Ribonucleic Acid
ROI
Return on Investment
RPC
Reversed Phase Chromatography
RP-HPLC
Reversed Phase HPLC
RPM
Regulatory Procedures Manual
SCADA
Supervisory control and data acquisition
SDS
Sodiumdodecylsulfate
SEC
Size Exclusion Chromatography
SIP
Sterilization in Place (also Steaming in Place)
SKU
Stock Keeping Unit
SOP
Standard Operating Procedure
SPC
Statistical Process Control
SPC
Supplementary Protection Certificate
TEM
Transmission Electron Microskopy
TFF
Tangential Flow Filtration
TOC
Total Organic Carbon
TOF
Time of Flight
TSE
Transmissible Spongiform Encephalopathie
UF
Ultrafiltration
UML
Unified modeling language
URS
User Requirements Specification
USP
United States Pharmacopoeia
UV
Ultra Violet
WCB
Working Cell Bank
WFI
Water for Injection
WHO
World Health Organization
ZLG
Zentralstelle fur Gesundheitsschutz bei Arzneimitteln und Medizinprodukten
Over recent years, pharmaceutical biotechnology has developed very dynamically. An important driver for this success has been the enormous increase of scientific know-how in the areas of genetics and immunology, which has created huge expectations for the development of innovative medicinal treatments.
The scientific pioneer spirit has been fueled by public and private sponsorship, resulting in a biotechnological landscape that has long been dominated by highly innovative, venture capital-based, small- and mid-size companies. However, before patients can benefit from scientific achievements, it is necessary that the identified molecule is transformed into a medicine – fit for achieving the therapeutic target – and tested in comprehensive trials in the field. The production of such a medicine has to be carried out in officially licensed, often tailor-made technical manufacturing facilities.
From project to product
This path from project to product usually lasts several years and is associated with enormous costs and risks. On average, the development costs of a new compound are in the region of US$ 500–1000 million and only 10% of all projects that enter clinical trials find their way into the market.
Owing to these immense investments in drug development, the costs of drug manufacturing often seem acceptable, particularly as the costs are absorbed by sales of the marketed drug in the same accounting period; however, safe and efficient product supply is the cornerstone of a company's success. In biotechnology, the overlap between development and market launch is particularly intensive, motivating companies to take care of manufacturing early on:
Many targets of process development result from requirements of large-scale manufacturing.
The classical separation of development (pre-marketing) and production (post-marketing) does not work for biologics, as both the manufacturing process and plant are factors that determine the quality of the final medicinal product.
Production is the basis for long-term market supply. Decisions about capital investment or outsourcing of manufacturing mostly have to be taken long before the market launch of the product.
Biotechnological processes are much more difficult to control than small-molecule preparations. The limited ability to monitor and characterize the product results in increased manufacturing risks.
Significance of production in the value chain
The main target of production is to supply the product safely and cost-efficiently. It is positioned between the development and marketing of a product. Figure 1.1 illustrates its significance in the value chain.
The chain starts with research that has a clear focus on the identification of targets, which involves analyzing the interaction between the biochemical molecule and its potential therapeutic functionality. In the subsequent development phase, a process for the scale-up and more consistent manufacturing of the molecule is designed. Here, the target structure is developed into a pharmaceutical form and tested in animals and humans as to its safety and efficacy. Once this is achieved, production kicks in, taking care of a high-quality and profitable product supply, addressing the following main tasks:
When, where, and in what quantities should the drug be produced? (
Production concept
)
Figure 1.1 Role and tasks of production.
How should market supply be organized? (
Supply-chain organization
)
How should the quality of the product and
Good Manufacturing Practice
(
GMP
) compliance be assured? (
Quality assurance
)
What are costs of manufacturing and how can these costs be controlled? (
Manufacturing costs
)
How attractive is an investment in one's own facilities? (
Investment decision
)
The marketing of the product stands at the end of the value chain; from this position, essential goals are formulated for production: supply safety and cost efficiency.
