SlideShare a Scribd company logo
Pyrogens, Endotoxin and the LAL test: An Introduction in Relation to 
Pharmaceutical Processing 
By Dr. Tim Sandle 
(email: timsandle@btinternet.com; website http://www.pharmig.blogspot.com) 
Introduction 
Pharmaceutical products which are injected into the human body are tested for 
pyrogenic substances. The most common, and arguably most important test, is for 
bacterial endotoxin. The pathological effects of endotoxin, when injected, are a rapid 
increase in core body temperature followed by extremely rapid and severe shock, 
often followed by death before the cause is even diagnosed. However, there needs to 
be large quantities of endotoxin within the human body for this to happen, and the 
endotoxin needs to be injected into the blood stream. 
Bacterial endotoxin is the lipopolysaccharide (LPS) component of the cell wall of 
Gram-negative bacteria. It is pyrogenic, and it is a risk to patients who are 
administered intravenous and intramuscular preparations. The purpose of the LAL test 
is to detect endotoxin (Guy, 2003). 
Pyrogenicity 
Bacterial endotoxin can be classed, among other things, as a “pyrogen.” Pyrogens are 
substances which, when injected into the mammalian body, will cause a variety of 
symptoms, the most recognizable of which is an increase in core body temperature. In 
the early days of the pharmacopeia, drug substances were classed as apyrogenic or 
pyrogenic, based, from 1942 and until the 1980’s, on the “pyrogen test” (whereby a 
quantity of the drug was injected into three rabbits, and the temperature response of 
the rabbits was noted). The rabbit pyrogen test was first described by Florence Seibert 
in 1925. 
The rabbit test is no longer widely used, and it has largely been replaced, for the 
testing of parenteral drug products, by the LAL (Limulus amoebocyte lysate) test. The 
LAL test is a method of the Bacterial Endotoxin Test (BET), for detecting the 
presence, and to go some way to determining the level of Gram-negative bacterial 
endotoxin in a given sample or substance. Current editions of the pharmacopoeia 
carry statements to the effect that where the term apyrogenic or pyrogen-free is used, 
it should be interpreted as meaning that samples of the product will comply with a 
limit for bacterial endotoxin. 
GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
The association of fevers (pyrexia) has a long history. However, it was not until the 
early 20th century with the development of a rabbit pyrogen test that an understanding 
began to emerge in which bacteria could be classified into pyrogenic and non-pyrogenic 
types that correlate to their Gram-stain. Gram-negative bacteria were 
found to be pyrogenic, Gram-positive bacteria were generally not, and killed cultures 
of Gram-negative bacteria were comparable to live cultures in their ability to induce 
fevers. 
By the 1920’s it was apparent that sterility in parenteral pharmaceuticals could be no 
guarantee of nonpyrogenicity, and that if pyrogenicity was to be avoided, it was 
imperative to avoid bacterial contamination at every stage of manufacture of 
parenteral pharmaceuticals. The first official rabbit pyrogen test was included in 
United States Pharmacopeia (USP) in 1942. 
In recognition that the causative agent of pyrogenicity was filterable and heat stable, 
efforts were applied to identifying its chemical composition. Trichloracetic acid and 
phenol-water extractions of bacteria were found to be effective in isolating the 
pyrogenic element from bacteria. These extracts were chemically identifiable as 
lipopolysaccharide (or what is commonly described as bacterial endotoxin). 
Bacterial Endotoxin 
The structural rigidity of the bacterial cell wall is conferred by a material called 
peptidoglycan (also known as murein). It is a polymer consisting of sugars and amino 
acids that form a mesh-like layer outside the plasma membrane of bacteria, forming 
the cell wall (Madigan et al, 2009). 
In Gram-positive bacteria, peptidoglycan is present as a thick layer, which is 
outermost in the cell wall. In Gram-negative bacteria, the peptidoglcan is only a thin 
layer, and it is not the outermost layer. Gram-negative bacteria are sometimes 
described as having a cell envelope rather than a cell wall. The term envelope better 
describes the loosely attached layer of material called lipopolysaccharide, which is 
located outside a thin structural layer of peptidoglycan, as shown in Figure 1 below: 
GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
Figure 1: Diagram of the Outer Bacterial Cell Wall (from Creative Commons 
Library) 
The outer layer of this lipopolysaccharide envelope is a permeability barrier effective 
against diffusion of exoenzymes into the external environment. This is an 
evolutionary feature which has arisen to allow Gram-negative bacteria (illustrated in 
Figure 2) to survive and increase in numbers in environments such as water, in which 
there are only low concentrations of organic nutrients. Macromolecular organic 
nutrients are trapped in the cell envelope as the water flows by, and then within the 
cell envelope they are hydrolyzed to smaller molecules, which can be taken into the 
cell. 
The important evolutionary advantages conferred by lipopolysaccharide are: 
 It contributes to the adhesion of Gram-negative bacteria to surfaces, allowing 
them to form as biofilms in aqueous environments. 
 It “attracts” and “entraps” organic macromolecules from aqueous 
environments. 
 It allows for entrapped organic macromolecules to be “recognized” by the cell 
so that specific enzymes can be synthesized to break them down into smaller 
fragments capable of passing through the peptidoglycan layer into the cell. 
 LPS also increases the negative charge of the cell membrane and helps 
stabilize the overall membrane structure. 
 It retains the enzymes synthesized by the cell so that they are not lost into the 
external environment [(Raetz and Whitfield,(2002)]. 
GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
Figure 2: Gram's Stain Light Microscope Image, Showing Rod-Shaped Gram- 
Negative Bacteria (image from Creative Commons Library). 
Lipopolysacccharide is pyrogenic. Bacterial endotoxin is a synonym for 
lipopolysaccharide. 
Although intimately associated with the cell envelope of Gram-negative bacteria, 
lipopolysaccharide is constantly shed by the bacteria into the environment, much like 
the shedding of the outer layers of human skin. When Gram-negative bacteria die and 
lyse, all of their lipopolysaccharide is shed into the environment. 
Furthermore, when bacterial cells are lysed by the immune system, fragments of 
membrane containing lipid A are released into the circulation, causing fever, diarrhea, 
and possible fatal endotoxic shock (also called septic shock) (Brandberg, K. et al, 
1996). There are some other substances which are also pyrogenic. They are unusual 
and are extremely rarely found associated with pharmaceutical preparations. 
Lipopolysaccharide has three distinct chemical regions, as illustrated in Figure 3 
below: 
GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
Figure 3: Diagram of LPS (from Creative Commons Library) 
 An inner core called Lipid A 
 An intermediate polysaccharide layer 
 An outer polysaccharide side chain 
Lipid A, embedded in the bacterial outer membrane, is responsible for pyrogenicity. 
The LAL Test 
The principle of the LAL test is a reaction between lipopolysaccharide and a 
substance (“clottable protein”) contained within amoebocyte cells derived from the 
blood of the Horseshoe Crab (of which Limulus polyphemus is the most commonly 
used species, as illustrated in Figure 4 below). LAL is an aqueous extract obtained 
after lysis of blood cells (amoebocytes).The reaction is specific. The reaction of the 
horseshoe crab to endotoxin (the formation of a clot) has been known since the 1950’s 
(Bang, 1956). 
GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
Figure 4: Image of the Limulus “Horseshoe crab” (image from Creative 
Commons Library) 
When endotoxin comes into contact with LAL, it initiates a series of enzymatic 
reactions that result in the activation of a pathway to the production of at least three 
serine protease zymogens (Factor C, Factor B and a proclotting enzyme). This 
pathway alters amoebocyte coagulogen (an invertebrate fibrinogen-like clottable 
protein) to form coagulin gel. 
Serine proteases are enzymes that cleave peptide bonds in proteins, in which serine 
serves as the nucleophilic amino acid at the active site. They are found in humans as 
well as in the horseshoe crab (and indeed in all mammals). In humans, they are 
responsible for coordinating various physiological functions, including digestion, 
immune response, blood coagulation and reproduction. It is the blood coagulation 
reaction which is similar in both humans and the horseshoe crab (Cooper, 2004). 
The reference test in the pharmacopoeias is called the gel clot (or gelation) and is 
conducted on the endpoint principle. The description of the test and the necessary 
GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
validation and accompanying controls is so detailed in both USP and Ph.Eur. 
(harmonized since 1999). 
The clotting mechanism of the blood of the crab is designed to prevent the spread of 
bacterial contamination throughout the horseshoe crab’s biochemical system. When 
the endotoxin of Gram-negative bacteria contacts with the horseshoe crab’s 
amebocytes, a series of enzymatic reactions begin. The pathway alters amebocyte 
coagulogen into a fibrinogen-like clottable protein, which forms a coagulin gel. The 
defence mechanism is also effective against fungi, hence, a similar reaction occurs in 
response to a fungal infection, which triggers the clotting cascade. In the reactions, 
glucans trigger the protease enzyme Factor G, whereas endotoxin triggers the Factor 
C enzyme, although the end result – coagulin – is the same (Moser, 2009). 
A considerable amount more glucan (1,000 times) is required to trigger the clotting 
cascade than the equivalent amount of endotoxin. The glucan required to trigger 
Factor G can be of varying molecular weights (such weights range from 3kD to 
100kD). (Tanaka et al, 1993). This is illustrated below in Figure 5: 
E n d o to x in 
F a c to r C A c tiv e F a c to r C ß -(1 ,3 )-D -G lu c a n 
F a c to r B A c tiv e F a c to r B ------------------------A c tiv e F a c to r G F a c to r G 
P r o c lo ttin g E n z ym e C lo ttin g E n z ym e 
C o a g u lo g en C o a g u lin 
G e l F o rm a tio n 
Figure 5: LAL Clotting Cascade (adapted by the author) 
The LAL reagent is used for the gel-clot supplied with an identified sensitivity or 
label claim (lamda or λ), e.g., 0.03 endotoxin units (EU) per mL. This means that 
when mixed with an equal volume of the material under test, a gel or clot will form if 
the material contains 0.03 EU per mL or greater. For the kinetic methods, the lysate 
does not come with a label claim. The test sensitivity is determined by the lowest 
point of the standard curve used with each assay (Sandle, 2001). 
GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
When it is necessary to quantify endotoxin concentration in a material, it is usual to 
test a series of doubling dilutions against the reagent in temperature-controlled 
conditions. The greatest dilution that gives a positive result (formation of a gel or 
clot) is the end point, and the concentration of endotoxin in the original material can 
be calculated by multiplying the dilution factor at the end point by the sensitivity of 
the LAL reagent. 
LAL tests require validation for each technician and each product before being 
applied routinely, and even then valid assays require careful internal standardization. 
These requirements are prescribed in detail in the pharmacopoeia to the extent that 
they do not merit repeating here. They require the use of a standard endotoxin termed 
“positive product control” (PPC), which is a known amount of endotoxin mixed with 
a test material to confirm the absence of interference. 
In the early days of the LAL test, endotoxin standards were variable, potency varied 
with the method of purification, the bacteria of its origin, and how it was formulated. 
In the 1970’s, USP and FDA commissioned Rudbach (Rudbach et al, 1976) to resolve 
the issue by preparing a reference endotoxin which was stable, could be chemically 
characterized, could be lyophilized without loss of activity, and which was free from 
biologically active proteins. This is termed RSE (reference standard endotoxin). 
Since RSE is expensive and potentially exhaustible, certified or calibrated standard 
