SlideShare a Scribd company logo
1 of 176
Download to read offline
aca® discrete clinical analyzer 
Dimension® clinical chemistry system 
OPUS™/OPUS™ PLUS/ 
OPUS Magnum® Immunoassay System 
Stratus® CS STAT Fluorometric Analyzer 
1 
Clinical Significance
2 
Here’s How to Order 
Additional Copies 
Copies of “Clinical Significance” may be purchased for $20 
each, including shipping and handling charges. To order, 
please write to us on your letterhead, specifying the num-ber 
of copies, your ship-to address and phone number. 
Mail to: 
Dade Behring Inc. 
Glasgow Business Community 
Mailbox 531 
P.O. Box 6101 
Newark, Delaware 19714 
U.S.A. 
Enclose a check or money order in the amount for 
the total number of copies ordered and make payable to 
Dade Behring.
3 
Table of Contents 
Page 
Introduction .........................................................................5 
Cancer Markers 
Carcinoembryonic Antigen ...................................................9 
Prostate Specific Antigen .................................................. 10 
Cardiac Markers 
Creatine Kinase-MB .......................................................... 15 
Myoglobin ......................................................................... 16 
Troponin-I ......................................................................... 18 
Coagulation 
General Information .......................................................... 23 
Antithrombin III ................................................................ 26 
Fibrin Degradation Products ............................................. 27 
Fibrinogen ......................................................................... 28 
Heparin ............................................................................. 29 
Plasminogen ..................................................................... 31 
Electrolytes 
Carbon Dioxide ................................................................. 35 
Chloride ............................................................................ 37 
Potassium ......................................................................... 38 
Sodium ............................................................................. 40 
Endocrinology 
Thyroid Function Tests: ..................................................... 45 
Free Thyroxine Index ...................................................... 45 
Thyroxine ........................................................................ 45 
Thyronine Uptake ........................................................... 45 
Thyroid Stimulating Hormone ........................................ 47 
Enzymes 
Acid Phosphatase ............................................................. 53 
Alanine Aminotransferase ................................................. 54 
Alkaline Phosphatase ........................................................ 55 
Amylase ............................................................................ 57 
Aspartate Aminotransferase .............................................. 58 
Total Creatine Kinase ........................................................ 60 
g-Glutamyl Transferase ..................................................... 61 
Lactic Dehydrogenase ....................................................... 63 
Lactate Dehydrogenase Isoenzyme 1 ............................... 63 
Liver Lactic Dehydrogenase .............................................. 63 
Lipase ............................................................................... 65 
Pseudocholinesterase ....................................................... 66 
Fertility Hormones 
Follitropin .......................................................................... 69 
Human Chorionic Gonadotropin (Choriogonadotropin) .... 72 
Luteinizing Hormone ......................................................... 76 
Prolactin ............................................................................ 80 
Page 
General Chemistry 
Bilirubin (Conjugated and Total) ....................................... 85 
Calcium ............................................................................. 87 
Cerebrospinal Fluid Protein ............................................... 89 
Cholesterol ........................................................................ 90 
Creatinine .......................................................................... 92 
Glucose ............................................................................. 93 
High Density Lipoprotein Cholesterol ............................... 96 
Iron ................................................................................... 98 
Magnesium ..................................................................... 100 
Neonatal Bilirubin ........................................................... 101 
Phosphorus .................................................................... 103 
Total Protein and Albumin ............................................... 105 
Triglycerides .................................................................... 107 
Urea (Blood Urea Nitrogen) ............................................ 109 
Urinary Protein ............................................................... 110 
Uric Acid ......................................................................... 113 
Immunology/Serology 
General Information ........................................................ 117 
Hypergammaglobulinemias ............................................ 119 
Hypogammaglobulinemias (Decreases in Immunoglobulins) ... 120 
C-Reactive Protein .......................................................... 122 
Therapeutic Drug Monitoring 
Aminoglycoside Antibiotics ............................................ 127 
Amikacin ....................................................................... 127 
Gentamicin ................................................................... 127 
Tobramycin ................................................................... 127 
Antiarrhythmic Drugs ..................................................... 129 
Digitoxin ....................................................................... 129 
Digoxin ......................................................................... 129 
Lidocaine ...................................................................... 130 
N-Acetylprocainamide .................................................. 130 
Procainamide................................................................ 130 
Quinidine ...................................................................... 131 
Antiasthmatic Drugs ....................................................... 132 
Theophylline ................................................................. 132 
Anti-convulsant Drugs: ................................................... 134 
Carbamazepine ............................................................. 134 
Ethosuximide ................................................................ 135 
Phenobarbital ............................................................... 135 
Primidone ..................................................................... 136 
Phenytoin ..................................................................... 137 
Valproic Acid ................................................................ 138 
Chemotherapeutic Drug .................................................. 139 
Methotrexate ................................................................ 139 
Vancomycin .................................................................... 141
4 
Table of Contents (cont'd) 
Page 
Toxicology 
Acetaminophen ............................................................... 145 
Ethanol ("alcohol," ethyl alcohol) .................................... 146 
Drug Screen Tests: .......................................................... 148 
Barbiturate Screen ........................................................ 148 
Benzodiazepine Screen ................................................. 148 
Tricyclic Antidepressant Screen ................................... 148 
Salicylate ......................................................................... 150 
Urine Drugs of Abuse Screen Tests Introduction ............ 151 
Urine Amphetamines Screen .......................................... 152 
Urine Barbiturates Screen ............................................... 153 
Urine Benzodiazepines Screen ........................................ 155 
Urine Cannabinoids Screen ............................................. 156 
Urine Cocaine Metabolite Screen .................................... 158 
Urine Methadone Screen ................................................ 159 
Urine Opiates Screen ...................................................... 160 
Urine Phencyclidine Screen ............................................ 161 
Specialty 
Ammonia ........................................................................ 165 
b2 Microglobulin ............................................................ 167 
Ferritin ............................................................................ 168 
Lactic Acid ...................................................................... 170 
Urinary Albumin for the detection of microalbuminuria ................ 171
5 
Introduction 
This collection of papers is intended to accompany the 
technical descriptions of the procedures performed by the 
Dade Behring aca® discrete clinical analyzer, Dimension® 
clinical chemistry system, OPUS™/OPUS™ Plus/OPUS 
Magnum® Immunoassay System and the Stratus® CS 
STAT Fluorometric Analyzer. The purpose is to explain 
in a very general way why the physician is interested in 
the substances measured. The approach has been to de-scribe 
briefly the normal physiology of the substance and 
then to list some of the diseases in which abnormal val-ues 
might be expected to occur. These are by no means 
complete lists; only the most common diseases have 
been included. Those wishing to know more about the 
clinical relevance of these tests are referred to any one 
of the standard textbooks of clinical pathology. 
Some comments may be useful relative to the nature of 
the specimen examined. Most of the tests are done on 
serum. Blood, serum, and plasma are words which are 
sometimes used interchangeably, yet they refer in fact to 
different substances. Blood needs no explanation except 
to say that it ordinarily refers to whole blood drawn from 
the veins, that is, venous blood. There are differences 
between arterial, capillary, and venous blood, but none 
of these are of much importance with respect to the tests 
now performed. 
Plasma is the fluid part of the blood. It is obtained by 
removing the cellular components (red cells, white cells, 
and platelets) from whole blood by centrifugation. In order 
to prevent clotting, an anticoagulant is added when the 
blood is drawn. Plasma must be used when coagulation 
factors are assayed, but it may also be used for the usual 
chemistry procedures when a very rapid answer is re-quired. 
For this purpose, heparin may be used as the 
anticoagulant. 
