Sunteți pe pagina 1din 46

Drug Manufacturing

BIT 230 Walsh Chapter 3

Drug Manufacturing

Most regulated of all manufacturing industries Highest safety and quality standards Parameters include:

Design and layout of facility Raw materials Process itself Personnel Regulatory framework

Pharmacopeias

Discussed before in other units and classes Martindale- not a standards book Gives information about drugs

Physiochemical properties Pharmacokinetics Uses and modes of administration Side effects Appropriate doses

GMP guidelines

Different publications world wide, but generally have similar information Go over everything from raw materials to the facility US guidelines issues publications called Points to Consider for additional guidelines for newer biotech products (will go over these later in semester)

Manufacturing facility

Most manufacturing facilities have requirements, but some specifics to biotech products, especially

Clean room Water

Clean Rooms

Clean room views Environmentally controlled areas Critical steps for bio/injectable drugs are produced in clean rooms Contain high efficiency particulate air (HEPA) filters in the ceiling Figure 3.1 page 98 of chapter

Classification of Clean Rooms for Pharma industry


Class

# microrganisms/m3 of air

A
B

<1
5

C
D

100
500

See table 3.5 page 100 of chapter

Other considerations

Exposed surfaces smooth, sealed, nonpenetrable surface Chemically-resistant floors and walls Fixtures (lights, chairs, etc.) minimum and easily cleaned Proper entry of materials and personnel into clean room to reduce risk of contamination in clean room

Gowned person in Clean room

Clean Room clothing


Covers most of operators body Change in a separate room and enter clean room via an air lock Clothing made from non-shredding material Number of people in a clean room at once limited to only necessary personnel (helps with automated processes)

CDS

Cleaning, decontamination and sanitization C- removal or organic and inorganic material that may accumulate D-inactivation and removal of undesired materials S- destroying and removing viable microorganisms

CDS contd

Done on surfaces that either are direct or indirect contact with the product

Examples of surfaces in both categories?

CDS of process equipment


Of course trickier because comes in contact with the final product Clean equipment, then rid equipment of cleaning solution Last step involves exhaustive rinsing of equipment with pure water

WFI Followed by autoclaving if possible If possible use CIP (cleaning in place)

Examples of CIP agents used to clean chromatography columns


0.5-2.0 M NaCl Non-ionic detergents 0.1-1.0 M NaOH Acetic Acid Ethanol EDTA Protease

Water

WFI- talked about this extensively before 30,000 liters of WFI needed for 1kg of a recombinant protein Use tap water just for non-critical tasks Purified water not as pure as WFI, but used for limited purposes (in cough medicines, etc.) WFI used exclusively in downstream processing Will not cover pages 105-112- water and documentation pages

Sources of Biopharmaceuticals

Genetic engineering of recombinant expression systems Your talks will be about types of systems and how they are used- mammalian cells, yeast, bacteria etc. Most approved products so far produced in E. coli or mammalian cell lines

E. coli

Cultured in large quantities Inexpensive (relatively speaking) Generation of quantities in a short time Production facilities easy to construct anywhere in the world Standard methods (fermentation) used

Current products from E. Coli


tPA (Ekokinase) Insulin Interferon Interleukin-2 Human growth hormone Tumor necrosis factor

Heterologous systems

Expression of recombinant proteins in cells where the proteins do not naturally occur Insulin first in E. coli Remember the drawbacks of expression in E. coli?

Other problems with E. coli


Most proteins in E. coli expressed intracellularly Therefore, recombinant proteins expressed in E. coli accumulate in the cytoplasm Requires extra primary processing steps (e.g. cellular homogenization) and more purification (chromatography)

Other problems with E. coli, contd

Inclusion bodies

Insoluble aggregates of partially folded product Heterologous expressed proteins overload the normal protein-folding machinery Advantage- inclusion bodies are very dense, so centrifugation can separate them from desired material

Preventing inclusion bodies

Lower growth temperature (from 37C to 30C) Use a fusion protein (thioredoxin) - native in E. coli protein expressed at high levels and remains soluble

Expression in animal cells


Major advantage- correct PT modifications Naturally glycosylated proteins produced in:


CHO - Chinese hamster ovary BHK - baby hamster kidney HEK human embryonic kidney

Current products from animal cells


tPA FSH Interferon - Erythropoietin FSH Factor VIIa

Disadvantages of animal cells (compared to E. coli)