endotoxin (CSE) is normally used for routine work, but not necessarily for 
fundamental research. The potency of a given batch of CSE is determined relative to 
the current Ph.Eur., USP or FDA lot of reference standard endotoxin (RSE) and is 
specific to particular batches of LAL reagent. Although the LAL test today is more 
robust, it remains open to a degree of variation (McCullogh, K.C. and Weider- 
Loeven, C., 1992). 
Today, both RSE and CSE are manufactured from Escherichia coli bacteria. With 
respect to international standards, the World Health Organization reference standard 
remains the chief standard from which standards approved by the pharmacopoeia are 
derived and equivalent to one another. 
The units of measurement for the LAL test are Endotoxin Units (EU). These are a 
measure of the activity of the endotoxin. Endotoxins differ in their biological activity 
or potency; the pyrogenicity or LAL reactivity of one endotoxin preparation may be 
very different from that of another of the same weight. Conversely, two endotoxin 
molecules may be different sizes and different weights but may have the same 
reactivity in an LAL test. The potency of an endotoxin determined with one LAL 
reagent lot may differ from that determined with another lot. Expressing endotoxin 
concentrations in EU’s avoids the issues of different potencies of different endotoxins 
and allows us to compare the results of different LAL tests performed in different 
GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
labs. Consequently, it is not the usual practice to convert the results of an LAL test in 
EU/ml to units of weight of endotoxin per ml. 
The primary LAL test methods are the gel-clot, chromogenic and turbidimetric. The 
former is an end-point method whilst the latter two are kinetic methods. The kinetic 
methods are more sensitive than the gel clot method, because changes in turbidity and 
color are discernable by light-scattering devices at lower concentrations of endotoxin 
than those at which gels form. 
When using kinetic methods, the most important aspect is the standard curve for the 
endotoxin concentrations used in a standard curve to determine the test sensitivity. 
Therefore, the high point and low point in a standard curve determines the lower and 
upper level of endotoxin that can be detected. 
Rapid LAL tests are now commonplace on the market, and the industry is also 
moving towards the use of recombinant lysate, and the reliance upon the horseshoe 
crab may one day decrease. 
The LAL assay may be interfered with by the sample being tested. The interference 
may be caused by a number of different factors, such as pH, protein concentration, 
presence of chemicals (such as NaOH from rinsing cycles, etc). Interference may 
affect the lysate or the endotoxin. Inhibition or enhancement is normally detected 
through the use of spiked controls. Inhibition is arguably the greatest concern, because 
it can result in a failure to detect the true level of endotoxin in a sample. 
When conducting LAL tests, the dilution of samples is important. This is in order to 
minimize the effects of any component of the material being tested, which may be 
inhibitory to the LAL reaction. It is, however, imperative that the material being 
tested is not diluted too much, because this would ensure negative results, possibly 
false negatives. The way this is avoided is by having a Maximum Valid Dilution 
(MVD). 
LAL tests are only valid in situations where standard endotoxin can be shown to be 
detectable with the same efficiency in a test sample as in a control consisting of water 
known to be endotoxin free (this is termed LRW, or LAL reagent water). 
Sources of Endotoxin in Pharmaceutical Manufacturing 
A. Water 
Although all bacteria have some associated endotoxin, the most potent source of 
endotoxin is from Gram-negative bacteria. Endotoxins may be shed from viable 
bacteria during growth, or they may be associated with nonviable bacteria. 
GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
Endotoxicity is not a function of microbial viability. The most common habitat for 
Gram-negative bacteria is water, where they have evolved to be able to survive and 
increase in numbers with minimal nutritional support and can adapt their metabolism 
to metabolize complex organic macromolecules that other bacteria cannot. 
In any form of pharmaceutical manufacture, water is one of the most serious potential 
sources of microbiological contamination. Water cannot be totally excluded from 
sterile products manufacturing facilities. The primary focus on endotoxin control in 
pharmaceutical manufacturing is on controlling it at its source – water. If endotoxin 
is not controlled at its source, it has the potential to create difficulties through 
manufacture to the finished product, potentially leaving no recourse but rejection. 
Endotoxin is practically impossible to remove terminally from pharmaceutical dosage 
forms. 
The measures taken to control contamination from water in sterile manufacturing 
facilities can be seen as measures taken to minimize the risk of endotoxin 
contamination of products. Pivotal to the control chain for any source of 
microbiological contamination is identification of those purposes for which it is 
necessary; in the case of water these are: 
 “Ingredient” water for aqueous sterile products 
 Water supplied for the cleaning of equipment and components 
 Water supplied to laundries 
 Water supplied for hand washing 
 Steam supplies to autoclaves, SIP systems, etc. 
The pharmacopoeia deal with ingredient water under two monographs, Purified Water 
and Water for Injection (WFI). To comply with the FDA requirements, WFI may be 
prepared by distillation or by reverse osmosis. For Europe, WFI can only currently be 
produced from distillation. With distillation, lipopolysaccharide has a molecular 
weight of around 106, heavy enough to be left behind when water is rapidly boiled off, 
as in a still. 
Where water has an endotoxin limit, it does not mean that it will automatically 
comply with that limit. Suitably controlled means of preparation, storage and 
distribution must be employed to ensure that the limits are complied with at point of 
use. Systems and user points require monitoring. 
WFI as prepared by the pharmacopoeially approved processes of distillation will 
comply (if sampled directly from the preparation point and not contaminated in 
sampling or testing). However, it is immediately fed to a storage tank and pumped 
GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
round a distribution system where at least in theory, microbiological contamination 
may be lurking and shedding endotoxin. 
Figure 6: Laboratory Work in a Pharmaceutical Laboratory (image copyright 
Tim Sandle) 
When used in bulk for manufacturing purposes, the pharmacopoeia also apply a 
microbiological limit to WFI; this is not more than 10 cfu per 100 mL. This 
microbiological limit for WFI does not tie in with the endotoxin specification. The 
amount of endotoxin associated with Gram-negative bacteria is thought to be around 
10-15g per bacterium. The first batch of Reference Standard Endotoxin (RSE) 
titrated at 1 EU = 2 x 10-10g. Therefore the endotoxin limit of 0.25 EU/ml for Water 
for Injections can be understood to correspond to 5 x 10-11g/ml, or about 104 bacteria 
/ml. Despite the fact that there is a great deal of uncertainty about these figures, and 
that Gram-negative bacteria not only have associated endotoxin but also shed 
endotoxin continuously, and that Gram-positive bacteria are generally not very 
endotoxic, it is apparent that water which meets the limit for endotoxin may exceed 
the limit for viable microorganisms (10 cfu/100ml). 
GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
The means of controlling the development of Gram-negative (and other) 
microorganisms in water storage and distribution systems are: 
 Smooth internal surfaces in tanks and in pipework. Microorganisms adhere 
less well to smooth surfaces than to rough surfaces. Bear in mind that the 
degree of roughness and smoothness is a “micro” phenomenon; suitable and 
unsuitable internal finishes would not be discernable to the average triple 
blade shaver. Pipe joints can disrupt smoothness, as can welds. 
 Continuous movement of the water in tanks and rapid flow in pipework. 
Where shear forces are involved, microorganisms adhere poorly to surfaces. 
Where there is no movement of the water, there is no shear, and shear 
increases with speed of flow. 
 Avoidance of areas where water can remain stagnant. 
 These include “dead legs” – water may stagnate in branch pipes from a 
circulating main if the length of the branch is too long to allow the turbulence 
of the flowing main to disturb the contents of the branch pipe. FDA has 
defined “dead legs” according to their dimensions in the 1993 Guide to 
Inspection of High Purity Water Systems, but fundamentally the principle is to 
always minimize the length of branch pipes. 
 Water can also remain stagnant in valves, particularly at user points, and even 
more particularly at user points which are not in frequent and regular use. 
This is counteracted by use of so-called hygienic or “zero dead leg” valves, 
which although significantly better than the alternatives (say, ball valves), 
should not lead to a sense of false security; they can harbor endotoxin-shedding 
biofilms. 
 Ring mains should be sloped (have “drop”) from the point of origin to the 
point of return to ensure that systems are completely drainable. 
 Avoidance of leakage. Water leaks can cause bridging of water to the external 
environment through which bacteria may enter the system. Storage tanks 
should be equipped with filter on their air vents to prevent airborne 
microbiological ingress. They may even be held under a “blanket” of an inert 
gas, such as nitrogen. 
 High temperature storage and distribution. The risks of endotoxin-shedding 
biofilms, despite the best attempts at control above, are thought to be so 
consequential that the regulatory bodies require the temperature of storage and 
GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
distribution to be maintained higher than 75°C. It should, however, be considered 
that 75°C is too high a temperature for most pharmaceutical formulation purposes. 
This means that user points are generally equipped with some form of cooling – 
heat exchangers used for this purpose may be a source of endotoxin and bacterial 
contamination and may thus cancel out many of the benefits of high temperature 
circulation. 
B. Depyrogenation 
Endotoxicity is not necessarily lost with the loss of viability of microorganisms. 
Lipopolysaccharide is not destroyed to any significant extent by sterilization 
treatments, such as steam sterilization, gamma radiation, ethylene oxide, hydrogen 
peroxide, etc. Lipopolysaccharide also passes through 0.22 μm bacteria retentive 
filters. It is claimed that endotoxin may be removed from liquids by up to 4 log10 
reductions using 0.025 μm ultrafilters (which function as a molecular sieving 
process). 
There are two commonly used ways of eliminating endotoxin from materials:either by 
removing them or by inactivating them. The two primary means by which elimination 
is achieved is either by rinsing or by dry heat depyrogenation (Sandle, 2004). 
The normal method of removal is by rinsing the material with WFI. This is normally 
applied to rubber stoppers for vials. It is also what is done to vessels and major 
pieces of equipment used for sterile parenteral manufacture. A question arises as to 
whether this can be validated and assured? In answer, the sampling statistics are 
likely to be poor, and the test method is inaccurate, and probably there is not much 
there in the first place. This is an area where the regulators have looked at but there is 
no consistent standard, although it is implicit that a 3log10 reduction should be 
achieved by washing just as it should be by heat inactivation. From this, satisfactory 
validation of washing processes is not easy – or conversely, it can be quite easy, 
depending on the rigour of the validation design and the minutiae of process 
knowledge, for instance: 
 Where should the endotoxin “spike” be placed on the material or item 
being washed? The sensible location on vial stoppers must be that part of 
the stopper that potentially could come into contact with the product. 
 How much endotoxin should be “spiked” on items or materials? If too 
much endotoxin is added, for example, 1,000,000 EU/component, it could 
be too easily removed and may reveal little about the washing method 
used. Alternatively, too little may not even allow the demonstration of a 3 
log10 reduction. 
 
GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
 How effective are washing processes? Air bubbles can cling to the 
surfaces of rubber stoppers and create areas that do not interact properly 
with the washing process. 
 How effective are washing machines? Batch washing machines generally 
work on a siphonage principle, such that removed materials (including 
endotoxin) are flushed upwards out of the washer rather than allowing the 
removed materials to settle back on the washed items. Continuous (semi-continuous 
washers (usually used for glassware and linked to 
depyrogenating tunnels) hold items upside down throughout the washing 
process; the final water rinse is frequently recycled to be the first wash, 
and probably adds less endotoxin than a lower grade of water would. 
Inactivation is done by dry heat. If there are materials and glass vials which are 
required for sterile parenteral manufacture which can be depyrogenated by dry heat, 
they should be depyrogenated by dry heat in an oven or a tunnel. 
The regulatory standard for validation of an endotoxin inactivation (depyrogenation) 
process is that it should be capable of reducing an endotoxin challenge through 3 log10 
reduction. To ensure that this limit works, there is also a requirement to clean 
materials prior to dry heat depyrogenation with WFI – otherwise, at least in theory, an 
item could be contaminated with 10,000 EU prior to entering a validated endotoxin 
inactivation process and still emerge with 10 EU intact and be ready to contaminate 
the product. 
Dry heat depyrogenation is a complex process which is still poorly understood, with 
contradictory research data. The phenomenon which complicates the picture is that 
inactivation may approximate to Second Order1 chemical kinetics with a high initial 
rate of inactivation, then tail off to nothing. What this means in practice is that at any 
particular depyrogenating temperature will be subjected to some degree of 
inactivation in some period of time or other, but beyond that point, no further 
inactivation will occur by holding the material at that temperature. 
D-values for pure endotoxin (the time required to achieve one log10 reduction in 
activity) at 170°C are as high as 20 minutes, which would suggest that 3 log10 
reductions are achievable in 60 minutes. However, due to the complexity of the 
kinetics, this is not necessarily true. All work with validation of depyrogenating 
processes must be done empirically with endotoxin challenges. This in itself is not 
easy, as the amount of endotoxin dried on an item (depending on the material of the 
item) may not be the maximum amount which can be recovered. Furthermore, the 
amount that can be recovered after treatment may only be a fraction of the amount 
1For a second-order reaction, the rate of reaction is directly proportional to the square of the 
concentration of one of the reactants. 
GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
that remains. Validation of endotoxin inactivation is a complex experimental area and 
requires that a lot of knowledge and a lot of well-considered controls to be included. 
Oven temperatures above 180°C will inactivate endotoxin through 3 log10. USP 
recommends 250°C, and peak temperatures approaching 350°C are achieved in 
tunnels. Based on these data, it is worth noting that each valid depyrogenating 
process is also an overkill sterilizing process. 
Conclusion 
This paper has provided an introduction to endotoxin and to the primary method for 
detecting endotoxin: the LAL test. The paper has also examined the risks of endotoxin 
to pharmaceutical processing and some of the control measures in place to reduce the 
risk of endotoxin contamination. The aim of the paper was to provide an introduction 
to this important subject. 
References 
Bang, F.B. (1956): “A Bacterial Disease of Limulus Polyphemus,” Bull. Johns 
Hopkins Hosp. 98, p.325. 
Brandberg, K. et al (1996): “Conformation of Lipid A, the Endotoxic Center of 
Bacterial Lipopolysaccharide,”J. of Endotoxin Research, Vol.3, No.3: 173-178. 
Cooper, J.F. (2004):“Microbiological Contamination Control in Parenteral 
Manufacturing,” In Williams, K. (Ed.) Microbial Contamination Control in 
Parenteral Manufacturing New York: Marcel Dekker, pp. 531-540. 
Guy, D. (2003): “Endotoxins and Depyrogenation,” in Hodges, N. and Hanlon, G., 
“Industrial Pharmaceutical Microbiology: Standards and Controls,”Euromed, pp. 
12.1–12.15. 
Madigan, M. T., Martinko, J. M., Dunlap, P. V., and Clark, D. P. (2009): Brock 
Biology of Microorganisms. 12th ed., San Francisco, CA: Pearson/Benjamin 
Cummings, p.742. 
McCullogh, K.C. and Weider-Loeven, C. (1992): “Variability in the LAL Test: 
Comparison of Three Kinetic Methods for the Testing of Pharmaceutical Products,”J. 
of Parenteral Science and Technology, Vol. 46, 3: 69-72. 
Raetz, C.R.H. and Whitfield, C. (2002):“Lipopolysacchride Endotoxin,”Annual 
Review of Biochemistry, 71, pp. 635-700. 
GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
Moser, K. (2009): “Playing Hide and Seek with Endotoxin,”LAL User Group 
Newsletter, Issue 3, Number 2, pp.1-5. 
Rudbach, J.A., Akiya, F.I., Elin, R.J., Hochstein, H.D., Luoma, M.K., Milner, C.B., 
Milner, K.C. and Thomas, K.R. (1979): "Preparation and Properties of a National 
Reference Endotoxin," J. of Clin.Microbiol., 3(10): pp. 21-25. 
Seibert, F.B. (1925): “The Cause of Many Febrile Reactions Following Intravenous 
Injection,”Am. J. Physiol., 71: pp. 621-651. 
Sandle, T. (April 2001): “Performance Characteristics of Automated LAL 
Tests,”PharMIG News No. 4, pp. 3-10. 
Sandle, T. (June/July 2004): “Three Aspects of LAL Testing: Glucans, 
Depyrogenation and Water System Qualification,”PharMIG News No. 16., pp.3-12. 
Tanaka, S., Aketagawa, J., Takahashi, S., Shibata, Y., Tsumuraya, Y., and Hashimoto, 
Y. (1993): “Inhibition of High-Molecular-Weight-(1β3)-β-D-Glucan-Dependent 
Activation of Limulus Coagulation Factor G by Laminaran Oligosaccharides and 
Curdlan Degradation Products,” Carbohydrate Research, 244, pp. 115-127. 
GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor

More Related Content

What's hot

PYROGEN TESTING
PYROGEN TESTINGPYROGEN TESTING
Expt. 9 Test for pyrogens ( rabbit method)
Expt. 9 Test for pyrogens ( rabbit method)Expt. 9 Test for pyrogens ( rabbit method)
Expt. 9 Test for pyrogens ( rabbit method)
VISHALJADHAV100
 
Pyrogens test
Pyrogens testPyrogens test
Pyrogens test
Ahmad Ali
 
BET testing USP85 an Introduction
BET testing USP85 an Introduction BET testing USP85 an Introduction
BET testing USP85 an Introduction
Amrutha Raiker
 
Pyrogen testing 112070804005
Pyrogen testing  112070804005Pyrogen testing  112070804005
Pyrogen testing 112070804005Patel Parth
 
Immunohisto Chemistry Antigen retrieval
Immunohisto Chemistry Antigen retrievalImmunohisto Chemistry Antigen retrieval
Immunohisto Chemistry Antigen retrieval
Atifa Ambreen
 
Clinical isolates of urinary tract infection and candidiasis inhibited by T. ...
Clinical isolates of urinary tract infection and candidiasis inhibited by T. ...Clinical isolates of urinary tract infection and candidiasis inhibited by T. ...
Clinical isolates of urinary tract infection and candidiasis inhibited by T. ...
Diganta Dey
 
Lal presentation
Lal presentation Lal presentation
Lal presentation
Tim Sandle, Ph.D.
 
Antigen retrieval (ANATOMY)
Antigen retrieval (ANATOMY)Antigen retrieval (ANATOMY)
Antigen retrieval (ANATOMY)
sartika Amran
 
Antigen retrieval
Antigen retrievalAntigen retrieval
Antigen retrieval
Atifa Ambreen
 
Immunohistochemistry
ImmunohistochemistryImmunohistochemistry
Immunohistochemistry
NuhaJuma
 
principle of Immunohistochemistry and its use in diagnostics
principle of Immunohistochemistry and its use in diagnosticsprinciple of Immunohistochemistry and its use in diagnostics
principle of Immunohistochemistry and its use in diagnostics
Ekta Jajodia
 
Antibody purification
Antibody purificationAntibody purification
Antibody purification
Sulov Saha
 
Usage of Immunological reactions in diagnostics of infectious diseases.
Usage of Immunological reactions in diagnostics of infectious diseases. Usage of Immunological reactions in diagnostics of infectious diseases.
Usage of Immunological reactions in diagnostics of infectious diseases.
Eneutron
 
ELISA: Enzyme-linked Immunosorbent Assay
ELISA: Enzyme-linked Immunosorbent AssayELISA: Enzyme-linked Immunosorbent Assay
ELISA: Enzyme-linked Immunosorbent Assay
Syed Muhammad Khan
 
Phytochemical screening and in vitro antioxidant activity of extracts of jasm...
Phytochemical screening and in vitro antioxidant activity of extracts of jasm...Phytochemical screening and in vitro antioxidant activity of extracts of jasm...
Phytochemical screening and in vitro antioxidant activity of extracts of jasm...
SriramNagarajan16
 
Elisa ppt
Elisa pptElisa ppt
Elisa ppt
Saba Ahmed
 
Usage of immunological reactions in diagnostics of infectious diseases. Compl...
Usage of immunological reactions in diagnostics of infectious diseases. Compl...Usage of immunological reactions in diagnostics of infectious diseases. Compl...
Usage of immunological reactions in diagnostics of infectious diseases. Compl...
Eneutron
 
Effects of silver nanoparticle (Ag NP) on oxidative stress biomarkers in rat
Effects of silver nanoparticle (Ag NP) on oxidative stress biomarkers in ratEffects of silver nanoparticle (Ag NP) on oxidative stress biomarkers in rat
Effects of silver nanoparticle (Ag NP) on oxidative stress biomarkers in rat
Nanomedicine Journal (NMJ)
 
Epidemiology and Characterization of lipophilic yeast (Malassezia) from human...
Epidemiology and Characterization of lipophilic yeast (Malassezia) from human...Epidemiology and Characterization of lipophilic yeast (Malassezia) from human...
Epidemiology and Characterization of lipophilic yeast (Malassezia) from human...
Premier Publishers
 

What's hot (20)

PYROGEN TESTING
PYROGEN TESTINGPYROGEN TESTING
PYROGEN TESTING
 
Expt. 9 Test for pyrogens ( rabbit method)
Expt. 9 Test for pyrogens ( rabbit method)Expt. 9 Test for pyrogens ( rabbit method)
Expt. 9 Test for pyrogens ( rabbit method)
 
Pyrogens test
Pyrogens testPyrogens test
Pyrogens test
 
BET testing USP85 an Introduction
BET testing USP85 an Introduction BET testing USP85 an Introduction
BET testing USP85 an Introduction
 
Pyrogen testing 112070804005
Pyrogen testing  112070804005Pyrogen testing  112070804005
Pyrogen testing 112070804005
 
Immunohisto Chemistry Antigen retrieval
Immunohisto Chemistry Antigen retrievalImmunohisto Chemistry Antigen retrieval
Immunohisto Chemistry Antigen retrieval
 
Clinical isolates of urinary tract infection and candidiasis inhibited by T. ...
Clinical isolates of urinary tract infection and candidiasis inhibited by T. ...Clinical isolates of urinary tract infection and candidiasis inhibited by T. ...
Clinical isolates of urinary tract infection and candidiasis inhibited by T. ...
 
Lal presentation
Lal presentation Lal presentation
Lal presentation
 
Antigen retrieval (ANATOMY)
Antigen retrieval (ANATOMY)Antigen retrieval (ANATOMY)
Antigen retrieval (ANATOMY)
 
Antigen retrieval
Antigen retrievalAntigen retrieval
Antigen retrieval
 
Immunohistochemistry
ImmunohistochemistryImmunohistochemistry
Immunohistochemistry
 
principle of Immunohistochemistry and its use in diagnostics
principle of Immunohistochemistry and its use in diagnosticsprinciple of Immunohistochemistry and its use in diagnostics
principle of Immunohistochemistry and its use in diagnostics
 
Antibody purification
Antibody purificationAntibody purification
Antibody purification
 
Usage of Immunological reactions in diagnostics of infectious diseases.
Usage of Immunological reactions in diagnostics of infectious diseases. Usage of Immunological reactions in diagnostics of infectious diseases.
Usage of Immunological reactions in diagnostics of infectious diseases.
 