Serum is used in the measurement of most chemical 
substances in blood. It is the fluid part of the blood which 
remains after whole blood has been allowed to clot. Except 
for the absence in serum of some of the coagulation fac-tors, 
the differences between serum and plasma are of 
little consequence. Serum has the advantage of remaining 
fluid while plasma tends to clot eventually. 
Many of the substances measured are referred to erro-neously 
as though they were measured in a whole blood 
sample when in fact it is serum (or plasma) which is as-sayed. 
Thus physicians (and clinical chemists) refer to 
“blood sugar”, “blood urea nitrogen”, etc., when the more 
accurate reference would be to serum glucose or serum 
urea nitrogen. 
Frank C. Larson, M.D. Myrna Traver, MD 
Professor Emeritus Pathologist 
Department of Medicine Madison, WI 
Medical School 
University of Wisconsin-Madison
6
Cancer Markers 
7
8
Carcinoembryonic Antigen 
9 
Structure and Function: CEA is one of approxi-mately 
20 related complex glycoproteins that are mem-bers 
of the immunoglobulin superfamily1. It’s molecular 
mass varies between 150 and 300 kd2. The alteration in 
molecular mass is due to extensive, but differing 
glycosylation of the individual CEA molecules. CEA is 
one of a number of proteins that are expressed during 
normal differentiation of fetal tissue but are partially or 
completely suppressed in the adult3. It resides on the 
surface of epithelial cells of the gastrointestinal tract and 
a variety of other tissues and functions to mediate inter-cellular 
adhesion. In the normal adult, CEA is expressed 
in only trace amounts and is attached to the cell surface 
by way of a GPI (glycosylphosphatidylinositol) anchor4. 
In cancerous states, surface CEA can be shed in larger 
amounts and detected in the circulation and other body 
fluids. No clear picture has emerged regarding the role 
of CEA in cancer, but several studies suggest that over 
expression in carcinomas interferes with cell differentiation. 
Clinical Application: CEA is the most commonly 
used tumor marker for gastrointestinal tract tumors. Once 
thought to be specific for bowel cancer, elevated levels 
are also detected in association with a number of other 
cancers such as colorectal (70%), lung (45%), gastric 
(50%), breast (40%), pancreatic (55%), ovarian (25%), 
and uterine (40%). However, an abnormal serum level is 
not specific for cancer or unique to malignancy. CEA lev-els 
may also be elevated in some patients with benign 
conditions such as cirrhosis (45%), pulmonary emphy-sema 
(30%), rectal polyps (5%), benign breast disease 
(15%), and ulcerative colitis (15%)2. Up to 19% of smok-ers 
with no evidence of malignancy and 3% of a healthy 
population may also demonstrate elevated CEA levels1. 
Expected normal range values for nonsmokers are < 3.0 
ng/ml and < 5.0 ng/ml for smokers. Age, sex, and race 
have no significant effect on serum CEA levels. 
Caution should be exercised when interpreting patient 
CEA values. Quantitation is method dependent and there-fore 
values should only be compared when using the 
same method. Measurement of CEA is not recommended 
as a screening tool given that false-positives arise from 
non-cancerous conditions and false negatives may oc-cur 
since many tumors do not produce CEA. It’s 
quantitation can be used as an aid in the prognosis and 
management of cancer patients in whom changing con-centrations 
of CEA are observed. The American Society 
of Clinical Oncology (ASCO) recommends the use of 
CEA in colorectal cancer for preoperative evaluation, 
postoperative follow-up, and monitoring of treatment for 
metastatic disease.5 
Preoperative Evaluation: CEA may be ordered pre-operatively 
in patients with colorectal carcinoma if it would 
assist in staging and surgical treatment planning. Al-though 
elevated preoperative CEA (>5 ng/ml) may cor-relate 
with poorer prognosis, data are insufficient to sup-port 
the use of CEA to determine whether to treat a pa-tient 
with adjunct therapy.5 
Postoperative Follow-up: If resection of liver me-tastases 
would be clinically indicated, it is recommended 
that postoperative serum CEA testing may be performed 
every 2 to 3 months in patients with stage II or III dis-ease 
for 2 or more years after diagnosis. An elevated 
CEA, if confirmed by retesting, warrants further evalua-tion 
for metastatic disease, but does not justify the insti-tution 
of adjunct therapy or systemic therapy for presumed 
metastatic disease.5 
Monitoring During Treatment for Metastatic 
Disease: Present data are sufficient to recommend rou-tine 
use of the serum CEA alone for monitoring response 
to treatment. If no other test is available to indicate a 
response, CEA should be measured at the start of treat-ment 
for metastatic disease, and every 2 to 3 months 
during active treatment. Two values above baseline are 
adequate to document progressive disease, even in the 
absence of corroborating radiographs. CEA is regarded 
as the marker of choice for monitoring colorectal cancer.5 
References: 
1. E. P. Mitchell: Role of carcinoembryonic antigen in the manage-ment 
of advanced colorectal cancer. Seminars in Oncol-ogy. 
25:12-20, 1998. 
2. D. W. Chan and S. Sell. Tumor Markers. In: C.A. Burtis and E.R. 
Ashwood, eds. Textbook of Clinical Chemistry. Philadelphia: W.B. 
Saunders, 1994:897-927. 
3. J. Mendelsohn: Principles of Neoplasia. In: E. Braunwald, et al, 
eds. Principles of Internal Medicine. New York: McGraw-Hill, 
1987:412-431. 
4. B. Obrink: CEA adhesion molecules: multifunctional proteins with 
signal-regulatory properties. Current Opinion in Cell Biology. 
9:616-626, 1997. 
5. 1997 Update of recommendations for the use of tumor markers 
in breast and colorectal cancer. J. Clinical Oncology. 16:793-795, 
1998.
Prostate Specific Antigen 
10 
Structure and Function 
Prostate specific antigen (PSA) is a single chain protein 
consisting of 237 amino acids and one N-linked carbo-hydrate 
chain. The molecular weight of 28.5 kDa includes 
the 7% carbohydrate content. PSA is a serine protease 
member of the kallikrein family and shares amino acid 
sequence homology with at least two other family mem-bers, 
human glandular kallikrein and pancreatic renal 
kallikrein. PSA is produced mainly by the epithelial and 
ductal cells of the prostate gland, but is also present in 
the urethra, periurethral and anal glands of the male. Low 
level PSA production has also been observed in the 
female salivary glands and breast. PSA is a normal con-stituent 
of seminal fluid where its role is liquefaction of 
the coagulum to increase sperm motility. PSA may also 
function as a growth factor modulator via proteolytic 
cleavage of growth factor, as is the case with pro-EGF 
(epidermal growth factor) or the transport protein for the 
growth factor as is the case for the binding protein-3 for 
insulin-like growth factor. PTH-rp (parathyroid hormone 
related peptide) has also been shown to be a substrate 
for PSA suggesting a role for PSA in intracellular cal-cium 
regulation. The serum PSA test has become an 
important aid in the clinical management of patients with 
prostate cancer. 
Serum PSA 
Molecular Forms: In serum, PSA circulates as a “com-plex” 
with proteolytic enzyme inhibitors and also in a “free” 
form. The majority of measurable PSA consists of PSA 
complexed in a 1:1 ratio with a-1 antichymotrypsin (ACT). 
Smaller amounts of PSA are bound to a-1 antitrypsin 
and protein C inhibitor. PSA complexed with a-2 macro-globulin 
(2:1) ratio is not measurable with conventional 
immunoassays, as the PSA is engulfed by the larger 
a-2 macroglobulin. In some healthy subjects, the free 
form of PSA in serum may constitute a significant frac-tion 
of the total PSA (up to 40% for PSA values in the 
range of 1.0 - 4.0 ng/mL). The free form may consist of 
the proenzyme that is inappropriately released by the 
prostate cell, or “nicked” PSA which is enzymatically in-activated 
as a result of peptide bond cleavage at the 
lysine-lysine 145-146 position and possibly at other sites 
as well. The complexed form of PSA is cleared by the 
liver with an apparent half-life of 2.7 days, while free PSA 
has a half-life of approximately 1.5 hours. 