Production is interdisciplinary
The integrated position of production in the value chain results in interdisciplinary tasks that are best treated by multilateral teams managed by experts in different disciplines such as biology, engineering, chemistry, economics, law, pharmacy, and medicine.
Figure 1.2 shows the subject areas that are important for the understanding and control of production processes and workflows. This volume provides an overview of these subject areas, while special emphasis is given to the interaction between these areas.
Figure 1.2 Subject areas in production. Inner circle = sections of this book; outer circle = subject areas treated in the sections.
Following this introductory part, Part II, “Technology,” focuses on processes and analytics. This section illustrates why the manufacturing process plays such a large role in biotechnology, and to what extent product quality is determined by processes and analytics. Moreover, essential technologies for industrial manufacturing as well as methods and areas of application of analytical testing are described.
Part III, “Pharmacy,” briefly elaborates on the basic principles of drug effects on humans and the essential steps of pre-clinical and clinical drug studies. The successful end of the clinical test marks the starting point of commercialization.
Product quality plays a crucial role in pharmaceutical manufacturing. Part IV, “Quality Assurance,” elucidates the organizational and operative workflows for quality assurance, including the rules of GMP.
Almost all activities of commercial production happen in the framework of legal regulations. Part V, “Pharmaceutical Law,” describes drug regulations and laws as well as institutions and enforcing official authorities.
The translation of process technology into large-scale manufacturing capacities is described in Part VI, “Production Facilities.” Basic principles of the design of GMP-compliant manufacturing facilities are given and different building concepts compared. The planning process that leads to industrial plants is illustrated. Here, we include a brief look at the regulations regarding health, safety, environment, and construction that form the legal framework of industrial production facilities.
Commercial thinking is the spine of efficient production. Part VII, “Economy,” introduces essential principles around product sales and cost of goods accounting. It compares concepts of in-house manufacturing with outsourcing strategies and elucidates the decision factors leading to capital investments in biotechnological plants.
The book closes with a Bibliography providing literature and web references and an appendix providing a list of abbreviations and an alphabetical index of keywords.
It is widely understood that production starts when development provides a marketable product and a commercially feasible manufacturing process. Ongoing market supply is secured by process optimization or the provision of additional manufacturing capacities, depending on how market demand develops. For biotechnological pharmaceuticals, the flexibility to react to demand changes is reduced due to the following reasons.
Drug application and the manufacturing process are described and fixed in the regulatory license. As the biotechnological manufacturing process is a quality-determining factor, it has to be finally defined at the time point of regulatory submission and can thereafter be changed only with relatively high effort. The market application contains proof of the safety and efficacy of the drug; it adds to the complexity that in biotechnology, this proof has to be made – at least partly – with material from the commercial process and manufacturing site. Changes to the process or site require comparability exercises that can be more or less complex depending on the risk associated with the change. All of this means that the manufacturing process is fixed at a relatively early time point during development and can only be changed with quite some effort.
Clinical and process development
This coherence is illustrated in Figure 1.3. Product development consists of clinical development, on the one hand, and development of the manufacturing process and the analytical methods, on the other hand. The clinical development renders proof of safe and efficacious use of the drug in humans. Ideally, this proof is generated with material from the process and the site designated for commercial supply. There is a challenge with this ideal approach: if the process would be finally established and only after that clinical development be initiated, the timelines of development would add up unacceptably. Therefore, the different branches in the development workflow occur in parallel; different stages of the clinical development are supplied with different development stages of the manufacturing process.
Figure 1.3 Relationship between production and development.
From lab to large-scale process
Validation and critical parameters
Coming from the laboratory scale, the process is evolved step by step into the final and mature manufacturing process. The scale and maturation a technical process achieves until first used for commercial supply depend on the characteristics of the process, on the requested product demand, and, often, on the available time for development. At the endpoint of development, the process is implemented in the designated commercial supply facility. Product generated in these so-called full-scale runs must be used in representative amounts in the clinical trial. Process validation shows that the drug can be manufactured reproducibly and in good quality under consideration of applicable operating procedures. Product generated in these so-called “validation runs” must usually be used in representative amounts in the clinical trial.