ELISA: Enzyme-linked Immunosorbent Assay
ELISA: Enzyme-linked Immunosorbent AssayELISA: Enzyme-linked Immunosorbent Assay
ELISA: Enzyme-linked Immunosorbent Assay
 
Phytochemical screening and in vitro antioxidant activity of extracts of jasm...
Phytochemical screening and in vitro antioxidant activity of extracts of jasm...Phytochemical screening and in vitro antioxidant activity of extracts of jasm...
Phytochemical screening and in vitro antioxidant activity of extracts of jasm...
 
Elisa ppt
Elisa pptElisa ppt
Elisa ppt
 
Usage of immunological reactions in diagnostics of infectious diseases. Compl...
Usage of immunological reactions in diagnostics of infectious diseases. Compl...Usage of immunological reactions in diagnostics of infectious diseases. Compl...
Usage of immunological reactions in diagnostics of infectious diseases. Compl...
 
Effects of silver nanoparticle (Ag NP) on oxidative stress biomarkers in rat
Effects of silver nanoparticle (Ag NP) on oxidative stress biomarkers in ratEffects of silver nanoparticle (Ag NP) on oxidative stress biomarkers in rat
Effects of silver nanoparticle (Ag NP) on oxidative stress biomarkers in rat
 
Epidemiology and Characterization of lipophilic yeast (Malassezia) from human...
Epidemiology and Characterization of lipophilic yeast (Malassezia) from human...Epidemiology and Characterization of lipophilic yeast (Malassezia) from human...
Epidemiology and Characterization of lipophilic yeast (Malassezia) from human...
 

Similar to Pyrogen article

Bacteriology&immunology lab2
Bacteriology&immunology lab2Bacteriology&immunology lab2
Bacteriology&immunology lab2
Brian Musalo
 
Antibiotics_Understanding_Basics
Antibiotics_Understanding_BasicsAntibiotics_Understanding_Basics
Antibiotics_Understanding_Basics
Ossama Motawae
 
Gram positive and gram negative
Gram positive and gram negativeGram positive and gram negative
Gram positive and gram negativeLeona Sucilan
 
Organelles & Diseases Related
Organelles & Diseases RelatedOrganelles & Diseases Related
Organelles & Diseases Relatedxoxositi
 
In vitro experiments of prokaryotic and eukaryotic antimicrobial peptide cyto...
In vitro experiments of prokaryotic and eukaryotic antimicrobial peptide cyto...In vitro experiments of prokaryotic and eukaryotic antimicrobial peptide cyto...
In vitro experiments of prokaryotic and eukaryotic antimicrobial peptide cyto...
AI Publications
 
Chloramphenicol Essay
Chloramphenicol EssayChloramphenicol Essay
Chloramphenicol Essay
Jill Ailts
 
INFLAMMATION PPT.pptx
INFLAMMATION PPT.pptxINFLAMMATION PPT.pptx
INFLAMMATION PPT.pptx
PushpanjaliDas4
 
Bacteria; structure, functions.pptx
Bacteria;  structure, functions.pptxBacteria;  structure, functions.pptx
Bacteria; structure, functions.pptx
MusharafRehman1
 
Antigens and toxins
Antigens and toxinsAntigens and toxins
Antigens and toxins
mohammed Qazzaz
 
application of biotechnology in pharmaceuticals
application of biotechnology in pharmaceuticalsapplication of biotechnology in pharmaceuticals
application of biotechnology in pharmaceuticals
SansarBookdepot
 
Semipermeability Of The Cell Membranes
Semipermeability Of The Cell MembranesSemipermeability Of The Cell Membranes
Semipermeability Of The Cell Membranes
Jill Lyons
 
In Vitro Cell Tests for Functional Food
In Vitro Cell Tests for Functional FoodIn Vitro Cell Tests for Functional Food
In Vitro Cell Tests for Functional Food
Institut Kurz
 
roozbahani-140104225550-phpapp01.pdfffff
roozbahani-140104225550-phpapp01.pdfffffroozbahani-140104225550-phpapp01.pdfffff
roozbahani-140104225550-phpapp01.pdfffff
MariamMansour32
 
sakshi yadav major project ppt.pptx
sakshi yadav major project ppt.pptxsakshi yadav major project ppt.pptx
sakshi yadav major project ppt.pptx
JMuthukumaran
 
Characteristics used in classification.pptx
Characteristics used in classification.pptxCharacteristics used in classification.pptx
Characteristics used in classification.pptx
Micro biology
 
Microbial Biotechnology Scope, Technique and Examples in Therapeutics
Microbial Biotechnology Scope, Technique and Examples in Therapeutics Microbial Biotechnology Scope, Technique and Examples in Therapeutics
Microbial Biotechnology Scope, Technique and Examples in Therapeutics
Zohaib HUSSAIN
 

Similar to Pyrogen article (20)

Bacteriology&immunology lab2
Bacteriology&immunology lab2Bacteriology&immunology lab2
Bacteriology&immunology lab2
 
Antibiotics_Understanding_Basics
Antibiotics_Understanding_BasicsAntibiotics_Understanding_Basics
Antibiotics_Understanding_Basics
 
Bacteriology
BacteriologyBacteriology
Bacteriology
 
Bacteriology
BacteriologyBacteriology
Bacteriology
 
Bacteriology
BacteriologyBacteriology
Bacteriology
 
Gram positive and gram negative
Gram positive and gram negativeGram positive and gram negative
Gram positive and gram negative
 
Organelles & Diseases Related
Organelles & Diseases RelatedOrganelles & Diseases Related
Organelles & Diseases Related
 
11
1111
11
 
In vitro experiments of prokaryotic and eukaryotic antimicrobial peptide cyto...
In vitro experiments of prokaryotic and eukaryotic antimicrobial peptide cyto...In vitro experiments of prokaryotic and eukaryotic antimicrobial peptide cyto...
In vitro experiments of prokaryotic and eukaryotic antimicrobial peptide cyto...
 
Chloramphenicol Essay
Chloramphenicol EssayChloramphenicol Essay
Chloramphenicol Essay
 
INFLAMMATION PPT.pptx
INFLAMMATION PPT.pptxINFLAMMATION PPT.pptx
INFLAMMATION PPT.pptx
 
Bacteria; structure, functions.pptx
Bacteria;  structure, functions.pptxBacteria;  structure, functions.pptx
Bacteria; structure, functions.pptx
 
Antigens and toxins
Antigens and toxinsAntigens and toxins
Antigens and toxins
 
application of biotechnology in pharmaceuticals
application of biotechnology in pharmaceuticalsapplication of biotechnology in pharmaceuticals
application of biotechnology in pharmaceuticals
 
Semipermeability Of The Cell Membranes
Semipermeability Of The Cell MembranesSemipermeability Of The Cell Membranes
Semipermeability Of The Cell Membranes
 
In Vitro Cell Tests for Functional Food
In Vitro Cell Tests for Functional FoodIn Vitro Cell Tests for Functional Food
In Vitro Cell Tests for Functional Food
 
roozbahani-140104225550-phpapp01.pdfffff
roozbahani-140104225550-phpapp01.pdfffffroozbahani-140104225550-phpapp01.pdfffff
roozbahani-140104225550-phpapp01.pdfffff
 
sakshi yadav major project ppt.pptx
sakshi yadav major project ppt.pptxsakshi yadav major project ppt.pptx
sakshi yadav major project ppt.pptx
 
Characteristics used in classification.pptx
Characteristics used in classification.pptxCharacteristics used in classification.pptx
Characteristics used in classification.pptx
 
Microbial Biotechnology Scope, Technique and Examples in Therapeutics
Microbial Biotechnology Scope, Technique and Examples in Therapeutics Microbial Biotechnology Scope, Technique and Examples in Therapeutics
Microbial Biotechnology Scope, Technique and Examples in Therapeutics
 