Expected Ranges for Total Serum PSA: Normal 
values for serum PSA are age related, reflecting the effects 
of increasing prostate size as a man ages. The age-referenced 
upper limits of normal (95th percentile) for 
Caucasian males are: 40-49 years, 2.5 ng/mL; 50-59 years, 
3.5 ng/mL; 60-69 years, 4.5 ng/mL and 70-79, 6.5 ng/mL. 
Asian men may have lower PSA values and African 
American men may have higher values than those given 
for Caucasian males. The value of 4.0 ng/mL has become 
the established cut-off value for diagnostic purposes. There 
have been no reports of significant diurnal or circadian 
variation for serum PSA, but some men may demonstrate 
significant biological variation, showing coefficients of 
variation as high as 50% for mean PSA concentrations 
below 4.0 ng/mL. 
Pre-Analytical Considerations: In some men, se-rum 
PSA values may be increased by prostatic massage 
as occurs with the digital rectal exam (DRE) or transrectal 
ultrasonography (TRUS). Thus, it is recommended that 
serum PSA values be determined prior to any of these 
activities in order to minimize false positive PSA results. 
Drug therapy with finasteride for benign prostatic hyper-trophic 
(BPH) disease may cause serum PSA values to 
decrease. Biopsy or surgery of the prostate will cause 
significant false elevation of serum PSA values and it is 
recommended that PSA measurements be delayed for 
at least six weeks after these procedures are performed. 
Human glandular kallikrein (HgK) is a potential source 
of false positive error in PSA immunoassays due to its 
high homology with PSA. However, careful selection of 
immunoreagents and structural differences between PSA 
and HgK appears to have minimized this potential problem. 
Non-prostatic sources of PSA in males are secreted into 
the urine tract and do not affect serum measurements. 
Clinical Application 
Patient Monitoring: Patients with cancer confined to 
the prostate are usually treated by prostatectomy or ra-diation, 
or may be offered “watchful waiting.” The “watch-ful 
waiting” approach consists of close monitoring to as-certain 
the rate of tumor growth. It is considered by many 
to be most useful for older men who have less aggres-sive 
tumors, that is, tumors with small volumes (less than 
1.0 cc) of low Gleason grade.
11 
After radical prostatectomy, serum PSA values should 
decline to values of less than 0.2 ng/mL after 2-3 days. 
In many urologic practices, post-surgical PSA values 
which rise above 0.5 ng/mL are strongly suggestive of 
recumbinant tumor or metastasis and will signal additional 
work-up or treatment. 
The pattern of PSA change for patients undergoing ra-diation 
therapy reflects the slower elimination of PSA 
producing tumor cells compared to the more immediate 
and complete removal of the tumor by surgery. Serum 
PSA values may decrease for 6-12 months before stabi-lization. 
The level to which the PSA must drop to reflect 
effective radiation therapy is controversial. However, it is 
apparent that the probability of relapse decreases with 
the extent to which the PSA concentration drops. The 
lowest relapse rate for radiation treated patients has been 
achieved when the serum PSA value drops to levels below 
0.2 ng/mL as demonstrated for patients treated by radical 
prostatectomy. Rising PSA values in patients treated by 
radiation therapy signals tumor recurrence and progres-sion 
of disease. It is hoped that the early treatment of these 
localized cancers will improve the overall cure-rate for pros-tate 
cancer. 
Hormonal treatments are commonly used to treat advanced 
prostate cancer. The primary strategy is to reduce the con-centration 
of testosterone by surgical castration (bilateral 
orchiectomy), estrogen therapy, or androgen blockage with 
the use of an LH-RH analogue and an anti-androgen. The 
response to hormonal therapy and the resulting decline in 
serum PSA concentration can be dramatic. However, the 
response to hormone therapy as well as the PSA decline is 
related to the composition of the tumor which consists of 
both hormonally sensitive and insensitive cell clones. Ulti-mately, 
the tumor will progress with the proliferation of the 
androgen resistant clone. Serum PSA may not increase to 
reflect the tumor burden in some of these cases, as the 
hormonally insensitive clone is less well-differentiated and 
may lack the ability to produce PSA to the same extent as 
hormonally sensitive clones. 
Staging and Prognosis: In routine practice, serum 
PSA provides little discrimination between patients with 
localized disease and those with extracapsular spread 
of cancer. While serum PSA varies directly with tumor 
volume, there is wide variation of PSA values and also 
considerable overlap of PSA ranges for the various dis-ease 
stages. Approximately 80% of prostate cancer pa-tients 
have an elevated PSA value defined by the cut-off 
value of 4.0 ng/mL. The majority of patients with PSA 
values of less than 4.0 ng/mL will have disease confined 
to the prostate, and most patients with PSA values greater 
than 10.0 ng/mL will have metastatic disease to lymph 
nodes or bone. Bone scans are used to confirm metasta-sis, 
however, bone scans may not be necessary in the 
routine workup of patients who present with no skeletal 
symptoms and have PSA values of less than 20.0 ng/ 
mL. Since serum PSA cannot accurately identify patients 
who have disease confined to the prostate, pretreatment 
serum PSA measurement provides no prognostic infor-mation 
for the individual patient. 
Other Prostate Diseases: The primary cause of 
false positive PSA values is BPH disease. While PSA 
production in BPH tissue is estimated to be only one-tenth 
that of prostate cancer tissue, the larger volume of 
BPH tissue leads to serum PSA values greater than 4.0 
ng/mL in 20-30% of patients. Most of these elevated BPH 
values are in the range of 4.0-10.0 ng/mL. Acute urinary 
retention, prostatic infarction and prostatitis may also 
cause elevated serum PSA values. 
Conclusion 
In the management of prostatic carcinoma, serum PSA 
values provide accurate assessment of the patient’s re-sponse 
to therapy. 
Prepared by: 
Herbert A. Fritsche, Ph.D. 
M. D. Anderson Cancer Center 
Houston TX
12
Cardiac Markers 
13
14
Creatine Kinase MB (CK Isoenzyme) 
15 
Physiology: Creatine phosphate and creatine kinase, the 
enzyme which synthesizes and degrades it, are important 
compounds in the storage and transfer of chemical energy 
within cells. When food from the diet is oxidized, its poten-tial 
energy is trapped as a high-energy compound which 
may be utilized immediately or stored in the cell until it is 
needed to perform mechanical work or to drive some en-ergy- 
requiring metabolic reaction. The most common of the 
“energy transfer” compounds is adenosine triphosphate, 
or ATP. For example, the energy for muscle contraction (and 
for many other metabolic reactions) is supplied by the hy-drolysis 
of the terminal pyrophosphate bond of ATP to yield 
adenosine diphosphate (ADP) and inorganic phosphate ion. 
ATP is normally produced by the oxidation of foods through 
a long series of reactions - a relatively slow process. There 
is only a small amount of ATP in muscle cells - about 6 
mmol/g. This is sufficient to sustain muscle contraction for 
only a short period of time. Some additional ATP can be 
produced fairly quickly by the partial degradation of glu-cose 
liberated by hydrolysis of stored muscle glycogen. 
However, at the onset of a sudden burst of intense muscle 
contraction, muscle cells require some form of “stored” 
energy which can be quickly converted to ATP. The high-energy 
compound which serves to “store” energy in the 
muscle cell is creatine phosphate (also called phosphocre-atine) 
which has a high-energy guanidophosphate bond. 