Complex nutritional requirements Slower growth More susceptible to damage Increased costs

WILL NOT cover bottom of page 116 to page 124 (up to biopharmaceuticals)- you will cover these in your presentations

Final Product Production


Focus on E. coli and mammalian systems Process starts with a single aliquot of the Master Cell Bank Ends when final products is in labeled containers ready to be shipped to the customer

Production: Upstream and Downstream

Upstream: initial fermentation process; yields initial generation of product Downstream: purification of initial product and generation of finished product, followed by sealing of final containers biomanufacturing process overview

Upstream processing

Remove aliquot from MCB Inoculate sterile medium and grow (starter culture) Starter culture used to inoculate larger scale production culture Production culture inoculates bioreactor Bioreactors few to several thousand liters See figure 3.13 of chapter (page 129)

Upstream contd

Pages 129-133 go over specific details for microbial fermentation Pages 133-134 go over specific details for animal cell culture Properties of animal cells Anchorage dependent Grow as a monolayer Contact inhibited Finite lifespan Longer doubling times Complex media requirements

Downstream processing

Diagram page 135 of chapter 3

Detailed steps considered confidential


Clean room conditions for downstream

Downstream contd

Steps involved (intracellular products E. coli.) mammalian products secreted in media, so easier to isolate)

Centrifugation or filtration Homogenization Removal of cellular debris Concentration of crude material (by precipitation or ultra filtration) High resolution chromatography (HPLC) Formulation into the final product

Downstream contd

Final product formulation


Chromatography yields 98-99% pure product Add excipients (non active ingredients), which may stabilize the final product Filtration of final product, to generate sterile product Freeze drying (lyophilization) if product if to be sold as a powder (dictated by product stability)

Separation methods

Page 142,tables 3.18 and 3.19 Familiar with:


Ion-exchange Gel-filtration Affinity chromatography

Protein A chromatography Immunoaffinity chromatography

Factors that influence biological activity


Denature or modify proteins Results in loss of/reduced protein activity Need to minimize loss in downstream work Problems can be chemical (e.g., oxidizing, detergents); physical (e.g., pH, temperature); or biological (e.g., proteolytic degradation) Table 3.20 page 143

Proteolytic degradation

Hydrolysis of one or more peptide bonds Results in loss of biological activity Trace quantities of proteolytic enzymes or chemical influences Several classes of proteases:

Serine Cysteine Aspartic Metalloproteases (also in other ppt)

Protease inhibitors

PMSF serine and cysteine proteases Benzamidine serine proteases Pepstatin A aspartic proteases EDTA metalloproteases a.a residue known to be present at active site of protein, so disruption of it causes loss of activity

Others (mentioned before)

Deamidation hydrolysis of side chain of asparagine and glutamine

Happens at high temp and extreme pH Oxidation by air (met and cys in particular)

Oxidation and disulphide exchange

Alterations of glycosylation patterns in glycoproteins (more than one sugar)

Affect activity or immunological properties

Excipients

Substances added to final product to stabilize it Serum albumin


Withstands low pH or elevated temps Keeps final product from sticking to walls of container Stabilize native conformation of protein

Excipients contd

Amino acids

Glycine stabilizes interferon, factor VIII, stabilizes against heat

Alcohols (and other polyols)

Stabilize proteins in solution

Surfactants

Reduces surface tension; proteins dont aggregate, so dont denature

Final product fill


See figure 3.27 page 153 Bulk product gets QC testing Passage through 0.22 m filter for final sterility Aceptically filled into final product containers Uses automated liquid handling systems

Final product fill contd


Freeze drying (lyophilization) Yields a powdered product Reduces chemical and biological degradation of final product Longer shelf life than products in solution Storage for parenteral products (those administered intravenously or injected)

Freeze drying contd

Need to add cryoprotectors


Glucose or sucrose Serum albumin Amino acids Polyols

Freeze drying can be done in many steps

Labeling and Packing


After sealed in final container, product quarantined Samples are QCd Check potency, sterility and final volume Detection and quantitation of excipients Highly automated procedures Labeling function critical- biggest error where many products are made

Label

Name and strength of product Specific batch number Date of manufacture and expiry date Required storage conditions Name of manufacturer Excipients included Correct mode of usage

Other final product items


Biopharmaceutical products undergo more testing than traditional pharma products Products made in recombinant systems have more potential to be contaminated than synthetic chemical drugs Larger, more complex molecules

S-ar putea să vă placă și