Pyrogen article

  • 1. Pyrogens, Endotoxin and the LAL test: An Introduction in Relation to Pharmaceutical Processing By Dr. Tim Sandle (email: timsandle@btinternet.com; website http://www.pharmig.blogspot.com) Introduction Pharmaceutical products which are injected into the human body are tested for pyrogenic substances. The most common, and arguably most important test, is for bacterial endotoxin. The pathological effects of endotoxin, when injected, are a rapid increase in core body temperature followed by extremely rapid and severe shock, often followed by death before the cause is even diagnosed. However, there needs to be large quantities of endotoxin within the human body for this to happen, and the endotoxin needs to be injected into the blood stream. Bacterial endotoxin is the lipopolysaccharide (LPS) component of the cell wall of Gram-negative bacteria. It is pyrogenic, and it is a risk to patients who are administered intravenous and intramuscular preparations. The purpose of the LAL test is to detect endotoxin (Guy, 2003). Pyrogenicity Bacterial endotoxin can be classed, among other things, as a “pyrogen.” Pyrogens are substances which, when injected into the mammalian body, will cause a variety of symptoms, the most recognizable of which is an increase in core body temperature. In the early days of the pharmacopeia, drug substances were classed as apyrogenic or pyrogenic, based, from 1942 and until the 1980’s, on the “pyrogen test” (whereby a quantity of the drug was injected into three rabbits, and the temperature response of the rabbits was noted). The rabbit pyrogen test was first described by Florence Seibert in 1925. The rabbit test is no longer widely used, and it has largely been replaced, for the testing of parenteral drug products, by the LAL (Limulus amoebocyte lysate) test. The LAL test is a method of the Bacterial Endotoxin Test (BET), for detecting the presence, and to go some way to determining the level of Gram-negative bacterial endotoxin in a given sample or substance. Current editions of the pharmacopoeia carry statements to the effect that where the term apyrogenic or pyrogen-free is used, it should be interpreted as meaning that samples of the product will comply with a limit for bacterial endotoxin. GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
  • 2. The association of fevers (pyrexia) has a long history. However, it was not until the early 20th century with the development of a rabbit pyrogen test that an understanding began to emerge in which bacteria could be classified into pyrogenic and non-pyrogenic types that correlate to their Gram-stain. Gram-negative bacteria were found to be pyrogenic, Gram-positive bacteria were generally not, and killed cultures of Gram-negative bacteria were comparable to live cultures in their ability to induce fevers. By the 1920’s it was apparent that sterility in parenteral pharmaceuticals could be no guarantee of nonpyrogenicity, and that if pyrogenicity was to be avoided, it was imperative to avoid bacterial contamination at every stage of manufacture of parenteral pharmaceuticals. The first official rabbit pyrogen test was included in United States Pharmacopeia (USP) in 1942. In recognition that the causative agent of pyrogenicity was filterable and heat stable, efforts were applied to identifying its chemical composition. Trichloracetic acid and phenol-water extractions of bacteria were found to be effective in isolating the pyrogenic element from bacteria. These extracts were chemically identifiable as lipopolysaccharide (or what is commonly described as bacterial endotoxin). Bacterial Endotoxin The structural rigidity of the bacterial cell wall is conferred by a material called peptidoglycan (also known as murein). It is a polymer consisting of sugars and amino acids that form a mesh-like layer outside the plasma membrane of bacteria, forming the cell wall (Madigan et al, 2009). In Gram-positive bacteria, peptidoglycan is present as a thick layer, which is outermost in the cell wall. In Gram-negative bacteria, the peptidoglcan is only a thin layer, and it is not the outermost layer. Gram-negative bacteria are sometimes described as having a cell envelope rather than a cell wall. The term envelope better describes the loosely attached layer of material called lipopolysaccharide, which is located outside a thin structural layer of peptidoglycan, as shown in Figure 1 below: GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
  • 3. Figure 1: Diagram of the Outer Bacterial Cell Wall (from Creative Commons Library) The outer layer of this lipopolysaccharide envelope is a permeability barrier effective against diffusion of exoenzymes into the external environment. This is an evolutionary feature which has arisen to allow Gram-negative bacteria (illustrated in Figure 2) to survive and increase in numbers in environments such as water, in which there are only low concentrations of organic nutrients. Macromolecular organic nutrients are trapped in the cell envelope as the water flows by, and then within the cell envelope they are hydrolyzed to smaller molecules, which can be taken into the cell. The important evolutionary advantages conferred by lipopolysaccharide are:  It contributes to the adhesion of Gram-negative bacteria to surfaces, allowing them to form as biofilms in aqueous environments.  It “attracts” and “entraps” organic macromolecules from aqueous environments.  It allows for entrapped organic macromolecules to be “recognized” by the cell so that specific enzymes can be synthesized to break them down into smaller fragments capable of passing through the peptidoglycan layer into the cell.  LPS also increases the negative charge of the cell membrane and helps stabilize the overall membrane structure.  It retains the enzymes synthesized by the cell so that they are not lost into the external environment [(Raetz and Whitfield,(2002)]. GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
  • 4. Figure 2: Gram's Stain Light Microscope Image, Showing Rod-Shaped Gram- Negative Bacteria (image from Creative Commons Library). Lipopolysacccharide is pyrogenic. Bacterial endotoxin is a synonym for lipopolysaccharide. Although intimately associated with the cell envelope of Gram-negative bacteria, lipopolysaccharide is constantly shed by the bacteria into the environment, much like the shedding of the outer layers of human skin. When Gram-negative bacteria die and lyse, all of their lipopolysaccharide is shed into the environment. Furthermore, when bacterial cells are lysed by the immune system, fragments of membrane containing lipid A are released into the circulation, causing fever, diarrhea, and possible fatal endotoxic shock (also called septic shock) (Brandberg, K. et al, 1996). There are some other substances which are also pyrogenic. They are unusual and are extremely rarely found associated with pharmaceutical preparations. Lipopolysaccharide has three distinct chemical regions, as illustrated in Figure 3 below: GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
  • 5. Figure 3: Diagram of LPS (from Creative Commons Library)  An inner core called Lipid A  An intermediate polysaccharide layer  An outer polysaccharide side chain Lipid A, embedded in the bacterial outer membrane, is responsible for pyrogenicity. The LAL Test The principle of the LAL test is a reaction between lipopolysaccharide and a substance (“clottable protein”) contained within amoebocyte cells derived from the blood of the Horseshoe Crab (of which Limulus polyphemus is the most commonly used species, as illustrated in Figure 4 below). LAL is an aqueous extract obtained after lysis of blood cells (amoebocytes).The reaction is specific. The reaction of the horseshoe crab to endotoxin (the formation of a clot) has been known since the 1950’s (Bang, 1956). GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
  • 6. Figure 4: Image of the Limulus “Horseshoe crab” (image from Creative Commons Library) When endotoxin comes into contact with LAL, it initiates a series of enzymatic reactions that result in the activation of a pathway to the production of at least three serine protease zymogens (Factor C, Factor B and a proclotting enzyme). This pathway alters amoebocyte coagulogen (an invertebrate fibrinogen-like clottable protein) to form coagulin gel. Serine proteases are enzymes that cleave peptide bonds in proteins, in which serine serves as the nucleophilic amino acid at the active site. They are found in humans as well as in the horseshoe crab (and indeed in all mammals). In humans, they are responsible for coordinating various physiological functions, including digestion, immune response, blood coagulation and reproduction. It is the blood coagulation reaction which is similar in both humans and the horseshoe crab (Cooper, 2004). The reference test in the pharmacopoeias is called the gel clot (or gelation) and is conducted on the endpoint principle. The description of the test and the necessary GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
  • 7. validation and accompanying controls is so detailed in both USP and Ph.Eur. (harmonized since 1999). The clotting mechanism of the blood of the crab is designed to prevent the spread of bacterial contamination throughout the horseshoe crab’s biochemical system. When the endotoxin of Gram-negative bacteria contacts with the horseshoe crab’s amebocytes, a series of enzymatic reactions begin. The pathway alters amebocyte coagulogen into a fibrinogen-like clottable protein, which forms a coagulin gel. The defence mechanism is also effective against fungi, hence, a similar reaction occurs in response to a fungal infection, which triggers the clotting cascade. In the reactions, glucans trigger the protease enzyme Factor G, whereas endotoxin triggers the Factor C enzyme, although the end result – coagulin – is the same (Moser, 2009). A considerable amount more glucan (1,000 times) is required to trigger the clotting cascade than the equivalent amount of endotoxin. The glucan required to trigger Factor G can be of varying molecular weights (such weights range from 3kD to 100kD). (Tanaka et al, 1993). This is illustrated below in Figure 5: E n d o to x in F a c to r C A c tiv e F a c to r C ß -(1 ,3 )-D -G lu c a n F a c to r B A c tiv e F a c to r B ------------------------A c tiv e F a c to r G F a c to r G P r o c lo ttin g E n z ym e C lo ttin g E n z ym e C o a g u lo g en C o a g u lin G e l F o rm a tio n Figure 5: LAL Clotting Cascade (adapted by the author) The LAL reagent is used for the gel-clot supplied with an identified sensitivity or label claim (lamda or λ), e.g., 0.03 endotoxin units (EU) per mL. This means that when mixed with an equal volume of the material under test, a gel or clot will form if the material contains 0.03 EU per mL or greater. For the kinetic methods, the lysate does not come with a label claim. The test sensitivity is determined by the lowest point of the standard curve used with each assay (Sandle, 2001). GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