ATP can be quickly resynthesized by the one-step trans-fer 
of the phosphate group of creatine phosphate to ADP. 
This reaction is catalyzed by the enzyme creatine kinase 
(CK - previously known as creatine phosphokinase, or 
CPK). 
All body cells probably contain some CK, but the largest 
amounts are found in tissues in which a large amount of 
energy is stored as creatine phosphate. Skeletal muscle 
contains 2000-3000 units of this enzyme per gram of 
tissue, heart muscle 400 U/g, and the smooth muscle of 
the intestinal wall about 150 U/g. In contrast, there are 
only 3 U/g in liver. 
As is true of many other enzymes, several molecular 
forms (isoenzymes) of CK exist. They differ in their 
Michaelis constants and pH optima, but the physiologi-cal 
significance of this is unknown. Each of the three 
isoenzymes of CK is composed of two polypeptide 
chains. In the case of skeletal muscle CK, the chains are 
identical and are called M chains. The enzyme is called 
type MM. Brain CK is also composed of two identical 
chains, but these are different from the chains in muscle 
CK. Brain CK is type BB, composed of two type B 
chains. The third isoenzyme is a hybrid molecule, 
called type MB, consisting of one M chain and one B 
chain. Heart muscle, although it contains primarily type 
MM CK, also contains some type MB. This is of major 
clinical significance since CKMB is usually found only in 
heart muscle. 
Clinical Significance: Normal serum contains a small 
amount of CKMM and practically no CKBB or CKMB. 
When any tissue is damaged some of the intracellular 
enzymes may leak into the blood. Thus injury to skeletal 
muscle or heart muscle may be followed by a rise in serum 
CK. Measurement of serum total CK has been widely 
used in the diagnosis of acute myocardial infarction 
(AMI).* This disease is the most common cause of death 
in the western world. A portion of the heart muscle is de-prived 
of its blood supply, usually because of occlusions 
of a coronary artery by fatty plaques and/or by thrombus 
formation. The heart muscle cells die, and CK is released 
into the blood along with other enzymes such as AST 
and LDH. Serum CK levels correlate with the volume of 
damaged heart muscle and have been used as an index 
of the extent of the infarction. The usefulness of serum to-tal 
CK in MI has been compromised by the fact that some 
injury to skeletal muscle may accompany AMI, and skel-etal 
muscle contains 10 times as much CK as does heart 
muscle. For example, injections of pain medication into 
muscle (a common event in the treatment of a patient 
with suspected AMI) may cause enough inflammation of 
the muscle to give a spurious elevation in total serum 
CK. This problem can be circumvented by measuring 
only serum CKMB. This is a remarkable, sensitive and spe-cific 
test for heart muscle damage. As with total CK, in-creases 
in serum CKMB can usually be seen at 4 hours 
after the onset of chest pain. The enzyme level peaks 
soon after and returns to normal within 72 hours. 
*CK is also used in the diagnosis of some diseases of muscle and 
brain. These are discussed in the section on CK.
16 
Physiology 
Structure and Function: Myoglobin is a 17,800 dalton 
porphyrin-containing protein consisting of 153 amino acids. 
Myoglobin is almost exclusively found in the cytoplasm of 
skeletal and cardiac striated muscle. It constitutes 2% of 
the total muscle protein, and there are approximately 
2.8 mg of myoglobin per gram of heart tissue. No organ-specific 
isoforms of myoglobin have been demonstrated 
in humans thus far. One of the primary roles of myoglobin 
in muscle tissue is to bind oxygen and release it when the 
partial pressure (pO2) within the cells becomes very low. 
Clinical Significance 
Myoglobin Release from Cells: Clinical studies suggest 
that myoglobin, owing to its small size and cytoplasmic 
location, is one of the first tissue specific markers to 
be released from the compromised skeletal or cardiac 
myocyte. It is this characteristic on which its clinical sig-nificance 
rests. Serum and plasma levels of myoglobin 
increase within one hour of the onset of chest pain in 
acute myocardial infarction (AMI), thus most clinical labo-ratories 
today measure myoglobin levels. 
Leakage by Molecular Size 
When the myocyte experiences ischemia, one of the first 
responses is swelling. This swelling results in a sequen-tial 
loss of cytoplasmic constituents into the interstitial 
spaces in a manner that is roughly dependent on the 
molecular weight of each constituent. In this leakage 
model, very small molecular weight molecules like elec-trolytes 
are lost first. Then the macromolecules of increas-ing 
size like myoglobin (17,800 daltons) and enzymes 
like creatine kinase and its MB isoenzyme (CKMB) at 
86,000 daltons are lost from the cytoplasm. Myoglobin 
at 17,800 daltons is lost from the myocyte cytoplasm 
before cell death in ischemic conditions such as unstable 
angina; thus, circulating levels of myoglobin have been 
found to increase in ischemia. In addition, myoglobin 
seems to move directly to the coronary blood flow, while 
CKMB and lactate dehydrogenase isoenzyme 1 (LD1) 
are likely carried by lymphatic drainage from the injured 
myocardium. 
Myoglobin in Acute Myocardial Infarction 
It has been known since the first reports by Kagen and 
Stone in 1975 that serum and plasma myoglobin levels 
increase with AMI. Information has begun to accumu-late 
suggesting patients with AMI show a rise in myoglobin 
levels before either mass CKMB or cardiac troponin I (cTnI). 
The rapid clearance of myoglobin from the circulation in 
uncomplicated AMI, makes this marker suitable for de-tection 
of reinfarction in patients with recurrent chest pain. 
Myoglobin in Emergency Department 
Diagnosis of AMI 
Until the early 1980’s most patients presenting with chest 
pain were routinely being admitted to the hospital for 
observation. However, two parallel events helped to 
change how laboratory tests to detect AMI would be used 
in the Emergency Department (ED) setting. The first of 
these events was the adoption of the Diagnostic Related 
Group (DRG) method of payment for in-patient treatment 
of Medicare and Medicaid patients in 1983 and the rise 
of managed care in the private sector. The payment 
change introduced for the first time significant monetary 
deincentives for inappropriate admission of patients who 
turned out to not have myocardial events. Next, in 1985 
the first reports began to appear on the use of tissue-type 
plasminogen activator (TPA) and then subsequently 
streptokinase as alternatives to angioplasty by their ability 
to dissolve coronary occlusions. The common link be-tween 
all the clot busting drugs was that the sooner they 
were administered after the arrival in the ED, the better 
the patient outcome. Therefore, the detection of 
reperfusion by lab tests could significantly alter subse-quent 
patient management. These two events contrib-uted 
to the increased interest in finding better methods 
to correctly identify patients actually having myocardial 
events and to detect refusion as soon as possible after 
administration of thrombolytic agents. 
One of the difficulties in trying to make the diagnosis of 
AMI in the ED is that patients present at widely variable 
times after onset of chest pain depending on personal, 
societal and logistical considerations. Some locations 
may show 90% of patients presenting within 6 hours of 
chest pain onset, while others in different parts of the 
United States commonly see over 50% of patients pre-senting 
more than 12 hours after onset of chest pain. A 
further complication is that only 10% of those with chest 
pain will have ST-segment elevations and AMI, the clas-sic 
indicator of coronary artery occlusion. Another 10% 
will have non-diagnostic ECG changes (e.g., ST-segment 
decreases) and AMI. About 30% of those presenting with 
chest pain will have acute coronary syndromes and no 
AMI, while the remaining 50% of those presenting with 
chest pain will have no cardiac etiology. 