An “easy-to-validate” process means that product quality is essentially independent of fluctuations of the critical process and equipment parameters. Critical parameters as well as measures to control them should be identified in the lab-scale process. These links result in interactions between production and development long before the actual supply to the commercial market.
Role of analytics
Owing to the heterogeneous composition of biological pharmaceuticals, analytical methods play a special role. Just like the process, the developed methods find their way into the regulatory license documentation. Concurrently – while being optimized itself – analytics has to support process development from very early on. Production requirements such as speed, robustness, and simplicity of testing methods have to be taken into account. Moreover, it has to be decided which method should support processing, which method is necessary for product characterization and process validation, and which method should be used only in the development phase.
This short outline illustrates how deeply the aspects of production reach into the development phase. An early recognition of production aspects can help to avoid detours and project delays.
Production facilities
An important interface, that is, not shown in Figure 1.3 is the one to the facility in which the manufacturing process is carried out. The capital investment in a manufacturing plant, and also the alternative contractual obligation with an outside source, means an additional financial risk of considerable size. This issue is further discussed in Part VII.
Production makes the final and packed product available for marketing (Figure 1.4). The packaging provides product protection and a possibility for attracting customers; especially in the pharmaceutical arena, the packaging contains a considerable amount of user information. The coordination and distribution of the country-specific final products is done by production logistics, which has to react flexibly to requirements from sales and marketing.
Figure 1.4 Relationship between production and marketing.
Life-cycle projects support the development of the project in the market place and usually affect not only the pharmaceutical dosage and form but also the indication of the product. In these cases, production has to adapt to changes in demand, packaging materials, or formulation processes.
Cost aspects
The acceptable manufacturing costs are determined by the achievable price in the marketplace, which is often regulated by country-specific reimbursement systems. The construction or maintenance of manufacturing plants has to be justified by adequate profitability calculations that are based on estimates and expectations of the market situation, and the desired profit margin, on the one hand, and the operating and capital expenses, on the other hand. These projections often reach far into the future (more than 10 years) and leave large room for variations.
Specialties of the pharma market
While the aforementioned characteristics also apply to other goods, there are indeed pharma-specific features, for example, the governmental monitoring system, the exceptionally high ethical responsibility of pharmaceutical companies, and the official regulation of drug reimbursement. Safety of patients is guaranteed by instruments used for pharmacovigilance and intensive product quality assurance. Pharmacovigilance systems serve to register unforeseen adverse effects of drugs and route them to the supervisory body. To achieve this goal, the pharmaceutical company collects and evaluates blinded patient data; in case of an unforeseen adverse event, a root-cause analysis has to be performed. To perform this analysis, it is necessary that the specific medicament used by the patient can be traced back to the manufacturing site and batch. It is the specific batch documentation that then provides insight into whether deviations have occurred during the operation that might have influenced the quality of the product. If yes, it needs to be clarified in a second step whether such a quality variation could have triggered the adverse reaction. Thus, the requirements of pharmacovigilance lead to a comprehensive documentation obligation of the entire manufacturing process.
The target of pharmacovigilance is to recognize risks retrospectively. As a complement to the framework of drug safety, there are intensive measures for prospective quality assurance. This has a significant impact on the operational workflows as will be shown in Part IV.
Pharmaceutical biotechnological products can be classified into:
Vaccines derived from non-genetically modified organisms or blood.
Therapeutics from blood or animal organs (e.g., Factor VIII and insulin).
Antibiotics manufactured traditionally in biological processes. Usually this is done with non-genetically modified organisms.
Recombinant proteins (i.e., active ingredients) derived from cultivation of genetically modified cells. Including monoclonal antibodies, these represent the biggest sector of current pharmaceutical biotechnology.
A new branch of therapy opens up with the possibilities of cell and gene therapeutics. These complex interventions into the human body require the reassessment of the pharmaceutical safety concept and demand special precautions from production technology and engineering.