  • 8. When it is necessary to quantify endotoxin concentration in a material, it is usual to test a series of doubling dilutions against the reagent in temperature-controlled conditions. The greatest dilution that gives a positive result (formation of a gel or clot) is the end point, and the concentration of endotoxin in the original material can be calculated by multiplying the dilution factor at the end point by the sensitivity of the LAL reagent. LAL tests require validation for each technician and each product before being applied routinely, and even then valid assays require careful internal standardization. These requirements are prescribed in detail in the pharmacopoeia to the extent that they do not merit repeating here. They require the use of a standard endotoxin termed “positive product control” (PPC), which is a known amount of endotoxin mixed with a test material to confirm the absence of interference. In the early days of the LAL test, endotoxin standards were variable, potency varied with the method of purification, the bacteria of its origin, and how it was formulated. In the 1970’s, USP and FDA commissioned Rudbach (Rudbach et al, 1976) to resolve the issue by preparing a reference endotoxin which was stable, could be chemically characterized, could be lyophilized without loss of activity, and which was free from biologically active proteins. This is termed RSE (reference standard endotoxin). Since RSE is expensive and potentially exhaustible, certified or calibrated standard endotoxin (CSE) is normally used for routine work, but not necessarily for fundamental research. The potency of a given batch of CSE is determined relative to the current Ph.Eur., USP or FDA lot of reference standard endotoxin (RSE) and is specific to particular batches of LAL reagent. Although the LAL test today is more robust, it remains open to a degree of variation (McCullogh, K.C. and Weider- Loeven, C., 1992). Today, both RSE and CSE are manufactured from Escherichia coli bacteria. With respect to international standards, the World Health Organization reference standard remains the chief standard from which standards approved by the pharmacopoeia are derived and equivalent to one another. The units of measurement for the LAL test are Endotoxin Units (EU). These are a measure of the activity of the endotoxin. Endotoxins differ in their biological activity or potency; the pyrogenicity or LAL reactivity of one endotoxin preparation may be very different from that of another of the same weight. Conversely, two endotoxin molecules may be different sizes and different weights but may have the same reactivity in an LAL test. The potency of an endotoxin determined with one LAL reagent lot may differ from that determined with another lot. Expressing endotoxin concentrations in EU’s avoids the issues of different potencies of different endotoxins and allows us to compare the results of different LAL tests performed in different GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
  • 9. labs. Consequently, it is not the usual practice to convert the results of an LAL test in EU/ml to units of weight of endotoxin per ml. The primary LAL test methods are the gel-clot, chromogenic and turbidimetric. The former is an end-point method whilst the latter two are kinetic methods. The kinetic methods are more sensitive than the gel clot method, because changes in turbidity and color are discernable by light-scattering devices at lower concentrations of endotoxin than those at which gels form. When using kinetic methods, the most important aspect is the standard curve for the endotoxin concentrations used in a standard curve to determine the test sensitivity. Therefore, the high point and low point in a standard curve determines the lower and upper level of endotoxin that can be detected. Rapid LAL tests are now commonplace on the market, and the industry is also moving towards the use of recombinant lysate, and the reliance upon the horseshoe crab may one day decrease. The LAL assay may be interfered with by the sample being tested. The interference may be caused by a number of different factors, such as pH, protein concentration, presence of chemicals (such as NaOH from rinsing cycles, etc). Interference may affect the lysate or the endotoxin. Inhibition or enhancement is normally detected through the use of spiked controls. Inhibition is arguably the greatest concern, because it can result in a failure to detect the true level of endotoxin in a sample. When conducting LAL tests, the dilution of samples is important. This is in order to minimize the effects of any component of the material being tested, which may be inhibitory to the LAL reaction. It is, however, imperative that the material being tested is not diluted too much, because this would ensure negative results, possibly false negatives. The way this is avoided is by having a Maximum Valid Dilution (MVD). LAL tests are only valid in situations where standard endotoxin can be shown to be detectable with the same efficiency in a test sample as in a control consisting of water known to be endotoxin free (this is termed LRW, or LAL reagent water). Sources of Endotoxin in Pharmaceutical Manufacturing A. Water Although all bacteria have some associated endotoxin, the most potent source of endotoxin is from Gram-negative bacteria. Endotoxins may be shed from viable bacteria during growth, or they may be associated with nonviable bacteria. GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
  • 10. Endotoxicity is not a function of microbial viability. The most common habitat for Gram-negative bacteria is water, where they have evolved to be able to survive and increase in numbers with minimal nutritional support and can adapt their metabolism to metabolize complex organic macromolecules that other bacteria cannot. In any form of pharmaceutical manufacture, water is one of the most serious potential sources of microbiological contamination. Water cannot be totally excluded from sterile products manufacturing facilities. The primary focus on endotoxin control in pharmaceutical manufacturing is on controlling it at its source – water. If endotoxin is not controlled at its source, it has the potential to create difficulties through manufacture to the finished product, potentially leaving no recourse but rejection. Endotoxin is practically impossible to remove terminally from pharmaceutical dosage forms. The measures taken to control contamination from water in sterile manufacturing facilities can be seen as measures taken to minimize the risk of endotoxin contamination of products. Pivotal to the control chain for any source of microbiological contamination is identification of those purposes for which it is necessary; in the case of water these are:  “Ingredient” water for aqueous sterile products  Water supplied for the cleaning of equipment and components  Water supplied to laundries  Water supplied for hand washing  Steam supplies to autoclaves, SIP systems, etc. The pharmacopoeia deal with ingredient water under two monographs, Purified Water and Water for Injection (WFI). To comply with the FDA requirements, WFI may be prepared by distillation or by reverse osmosis. For Europe, WFI can only currently be produced from distillation. With distillation, lipopolysaccharide has a molecular weight of around 106, heavy enough to be left behind when water is rapidly boiled off, as in a still. Where water has an endotoxin limit, it does not mean that it will automatically comply with that limit. Suitably controlled means of preparation, storage and distribution must be employed to ensure that the limits are complied with at point of use. Systems and user points require monitoring. WFI as prepared by the pharmacopoeially approved processes of distillation will comply (if sampled directly from the preparation point and not contaminated in sampling or testing). However, it is immediately fed to a storage tank and pumped GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
  • 11. round a distribution system where at least in theory, microbiological contamination may be lurking and shedding endotoxin. Figure 6: Laboratory Work in a Pharmaceutical Laboratory (image copyright Tim Sandle) When used in bulk for manufacturing purposes, the pharmacopoeia also apply a microbiological limit to WFI; this is not more than 10 cfu per 100 mL. This microbiological limit for WFI does not tie in with the endotoxin specification. The amount of endotoxin associated with Gram-negative bacteria is thought to be around 10-15g per bacterium. The first batch of Reference Standard Endotoxin (RSE) titrated at 1 EU = 2 x 10-10g. Therefore the endotoxin limit of 0.25 EU/ml for Water for Injections can be understood to correspond to 5 x 10-11g/ml, or about 104 bacteria /ml. Despite the fact that there is a great deal of uncertainty about these figures, and that Gram-negative bacteria not only have associated endotoxin but also shed endotoxin continuously, and that Gram-positive bacteria are generally not very endotoxic, it is apparent that water which meets the limit for endotoxin may exceed the limit for viable microorganisms (10 cfu/100ml). GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