Myoglobin
17 
Laboratory testing is not involved in the diagnostic deci-sion 
to use thrombolytic therapy since, with occlusion, 
serum and plasma markers may not reflect the extent of 
the myocardial damage due to their accumulation on the 
other side of the blockage. The correct clinical diagno-sis 
of the remaining AMI presenting population, which 
have no ST-segment increase, relies increasingly on 
laboratory tests. Since patients may present within less 
than an hour to over 24 hours after onset of chest pain, 
laboratory diagnostic markers of AMI need to be selected 
based on when the levels in serum and plasma become 
diagnostic. Myoglobin rises quickly after AMI and becomes 
diagnostic within 2-4 hours post-infarct, peaking at 9-12 
hours and returning to baseline within 24-36 hours. Myo-globin 
tends to rise and fall sooner than other cardiac mark-ers 
with AMI. 
Myoglobin Utility on ED Presentation 
The time of patient presentation in the Emergency De-partment 
(ED) with possible AMI is nearly always more 
reliably known than the time, based on patient recollec-tion, 
of chest pain onset. Therefore, many reports relate 
the clinical utility of the various markers for AMI to the 
time of initial patient presentation in the ED. Generally 
myoglobin is found to have a better sensitivity for AMI in 
the first 4 hours after chest pain onset than mass CKMB, 
cTnI and cardiac troponin-T (cTnT), but has a lower speci-ficity 
than these other markers. A number of reports sug-gest 
the measurement of myoglobin as a diagnostic aid 
in “ruling-out” AMI with negative predictive values of up 
to 100% reported at certain times after onset of symptoms. 
A study in 1996 by Gornall and Levinott using serial sam-pling 
at 1, 2, and 6 hours after presentation showed myo-globin 
had the highest sensitivity, specificity, and nega-tive 
predictive value for AMI in the first 2 hours of ED 
presentation. They found myoglobin’s sensitivity to be 
77% at 1 hour, and 93% at 2 hours, thus the serial pre-dictive 
efficiency at presentation was 96% vs. Baseline 
ECG at 70% and CKMB at 62%. Another study also 
based on collecting blood at presentation and 1 and 2 
hours later, using a criteria of myoglobin above 100 ng/ 
mL or a change (+/-) from baseline of more than 50% 
found similar results. Sensitivity in the 0-2 hour period 
was 93%, with a 95% confidence interval of 59-92%. The 
literature suggests that by using the change from baseline 
presentation, myoglobin may yield the most sensitive and 
specific assessments of AMI detection if myoglobin mea-surements 
alone must be employed. 
Myoglobin with Perioperative Myocardial 
Infarction 
The same characteristics which make myoglobin useful 
for the detection of AMI in the ED have been found in 
perioperative MI in both cardiac and noncardiac surgical 
patients. For example, one study of coronary artery by-pass 
grafting (CABG) surgical patients found that myo-globin 
levels peaked within 1 hour after aortic unclamping, 
and returned to baseline within 4 hours. When a 
perioperative MI occurred, myoglobin levels were elevated 
1 hour after aortic unclamping, and continued to be 
significantly elevated at least 3 hours post-surgery 
compared with levels observed in non-MI patients. Myo-globin 
levels in patients who suffered perioperative MIs 
were found to be greater than 400 ng/mL 4 hours after 
the procedure. 
Cautions with Use of Myoglobin 
There are several clinical situations where myoglobin 
results should be used with caution in assessing AMI. 
The most significant source of false myoglobin eleva-tions 
is skeletal muscle injury. It has been found that 
myoglobin and mass CKMB may be elevated in some 
patients after electrical cardioversion while cardiac tropo-nin 
I remains normal. Individuals running marathons 
have also been shown to have elevations of myoglobin, 
mass CKMB and CK isoforms but normal cTnI levels. 
The specificity of myoglobin for AMI detection was found 
to fall from 82% in non-cocaine users to 50% in individu-als 
who recently used cocaine. Other clinical situations, 
such as severe burn victims, Rhabdomyolysis patients 
or other disease states where skeletal muscle injury is 
involved could cause increased levels of myoglobin.
18 
Physiology 
Structure and Function: Troponin C, I and T are polypep-tides 
that combine to form a CIT complex on the thin fila-ment 
(actin) portion of the contractile apparatus of cardiac 
and skeletal striated muscle. This troponin complex is not 
present in smooth muscle. Troponin C (TnC) can bind up to 
four calcium ions and is responsible for regulating thin fila-ment 
activation; troponin I (TnI) binds and inhibits acto-myosin 
ATPase thereby blocking myosin movement, and 
troponin T (TnT) binds TnI and TnC to the actin-tropomyo-sin 
filament positioning the complex on the filament. The 
size of the troponins vary from 23,500 daltons for TnI to 
37,000 daltons for TnT. TnC has a variable molecular weight 
depending on the number of calcium molecules bound. 
The binding affinity of TnC for TnI is very high at about 
108 L/mol if calcium is present, this enables TnI to wrap 
around TnC contacting both N- and C-terminal globular 
domain Ca2+ binding sites on TnC. Removal of Ca2+ with 
EDTA decreases the TnC - TnI binding affinity 1,000 fold. 
TnI has three striated muscle isoforms. Cardiac troponin 
I (cTnI) is found only in heart striated muscle. Slow twitch 
Troponin I (stTnI) is the isoform found in “red” striated 
muscle such as in the soleus and diaphragm. Lastly, fast 
twitch troponin I (ftTnI) is the isoform associated with 
the “white” striated muscle in skeletal muscles such as 
the biceps and triceps. Production of each isoform is 
controlled by a different gene. Because the amino acid 
sequence of each isoform is different, e. g., cTnI has 31 
unique amino acids at its N-terminal end, antibodies can 
be developed which can specifically detect cTnI, even in 
the presence of high levels of the stTnI and ftTnI striated 
muscle TnI isoforms. cTnI is not seen in fetal skeletal 
muscle, in regenerating skeletal muscle or in chronic 
muscle disease. TnT also shows differences between 
cardiac and skeletal muscle isoforms; however, the pres-ence 
of cardiac TnT in fetal skeletal muscle raises con-cern 
that regenerating skeletal muscle may also produce 
the cardiac TnT isoform. TnC has the identical amino acid 
sequence in cardiac and skeletal muscle so it is not use-ful 
as a marker for cardiac tissue damage. 
Clinical Significance 
Troponin Release from Cells: Most troponin is bound into 
CIT complexes as part of the myofibril structure within the 
muscle myocytes. cTnI and cTnT are present in stoichio-metrically 
equal amounts in the contractile apparatus of 
striated muscle. A smaller amount of cTnI, cTnT and TnC, 
approximately 6% to 8% of cTnT and 2.5% of total cTnI, 
resides in the myocyte’s cytoplasm. When the myocyte 
experiences ischemia, one of the first responses is swelling. 
The cell swelling is believed to result in a 
sequential loss of cytoplasmic constituents into the in-terstitial 
spaces. This cytoplasmic leakage occurs in a 
manner that is roughly dependent on the molecular 
weight of each constituent. In this leakage model, very 
small molecular weight molecules such as electrolytes 
are lost first. Then macromolecules of increasing size 
such as myoglobin (17,800 daltons) and enzymes such 
as creatine kinase (both MM and MB) at 86,000 daltons 
and lactate dehydrogenase (LD) at 135,000 daltons are 
lost from the cytoplasm. The macromolecule size, which 
defines the threshold between leaking and ruptured cells, 
is not well defined. However, it is likely that molecules 
the size of CKMB and LD may not be able to escape 
from an intact living cell. However, there is evidence that 
cTnI, at approximately 23,500 daltons, is lost from myo-cyte 
cytoplasm before cell death in conditions such as 
unstable angina. 
cTnI seems to be released from the cytoplasm of 
myocytes in patients with acute myocardial infarction 
(AMI) at about the same time as CKMB based on the 
timing of cTnI’s appearance in serum and plasma. Both 
mass CKMB and cTnI first attain optimal diagnostic effi-ciencies 
for AMI in the range of 80% at about 6 hours 
after onset of chest pain. However, cTnI is about 13 times 
higher in concentration in the myocardium than CKMB, 
offering the possibility of greater sensitivity to myocar-dial 
damage than CKMB. If the ischemia is not reversed 
the myocyte will die releasing structural elements, includ-ing 
the myofibril troponin CIT complexes into the circula-tion. 