Manufacturing technologies of different product groups can be similar
The focus of the present work lies in the production of therapeutic recombinant proteins; however, the principles described can be applied to the other product groups as well. A closer look at the groups reveals interesting therapeutic and technological overlaps. For example, innovative gene therapy can learn from experiences in virus production gathered in the conventional vaccine field. Also, vaccines will face a new era due to the possibility to produce monoclonal antibodies (Section 1.3.2.1). In the following text, the product groups – with the exception of antibiotics – will be covered in more depth.
Figure 1.5 Schematic production workflows of important product groups. Product groups are shown on the right. Attenuation = elimination of reproducibility, but retention of infectivity; inactivation = elimination of reproducibility and infectivity.
Figure 1.5 schematically shows the production workflows for different product groups. There are differences regarding the genetic modification of the starting material. Genetically modified organisms are mainly deployed for recombinant proteins and gene therapeutics, but cell therapy can also use this technology. The products can be proteins, viruses, bacterial fragments, cells, or intact viruses for gene therapy.
There are two principles of vaccination:
Passive vaccination
: Antibodies against the pathogen are administered.
Active vaccination
: The immune system is confronted with alleviated pathogens and builds up its own immune defense against the causative organism.
Antibodies for passive vaccination are prepared by injecting the pathogen into animals. The immune system of the animals pours out so-called polyclonal antibodies into the blood system. Blood is collected from the animals, and the antibodies isolated and purified, so that they can be administered to humans.
Active vaccination uses inactivated germs that are no longer pathogenic, but still immunogenic. Activation allows the immune system to recognize the real pathogenic germs much faster and therefore fight them before they can spread out and cause the illness. It suffices to present only a moiety instead of the whole pathogen to enable the immune system to recognize the substance foreign to the body. This moiety can be the hull protein of a virus, whole inactivated cells, or pathogen-specific deoxyribonucleic acid (DNA). The general term for these immune response-inducing agents is “antigen.” Active vaccines like the influenza vaccine can be proliferated in chicken eggs and reworked to vaccines.
Many diseases can be attributed to the lack of certain proteins in the blood. In part, these proteins can be extracted from animal or human blood or organs, such as insulin against diabetes or Factor VIII against bleeding disorders. Biotechnology has made it possible for these proteins to be obtained without being tied to these expensive and – under aspects of safety – questionable raw materials from natural sources. In some cases, blood-derived products still play a role as it has not yet been possible to successfully replace them completely by recombinant proteins.
Plasma fractionation
Risks of protein extraction from blood
To isolate the proteins from the blood, it is first separated into its two main components: plasma and cells. The plasma is further fractionated to obtain the proteins. It is associated with considerable analytical and organizational effort to guarantee the safety of the raw material blood, especially the absence of viral contamination and transmissible spongiform encephalopathy (TSE)-inducing components. Despite the intensive surveillance of blood donors, the danger of safety-relevant incidents persists. It can be expected that the production of proteins will be more and more shifted to recombinant technologies, while whole-blood donations will remain irreplaceable for patient treatment in hospitals.
Recombinant proteins, including monoclonal antibodies, by far, make up the largest group of biotechnological pharmaceutical products. Table 1.1 shows some examples; in addition to the medical indication and the functionality in the human organism, it provides details regarding the size and type of the molecule. Section 1.3 gives further insight into the structure of proteins and the terms of amino acids and glycosylation.
A huge growth potential is expected for monoclonal antibodies and antibody fragments.
The starting point for all protein production is the genetic modification of the host cell in which the protein should be expressed. The endpoint usually is a parenterally (per injection) administered product in liquid or solid form.