  • 12. The means of controlling the development of Gram-negative (and other) microorganisms in water storage and distribution systems are:  Smooth internal surfaces in tanks and in pipework. Microorganisms adhere less well to smooth surfaces than to rough surfaces. Bear in mind that the degree of roughness and smoothness is a “micro” phenomenon; suitable and unsuitable internal finishes would not be discernable to the average triple blade shaver. Pipe joints can disrupt smoothness, as can welds.  Continuous movement of the water in tanks and rapid flow in pipework. Where shear forces are involved, microorganisms adhere poorly to surfaces. Where there is no movement of the water, there is no shear, and shear increases with speed of flow.  Avoidance of areas where water can remain stagnant.  These include “dead legs” – water may stagnate in branch pipes from a circulating main if the length of the branch is too long to allow the turbulence of the flowing main to disturb the contents of the branch pipe. FDA has defined “dead legs” according to their dimensions in the 1993 Guide to Inspection of High Purity Water Systems, but fundamentally the principle is to always minimize the length of branch pipes.  Water can also remain stagnant in valves, particularly at user points, and even more particularly at user points which are not in frequent and regular use. This is counteracted by use of so-called hygienic or “zero dead leg” valves, which although significantly better than the alternatives (say, ball valves), should not lead to a sense of false security; they can harbor endotoxin-shedding biofilms.  Ring mains should be sloped (have “drop”) from the point of origin to the point of return to ensure that systems are completely drainable.  Avoidance of leakage. Water leaks can cause bridging of water to the external environment through which bacteria may enter the system. Storage tanks should be equipped with filter on their air vents to prevent airborne microbiological ingress. They may even be held under a “blanket” of an inert gas, such as nitrogen.  High temperature storage and distribution. The risks of endotoxin-shedding biofilms, despite the best attempts at control above, are thought to be so consequential that the regulatory bodies require the temperature of storage and GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
  • 13. distribution to be maintained higher than 75°C. It should, however, be considered that 75°C is too high a temperature for most pharmaceutical formulation purposes. This means that user points are generally equipped with some form of cooling – heat exchangers used for this purpose may be a source of endotoxin and bacterial contamination and may thus cancel out many of the benefits of high temperature circulation. B. Depyrogenation Endotoxicity is not necessarily lost with the loss of viability of microorganisms. Lipopolysaccharide is not destroyed to any significant extent by sterilization treatments, such as steam sterilization, gamma radiation, ethylene oxide, hydrogen peroxide, etc. Lipopolysaccharide also passes through 0.22 μm bacteria retentive filters. It is claimed that endotoxin may be removed from liquids by up to 4 log10 reductions using 0.025 μm ultrafilters (which function as a molecular sieving process). There are two commonly used ways of eliminating endotoxin from materials:either by removing them or by inactivating them. The two primary means by which elimination is achieved is either by rinsing or by dry heat depyrogenation (Sandle, 2004). The normal method of removal is by rinsing the material with WFI. This is normally applied to rubber stoppers for vials. It is also what is done to vessels and major pieces of equipment used for sterile parenteral manufacture. A question arises as to whether this can be validated and assured? In answer, the sampling statistics are likely to be poor, and the test method is inaccurate, and probably there is not much there in the first place. This is an area where the regulators have looked at but there is no consistent standard, although it is implicit that a 3log10 reduction should be achieved by washing just as it should be by heat inactivation. From this, satisfactory validation of washing processes is not easy – or conversely, it can be quite easy, depending on the rigour of the validation design and the minutiae of process knowledge, for instance:  Where should the endotoxin “spike” be placed on the material or item being washed? The sensible location on vial stoppers must be that part of the stopper that potentially could come into contact with the product.  How much endotoxin should be “spiked” on items or materials? If too much endotoxin is added, for example, 1,000,000 EU/component, it could be too easily removed and may reveal little about the washing method used. Alternatively, too little may not even allow the demonstration of a 3 log10 reduction.  GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
  • 14.  How effective are washing processes? Air bubbles can cling to the surfaces of rubber stoppers and create areas that do not interact properly with the washing process.  How effective are washing machines? Batch washing machines generally work on a siphonage principle, such that removed materials (including endotoxin) are flushed upwards out of the washer rather than allowing the removed materials to settle back on the washed items. Continuous (semi-continuous washers (usually used for glassware and linked to depyrogenating tunnels) hold items upside down throughout the washing process; the final water rinse is frequently recycled to be the first wash, and probably adds less endotoxin than a lower grade of water would. Inactivation is done by dry heat. If there are materials and glass vials which are required for sterile parenteral manufacture which can be depyrogenated by dry heat, they should be depyrogenated by dry heat in an oven or a tunnel. The regulatory standard for validation of an endotoxin inactivation (depyrogenation) process is that it should be capable of reducing an endotoxin challenge through 3 log10 reduction. To ensure that this limit works, there is also a requirement to clean materials prior to dry heat depyrogenation with WFI – otherwise, at least in theory, an item could be contaminated with 10,000 EU prior to entering a validated endotoxin inactivation process and still emerge with 10 EU intact and be ready to contaminate the product. Dry heat depyrogenation is a complex process which is still poorly understood, with contradictory research data. The phenomenon which complicates the picture is that inactivation may approximate to Second Order1 chemical kinetics with a high initial rate of inactivation, then tail off to nothing. What this means in practice is that at any particular depyrogenating temperature will be subjected to some degree of inactivation in some period of time or other, but beyond that point, no further inactivation will occur by holding the material at that temperature. D-values for pure endotoxin (the time required to achieve one log10 reduction in activity) at 170°C are as high as 20 minutes, which would suggest that 3 log10 reductions are achievable in 60 minutes. However, due to the complexity of the kinetics, this is not necessarily true. All work with validation of depyrogenating processes must be done empirically with endotoxin challenges. This in itself is not easy, as the amount of endotoxin dried on an item (depending on the material of the item) may not be the maximum amount which can be recovered. Furthermore, the amount that can be recovered after treatment may only be a fraction of the amount 1For a second-order reaction, the rate of reaction is directly proportional to the square of the concentration of one of the reactants. GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
  • 15. that remains. Validation of endotoxin inactivation is a complex experimental area and requires that a lot of knowledge and a lot of well-considered controls to be included. Oven temperatures above 180°C will inactivate endotoxin through 3 log10. USP recommends 250°C, and peak temperatures approaching 350°C are achieved in tunnels. Based on these data, it is worth noting that each valid depyrogenating process is also an overkill sterilizing process. Conclusion This paper has provided an introduction to endotoxin and to the primary method for detecting endotoxin: the LAL test. The paper has also examined the risks of endotoxin to pharmaceutical processing and some of the control measures in place to reduce the risk of endotoxin contamination. The aim of the paper was to provide an introduction to this important subject. References Bang, F.B. (1956): “A Bacterial Disease of Limulus Polyphemus,” Bull. Johns Hopkins Hosp. 98, p.325. Brandberg, K. et al (1996): “Conformation of Lipid A, the Endotoxic Center of Bacterial Lipopolysaccharide,”J. of Endotoxin Research, Vol.3, No.3: 173-178. Cooper, J.F. (2004):“Microbiological Contamination Control in Parenteral Manufacturing,” In Williams, K. (Ed.) Microbial Contamination Control in Parenteral Manufacturing New York: Marcel Dekker, pp. 531-540. Guy, D. (2003): “Endotoxins and Depyrogenation,” in Hodges, N. and Hanlon, G., “Industrial Pharmaceutical Microbiology: Standards and Controls,”Euromed, pp. 12.1–12.15. Madigan, M. T., Martinko, J. M., Dunlap, P. V., and Clark, D. P. (2009): Brock Biology of Microorganisms. 12th ed., San Francisco, CA: Pearson/Benjamin Cummings, p.742. McCullogh, K.C. and Weider-Loeven, C. (1992): “Variability in the LAL Test: Comparison of Three Kinetic Methods for the Testing of Pharmaceutical Products,”J. of Parenteral Science and Technology, Vol. 46, 3: 69-72. Raetz, C.R.H. and Whitfield, C. (2002):“Lipopolysacchride Endotoxin,”Annual Review of Biochemistry, 71, pp. 635-700. GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor
  • 16. Moser, K. (2009): “Playing Hide and Seek with Endotoxin,”LAL User Group Newsletter, Issue 3, Number 2, pp.1-5. Rudbach, J.A., Akiya, F.I., Elin, R.J., Hochstein, H.D., Luoma, M.K., Milner, C.B., Milner, K.C. and Thomas, K.R. (1979): "Preparation and Properties of a National Reference Endotoxin," J. of Clin.Microbiol., 3(10): pp. 21-25. Seibert, F.B. (1925): “The Cause of Many Febrile Reactions Following Intravenous Injection,”Am. J. Physiol., 71: pp. 621-651. Sandle, T. (April 2001): “Performance Characteristics of Automated LAL Tests,”PharMIG News No. 4, pp. 3-10. Sandle, T. (June/July 2004): “Three Aspects of LAL Testing: Glucans, Depyrogenation and Water System Qualification,”PharMIG News No. 16., pp.3-12. Tanaka, S., Aketagawa, J., Takahashi, S., Shibata, Y., Tsumuraya, Y., and Hashimoto, Y. (1993): “Inhibition of High-Molecular-Weight-(1β3)-β-D-Glucan-Dependent Activation of Limulus Coagulation Factor G by Laminaran Oligosaccharides and Curdlan Degradation Products,” Carbohydrate Research, 244, pp. 115-127. GBPR, Inc. May 2012 Newsletter Evelyn Heitman, Editor