Support for this sequential release model comes 
from the well known sequence of appearance of bio-chemical 
markers in serum and plasma. The bimodal rise 
of serum and plasma cTnT is also believed to be due to 
an initial cytoplasmic release followed by a second cTnT 
rise due to the slower myofibrillar degradation occuring 
with cell death. 
Free vs Complexed cTnI 
Little free cTnI is measurable in serum and plasma after 
acute myocardial infarction. Because of the high binding 
affinity of cTnI for TnC in the presence of Ca2+, nearly all 
measurable serum and plasma cTnI exists as a cTnI-TnC 
complex. The use of blood collection tubes containing 
strong Ca2+ chelators like EDTA (purple top tubes) dra-matically 
lowers the cTnI-TnC binding affinity, resulting 
in more free cTnI in the sample. Studies using highly 
selective antibodies for free and complexed cTnI show 
that nearly all of the measurable cTnI in acute myocar-dial 
infarction (AMI) is due to an increase of the cTnI-Troponin- 
I
19 
TnC complexes. Peak concentrations of total cTnI lev-els 
with AMI were found to be 5 to 12 times higher than 
the corresponding free cTnI levels. This information sug-gests 
that measurement of free cTnI should not contrib-ute 
significantly to the diagnostic efficiency of serum and 
plasma cTnI assays for myocardial infarction detection. 
Cardiac Troponin I (cTnI) Measurement 
cTnI’s unique 31 amino acid N-terminal sequence allows 
antibodies to be developed to highly discrete epitope sites 
on the polypeptide molecule. A properly selected anti-body 
permits highly specific cTnI measurement in the 
presence of severe skeletal muscle trauma and the high 
levels of serum skeletal muscle troponin isoforms that 
would result. Whether an immunoassay measures 
complexed or free cTnI depends on the cTnI epitopes to 
which the antibody is directed. AMI is generally associ-ated 
with a serum rise of the cTnI-TnC complex; there-fore, 
assays must measure this form of cTnI to be clini-cally 
useful in AMI assessment. There is also evidence 
that oxidation of cysteine-SH groups on cTnI may alter 
the exposed epitopes and the ability of some assays to 
detect cTnI. Since serum and plasma cTnI levels are 
normally low to non-detectable, clinical laboratory mea-surement 
of cTnI requires a highly specific antibody de-tection 
system and use of a final measurement system 
that can generate a detectable signal at low analyte con-centrations. 
One of the biggest problems faced in the clinical appli-cation 
of cTnI tests is that assays frequently employ an-tibodies 
with different cTnI epitope specificities, and each 
commercial method employs a different calibrator mate-rial. 
The result is that commercial cTnI methods from dif-ferent 
companies do not produce the same analytical 
results and may vary as much as 30 fold on the same 
sample. As a practical matter this means that diagnostic 
cutoff values and clinical utility decision points presented 
in the literature are highly method specific and should 
not be exported to another cTnI method without repeating 
the clinical studies needed to establish new numerical 
decision points associated with, for example, unstable an-gina 
and myocardial infarction. Therefore, the method used 
(e. g., Dade Behring Stratus®) will be indicated as relevant 
clinical literature findings are discussed in this article. 
cTnI in Acute Myocardial Infarction 
Several lines of evidence suggest cTnI may be a good 
marker for myocardial injury. cTnI concentration is about 
13 times higher in heart muscle than CKMB; therefore, 
small amounts of injury should generate a large increase 
from normal background levels. In practice, some clini-cal 
decision points may be near the assay’s analytical 
sensitivity since there is no measurable cTnI in serum or 
plasma of individuals without cardiac involvement. 
Indeed, cTnI (as measured by Dade Behring Stratus®) 
has not been found to increase in serum or plasma in 
conditions with acute skeletal muscle damage like trauma 
or rhabdomyolysis, or with chronic skeletal muscle dam-age 
such as polymyositis or Duchenne’s muscular 
dystropy, or after running a marathon. Because those 
same acute and chronic skeletal muscle pathologies can 
result in increases in CKMB that are not associated with 
AMI, the cTnI offers significantly improved specificity 
for myocardial damage. Chronic renal failure with hemo-dialysis 
or peritoneal dialysis also shows no increase of 
cTnI. Elevations in cTnI that correlate closely with myo-cardial 
injury have been verified by echocardiography. 
These characteristics all suggest that cTnI should be a 
better overall serum and plasma marker for routine as-sessment 
of myocardial infarction than CKMB. 
cTnI in Emergency Department Diagnosis of 
AMI 
One of the difficulties associated with making the diagno-sis 
of acute myocardial infarction (AMI) in the emergency 
department is that patients present at widely variable 
times after the onset of chest pain depending on per-sonal, 
societal and logistical considerations. Some loca-tions 
may show 90% of patients presenting within 6 hours 
of chest pain onset, while others (in different parts of the 
United States) commonly see over 50% of patients pre-senting 
more than 12 hours after the onset of chest pain. 
To further complicate matters, currently only 50% of pa-tients 
actually having AMI and 10% of individuals who 
present with chest pain, show the ST-segment eleva-tions 
associated with occlusion of coronary arteries and 
require emergency thrombolytic therapy or angioplasty. 
Another 10% of individuals who present with chest pain 
will have nondiagnostic electrocardiogram (ECG) 
changes (e. g., ST-segment decreases), and AMI. About 
30% of those presenting with chest pain will have acute 
coronary syndromes and no AMI, and in the remaining 
50% of their chest pain will have no cardiac etiology. 
Laboratory testing is often not involved in the diagnostic 
decision to use thrombolytic therapy since, with occlusion, 
cardiac markers may not reflect the extent of myocardial 
damage due to their accumulation on the other side of a 
blockage. The correct clinical diagnosis of patients pre-senting 
with AMI who have no ST-segment increase 
relies increasingly on laboratory tests. Since patients may 
present within less than an hour to over 24 hours after 
onset of chest pain, laboratory diagnostic markers of AMI 
need to be selected based on when the levels in serum 
and plasma become diagnostic. Myoglobin rises quickly 
after AMI and becomes diagnostic at about 1 to 3 hours 
after chest pain onset and remains elevated for 12 to 24 
hours. It is a useful marker for “ruleout” AMI within 24 
hours. CKMB begins to rise at 3 to 4 hours after onset of 
chest pain, becomes diagnostic at about 6 hours, peaks 
at 12 to 24 hours, and remains elevated about 24 to 36 
hours. LD isoenzyme 1(LD1) becomes diagnostic at 
about 10 hours after chest pain onset and remains el-
20 
evated for at least 72 hours. cTnI begins to rise 4 to 6 
hours after chest pain onset, becomes diagnostic at about 
6 hours, and will remain elevated for from 4 to 14 days. 
Consequently, cTnI tends to rise like CKMB in serum 
but stays increased for as long as LD1. This release 
pattern enables the use of cTnI to detect AMI over a 
longer period of time from the onset of chest pain than 
the other three traditional markers of AMI. 