Cell therapy: implantation of intact cells
Therapeutic proteins are administered to compensate for the lack of the respective natural protein in the organism. As the molecule is eliminated either by degradation or excretion, the administration has to be repeated to achieve a constant active agent level. In contrast, cell and gene therapy is based on the idea of fighting the disease at its source and enabling body cells to express the missing proteins by themselves. Figure 1.6 schematically illustrates the differences between the philosophies of protein versus cell and gene therapy treatment. The starting point is a disease caused by a lack of the example protein X. In conventional therapy, the protein is produced ex vivo and injected into the patient. Owing to elimination processes, the protein disappears after a while. In gene therapy, a genetic sequence is introduced into the body, which contains the construction plan for the desired protein as well as the capability to infect suitable target cells. This combined capability is generated by means of biotechnological methods: the protein-encoding gene is linked to a molecular “ferry” (or vector) that carries the gene into the designated target cells. This ferry is a virus that has been modified in such a way that it retains its infectivity but lacks its ability to replicate. After having been infected the cells start to produce the desired protein. In the ideal case – if the construct is genetically very stable and the expression rate high – this process has to be carried out only once. If the modified viruses are injected directly into the body, it is called in vivo gene therapy, but it is also possible that the cells are extracted from the patient and re-implanted after being infected in the lab (ex vivo gene therapy). The latter is basically a crossover between cell and gene therapy.
Table 1.1 Examples for recombinant proteins.
Name
Indication
Functional group
Number of amino acids; glycosylation, and fraction of sugars of molecular weight; molecular weight
Insulin
Diabetes
Hormone
AA 51; Gly no; 5.8 kDa
Human growth hormone
Dwarfism
Hormone
AA 191; Gly no; 22.1 kDa
Factor VIII
Bleeding disorder
Clotting factor
AA 2332; Gly to 35%; 300 kDa
Lepirudin
Thrombosis
Anticoagulant
AA 64; Gly no; 7 kDa
Tissue plasminogen activator
Thrombosis
Thrombolytic agent
AA 72; Gly to 25%; 72 kDa
Interferon
s (
IFN
s)
Diverse: multiple sclerosis, hepatitis, arthritis, and so on
Immune modulator
IFN-β: AA 166; Gly yes and no; 18.5 kDa
Interleukin
s (
IL
s) (13 different types)
Diverse: asthma, HIV, cancer, mucositis, and so on
Immune modulator, signal agent between immune cells
IL-2: AA 133; Gly yes; 15.5 kDa
Erythropoietin
Anemia
Growth factor
AA 165; Gly to 40%; 34 kDa
Granulocyte colony-stimulating factor
(
G-CSF
),
granulocyte macrophage colony-stimulating factor
(
GM-CSF
)
Infections, cancer
Growth factor
G-CSF: AA 174–180; Gly yes; 19.6 kDa; GM-CSF: AA 127; Gly yes, 15.5, 16.8; and 19.5 kDa
Monoclonal antibodies
Cancer, transplantation, and so on
Antibodies
IgG: AA about 1300; Gly yes; about 150 kDa
Gly = glycosylation; AA = amino acids; Gly to 30% = molecular weight fraction of glycosylation can reach up to 30%.
Figure 1.6 Schematic comparison of traditional therapy and cell and gene therapies.
The basic principle of cell therapy is to convey cells to the body that have the desired functionality; thus, cell therapy does not aim at repairing dysfunctional cells, but rather replacing them. These cells can originate from animals (xenogenic) or humans, either patient proprietary (autologous) or non-patient proprietary (allogeneic). The allogeneic and xenogenic approaches raise questions regarding immunogenic responses, comparable to tissue rejection in organ transplantation. If the cells are genetically modified, they belong to transformed cell lines; cells that have not experienced any genetic modifications are called primary cells. Thus, “allogeneic cell therapy with primary cells” denotes a therapy in which cells of a foreign donor are implanted without genetic modification into the patient. The functionality of cells is not restricted to protein production – other molecules can also be expressed. A prominent area of cell therapy is the replacement of tissue or organs (tissue engineering). Consequently, the widely discussed therapy with human stem cells is only one form of cell therapy, which is characterized by the adoption of non-differentiated stem cells.
Cell production for therapy
Cell therapy production starts with isolating the cells intended for implantation. This is clearly a crucial issue of cell therapy, as the starting material is limited and the subsequent expansion of the cells is restricted by the natural limit of generation numbers. The implantable cells are available for surgical implantation after an additional manipulation step such as washing or buffer exchange.