Well controlled studies wherein serum or plasma levels 
of multiple markers of myocardial injury were examined 
in the same sample and related to time of chest pain 
onset show cTnI rises in serum at or before the time of 
CKMB. These studies consistently show that CKMB and 
cTnI have poor diagnostic sensitivity (e. g., less than 
50%) for myocardial injury before 6 hours after onset of 
chest pain. In one typical study, in the 7 to 36 hour pe-riod 
after chest pain onset cTnI sensitivity was 88 - 100% 
compared to CKMB sensitivity of 61% to 81%. 
Studies using serial testing of patients with possible acute 
myocardial events, but no ST-segment elevation, have 
consistently shown that absence of cTnI rise indicates a 
low risk of near term coronary death or infarction. In one 
study, 773 consecutive patients presenting with chest 
pain of less than 12 hours duration and no ST-segment 
elevation were tested on admission and again 4 hours 
later, or at a second time corresponding to at least 6 
hours after chest pain onset. The risk of coronary death 
or infarct in those with negative cTnI levels on both tests 
was only 0.3%. In another study, cTnI values less than 
0.6 ng/mL showed negative predictive values (NPV-%) 
results, to be 85% at presentation to the ED, 85.5 at 1 
hour later, 87.4 at 2 hours, 96.7 at 6 hours, and 98% at 
12-24 hours. The positive predictive value (NPV+%) at 
time of ED presentation was 25%, 40% 1 hour later, 
60% at 2 hours, 88% at 6 hours, and 88.9% at 12-24 
hours. This and other studies strongly suggest serial cTnI 
testing assists in the clinical decision to discharge or ad-mit 
a patient presenting at the ED with possible AMI. 
Available information suggests that cTnI or CKMB results 
from serum or plasma collected less than 4 hours after 
onset of chest pain may yield results still within the 
normal range that could change to positive in samples 
collected later. 
cTnI in Acute Coronary Syndromes 
Acute coronary syndromes (ACS) share a common 
pathophysiologic mechanism which is initiated when a 
coronary atherosclerotic plaque ruptures and a throm-bus 
forms that interrupts perfusion of myocardial tissue. 
Patients presenting with resting chest pain, but with no 
ST-segment elevation on the electrocardiogram (ECG), 
may have an ACS such as unstable angina or non-Q 
wave AMI. Detection of ACS frequently cannot be done 
clinically or by angiography. Consequently, analysis of 
serum markers of myocardial damage such as CKMB 
and cTnI have been routinely ordered to assist in detection 
of ACS. Making the diagnosis at the time the patient 
presents with the first ACS chest pain event is critical 
since, without intervention, 40% of those patients will 
have an AMI within 3 months. About 17% of those pre-senting 
with ACS will die within 3 months. Early inter-vention 
can reduce the risk of AMI to 8% and death to 
3% within the same 3 month period. 
Evidence now suggests that increased serum and 
plasma cTnI level in the first 24 hours from onset of chest 
pain is a much more sensitive and specific marker for 
ACS than CKMB. The prognostic value (predictive value 
of positive results) of cTnI increase is highest in patients 
presenting more than six hours after onset of chest pain. 
One study used samples collected from 1,404 patients 
presenting with unstable angina or non-Q wave AMI as 
part of the Thrombolysis in Myocardial Ischemia Phase 
IIIB (TIMI IIIB) study. It was found that 2.6% of patients 
presenting within 6 to 24 hours after chest pain onset, 
having cTnI (Dade Behring Stratus®) levels above 0.4 
ng/mL but negative mass CKMB levels, died from car-diac 
causes within 42 days. If cTnI was less than 0.4 ng/ 
mL, mortality within the 42 days was 5 fold less, at 0.5%. 
This and other studies suggest that cTnI is a useful di-agnostic 
marker for ACS, the utility of which will likely 
improve as detection methods for cTnI evolve. 
cTnI to Assess Cardiac Contusion 
Detection of cardiac injury in patients with chest trauma 
can be difficult because the serum and plasma level of mass 
CKMB could be elevated due to collateral skeletal muscle 
injury. Studies using transthoracic echocardiograms as the 
criteria for myocardial damage show that serum and plasma 
cTnI levels are much more specific than mass CKMB lev-els 
for detecting cardiac contusion. In one study (using Dade 
Behring Stratus® II) of 44 patients without cardiac contu-sion, 
none had serum cTnI levels above 0.35 ng/mL, sug-gesting 
no cardiac damage. In contrast, 26 patients had 
serum CKMB mass levels above 6.7 ng/mL, suggesting 
cardiac damage was present. That the commonly employed 
CKMB/Total CK ratio percent does not correct for all skel-etal 
muscle damage is suggested by the finding that 3% of 
the patients in this study had levels above 2.5%, a range 
generally attributed to cardiac damage. Several studies also 
suggest that cTnI may be useful for detection of unsus-pected 
perioperative myocardial injury or infarction. 
cTnI to Assess Myocardial Injury in Critically 
Ill Patients 
Critically ill patients are exposed to stresses that increase 
tissue oxygen demand at a time when oxygen supply to 
the myocardium could decrease due to a combination of 
variables such as hypoxia, fall in blood pressure, tachycar-dia, 
anemia and septicemia. Available evidence suggests 
that myocardial injury may go undetected in about 66% of 
critically ill patients in the absence of cTnI surveillance.
Coagulation 
21
22
23 
General Information 
Hemostasis, the process which prevents the loss of blood 
from a blood vessel, is accomplished through three inter-related 
events: vascular constriction, platelet aggregation, 
and coagulation. The initial vasoconstriction is short-lived. 
It enables the second event, platelet aggregation, to pro-duce 
a fragile, unstable platelet plug. Prolonged hemosta-sis 
depends upon the third phenomenon, the formation 
of a blood clot. To be effective a clot must form promptly 
and yet be closely confined to the damaged portion of 
the vessel. Coagulation is a highly complicated process 
in which the factors which prevent clotting are delicately 
balanced against those which promote it. Understanding 
the complex coagulation process is made more difficult 
by the lack of standard nomenclature. In some cases, 
the clotting factors are given their chemical names (i.e. 
calcium, prothrombin, fibrinogen). Other factors have also 
been named for their functions (proaccelerin, proconver-tin, 
antihemophilic factor). In still other cases factors were 
given the surname of the person in whom hereditary 
deficiency of that factor was first detected (Hageman, 
Fletcher, and Stuart). In an attempt to reduce confusion, 
Roman numerals have been assigned to most of the clot-ting 
factors. The numbers reflect the order of their dis-covery, 
and not the sequence in which they participate 
in the clotting reactions. Factors may exist in plasma as 
the inactive protein or precursor which is converted to 
the active form during the clotting process. The active 
form is identified by appending a lower case ‘a’ to the 
Roman numeral, e.g. factor Xa. Even in normal plasma 
the stage is constantly set for clotting. It can be triggered 
by events which take place either within the blood itself, 
in the blood vessel wall, or in the surrounding tissues. 
Regardless of how clotting is initiated, the last several 
steps are identical. These final steps result in the formation 
of a three-dimensional fibrin lattice in which platelets, 
red cells, and white cells are trapped. 
Fibrin is an insoluble protein which polymerizes spontane-ously. 
The initial polymers aggregate due to electrostatic 
and hydrophobic bonds. The immature fibrin aggregate 
is therefore gelatinous and easily broken up. As the clot 
matures, covalent (peptide) bonds are formed between 
the fibrin strands, strengthening and contracting the clot. 
This process is catalyzed by a transglutaminase, factor XIIIa. 
Fibrin cannot exist per se in plasma. It must neverthe-less 
be readily available in abundant quantity. Plasma 
therefore contains large amounts of a fibrin precursor, 
fibrinogen. Fibrinogen is converted to fibrin by the catalytic 
action of the enzyme thrombin. 