Gene production for therapy
The genetic construct for gene therapy is produced by proliferating the DNA in a host organism that can be first expanded and subsequently transfected with a modified virus. This infected cell produces the desired virus. To control infectivity, the virus can be weakened (attenuated); at the end of this production process, the product consists of the attenuated, modified virus.
Safety questions around cell and therapy
Cell and gene therapies are at the advent of their development. Despite the fact that the approaches seem plausible, complex questions around drug safety arise. The administration of generally replicable and propagatable substances is very different compared to conventional protein therapy. The scarce source and the handling of the living “cell” system impose new challenges on production and distribution processes.
Antibiotics have been produced biologically for many decades. A penicillin-producing yeast strain is cultivated in a biological fermentation step, and the expressed penicillin is further purified into a pharmaceutical product. In particular, fermentation resembles that with recombinant microbial expression systems (Section 2.3.1). In contrast, purification is different because penicillin is a relatively small and robust molecule compared to proteins.
This section is dedicated to some basic principles of biology and biochemistry that are relevant for the understanding of production processes on the level of this book. After a short outline of cell biology and microbiology, the four basic molecular entities of biochemistry are introduced: proteins, nucleic acids, polysaccharides, and lipids.
Each form of life – plants, animals, or microorganisms – consists of biological cells. While plants and animals constitute themselves as enormous networks of different cell types, microorganisms are predominantly single celled.
Microorganisms can survive in their selected habitat independently, while cells of higher organisms depend on their united cell structure. Cells are characterized by some general features:
Cells contain a carrier of genetic information (double-helical DNA) and a single-strand
ribonucleic acid
(
RNA
) derived thereof. During replication – in the process of propagation – its genetic information is prone to erratic variations (mutation).
Cells exchange nutrients and waste products with the environment (metabolism) for the purpose of energy recovery (catabolism) or substance construction (anabolism).
They are confined by a membrane that allows for controlled substance exchange with the environment.
They communicate via so-called receptors with the environment and can react to changes in external conditions.
They are capable of replicating themselves and a number of higher cells differentiate into other cell types.
In addition to genotypic characterization of cells and organisms, based on gene technology, there is a phenotypic characterization based on the differences in shape, movement pattern, staining (Gram staining), metabolic pattern, and preferred habitat.
Mycoplasmas are special bacteria. They do not possess a cell wall and are exceptionally small. Obviously, they are resistant against types of antibiotics that attack cell walls. As with viruses, they are not retained by filters of a pore size of 0.22 μm. Some members of this family are pathogenic (e.g., Mycoplasma pneumoniae, Mycoplasma genitalium).
Viruses are not cells but consist of encapsulated genetic information in the form of DNA or RNA. They do not have their own metabolism and depend on other biological cells for their replication. Many representatives of this group are pathogenic [e.g., human immunodeficiency virus (HIV), hepatitis, and herpes].
There are two types of cells: the simple prokaryotic cells and the more complicated eukaryotic cells.
Chromosome and plasmids
Structure of Prokaryotic Cells Prokaryotic cells consist of a cytoplasm surrounded by a membrane, which is itself surrounded by a stabilizing cell wall. The most important functional units embedded in the cytoplasm are the ribosomes, the chromosome, and the plasmids (Figure 1.7).
Figure 1.7 Schematic of a prokaryotic cell.
Inclusion bodies and secretion
The chromosome is a single-stranded DNA double helix and contains the genetic information for the construction and replication of the cell. Protein biosynthesis from the DNA happens at the ribosomes after the DNA information has been transcribed into RNA. In addition to the chromosome, prokaryotes often carry further genetic information in the so-called plasmids. These are ring-shaped DNA molecules located in the cytoplasm. They usually encode secondary functional proteins such as the substances enabling penicillin resistance. The inclusion bodies shown in Figure 1.7 are storage locations for substances that for the time being are not required.
Gene expression
Inclusion bodies