Figure 1 
Xa 
Prothrombin Thrombin 
Fibrinogen 
Fibrin Monomer 
Soluble Fibrin 
Stabilized Fibrin 
(Fibrin Clot) 
Ca++, PF3 
Va 
Ca++ 
XIII XIIIa 
Thrombin also is responsible for the activation of factor 
XIII (i.e. the conversion of protransglutaminase to trans-glutaminase) 
in the final step of clot formation. 
Thrombin is a key enzyme in coagulation. If thrombin 
existed in the active form in plasma, uncontrolled clotting 
would occur. It, too, circulates as an inactive precursor, 
prothrombin. The conversion of prothrombin to thrombin 
is catalyzed by factor Xa. This has proved to be a com-plex 
enzymatic reaction requiring the presence of factor 
Va, calcium, and a phospholipid-Platelet Factor 3 (PF3) 
(See figure 1). 
The activation of factor X is pivotal in clotting. It can come 
about in two different ways, referred to as the intrinsic 
and extrinsic pathways; the distinction is based upon the 
source of the activators. The intrinsic pathway is acti-vated 
by trauma within the vascular system such as ex-posed 
endothelium. In contrast, the extrinsic pathway is 
activated by external trauma such as a cut. Of the two 
systems, the intrinsic is slower but more important. 
The intrinsic system involves a series of reactions in 
which the product of one acts as a catalyst or cofactor 
for the succeeding reaction. This reaction chain is often 
referred to as a “cascade” (See figure 2). The intrinsic 
pathway is triggered when blood comes in contact with 
an unfamiliar surface such as a damaged blood vessel 
wall, causing a conformational change in factor XII and 
converting it to factor XIIa. Factor XIIa is a weak initiator 
of several reactions, including the activation of factor XI 
to XIa and the conversion of prekallikrein (Fletcher factor) 
to kallikrein. Factor XIa activates factor IX. Factor IXa is 
an endopeptidase which, in the presence of factor VIIIa 
converts factor X to factor Xa. The concomitant presence 
of factor VIIIa, platelet phospholipids (PF3), and calcium 
is required for the activation of factor X to proceed rapidly.
24 
Figure 2 
XII 
Activator 
Chemicals 
Collagen 
Exposed 
Endothelium 
Platelets 
XIIa 
kallikrein 
prekallikrein 
Intrinsic Pathway 
XI XIa 
IX IXa 
Ca++, PF3 
VIIIa 
X Xa 
Clotting is initiated through the extrinsic system when 
blood escapes from the vascular system and contacts 
damaged tissues. A poorly understood tissue factor called 
thromboplastin is released from the damaged cells. It 
forms a complex with calcium and factor VII which is able 
to activate factor X directly (See figure 3). Thus several 
of the early steps which must occur in the intrinsic system 
are bypassed in the extrinsic system. Clotting occurs 
much more quickly by this route. 
Figure 3 
Extrinsic Pathway 
VII 
Tissue Factor 
VIIa 
X Xa 
Control of Clotting 
Once clotting has started it is important to control its rate 
and, when hemostasis is achieved, to stop it altogether. 
This is accomplished by several mechanisms. The flow 
of blood past the site of the developing clot washes away 
the activated clotting factors and delivers them to the 
liver where they are removed from the blood. In addition 
there are several circulating clotting inhibitors, including 
antithrombin III (AT III), alpha-1-antitrypsin, C1 inhibitor, 
and alpha-2-macroglobulin. Of these, AT III is the most 
important. As its name implies, its primary role is to com-bine 
with and thereby inactivate thrombin. This inhibition 
proceeds relatively slowly but is greatly accelerated in 
the presence of heparin. AT III also inactivates factors 
Xa, and to a lesser extent, inactivates IXa, XIa, and XIIa. 
Inhibition of these factors is also accelerated by heparin 
(See figure 4). 
Figure 4 
Antithrombin III 
+ Heparin 
Inactive 
Xa 
Complex 
Inactive 
Thrombin 
Complex 
Xa Thrombin
Inactive Xa 
Complex 
Activators 
Kallikrein (Intrinsic) 
Tissue Activator (Extrinsic) 
Urokinase 
Streptokinase 
Antithrombin III 
+ 
Heparin 
Prothrombin Thrombin 
25 
Clot Dissolution 
Blood clots are not intended to be permanent. Their pur-pose 
is to stop the flow of blood until the damaged blood 
vessel can be repaired. Normally all but the largest clots 
will be completely dissolved. Dissolution, which begins 
almost as soon as the clot is formed, is accomplished by 
the hydrolytic action of the enzyme plasmin. Plasmin also 
hydrolyzes fibrinogen and other clotting factors. Therefore, 
it cannot exist in its active form in plasma. An elaborate 
control system exists which allows clot lysis to proceed while 
protecting clotting factors against destructive hydrolysis. 
Normal plasma contains an inactive precursor of plasmin 
called plasminogen. Plasminogen has an affinity for fibrin 
molecules and is incorporated within the fibrin strands 
as the clot forms. Plasminogen remains dormant inside 
the clot until activated by specific enzymes called plas-minogen 
activators. Such activators have been found in 
tissues, in urine, in plasma, and, most importantly, in the 
endothelial cells which line blood vessels. Factor XIIa 
also accelerates the activation of plasminogen. Thus, the 
factor which triggers the clotting process also contributes 
to clot dissolution. The fact that a clot forms at all is ex-plained 
by the relative rates of the two processes. Clot for-mation 
occurs rapidly until it is slowed by specific limiting 
mechanisms. Clot dissolution, on the other hand, is slow in 
starting, but is a continuous process (See figure 5). 
Hemostasis 
XII XIIa 
Activator 
Chemicals 
Collagen 
Exposed 
Endothelium 
Platelets 
Kallikrein 
Prekallikrein 
Kallikrein 
Extrinsic – Intrinsic Interaction 
Extrinsic System 
Intrinsic System 
Common Pathway 
Fibrinolytic Pathway 
Inhibition 
VIIa 
XIa 
Tissue Factor 
XI 
VII 
IX IXa 
X Xa 
XIII XIIIa 
Ca++, PF3 
VIIIa 
Ca++, PF3 
Va 
Activators 
Kallikrein (Intrinsic) 
Tissue Activator 
(Extrinsic) 
Urokinase 
Streptokinase 
Inactive 
Thrombin 
Complex 
inactive 
plasmin 
complex 
Fibrin (ogen) 
Degradation 
Product 
Stabilized Fibrin 
Plasminogen 
a2antiplasmin 
Plasmin 
Fibrinogen 
FpA 
FpB 
Fibrin Monomer 
Soluble Fibrin 
Figure 5 
inactive 
plasmin 
complex 
Fibrin (ogen) 
Degradation 
Product 
Fibrinogen 
Fibrin Monomer 
Soluble Fibrin 
Stabilized Fibrin 
Plasminogen 
a2antiplasmin 
Plasmin 
Active plasmin sometimes finds its way into the plasma; 
it may escape from a dissolving clot or be formed if plas-minogen 
activators gain access to the blood stream. 
When this happens, fibrinogen and other essential clotting 
proteins, including factors V and VIII, are destroyed. To 
prevent this, plasmin inhibitors, the most important 
of which is alpha-2-antiplasmin, quickly inactivate any 
plasmin in the circulation.
26 
Antithrombin III 
Physiology: Antithrombin III (AT III) is a plasma pro-tein 
which is an inhibitor of serine proteases. As the name 
implies, its main physiologic function is the inactivation 
of the key clotting enzyme, thrombin. It also inactivates 
other clotting and clot-lysing enzymes, including factors 
Xa, IXa, XIa, XIIa, kallikrein, urokinase, and plasmin, all 
of which are serine proteases. AT III is a glycoprotein 
with a molecular weight of 65,000. It migrates as an al-pha- 
globulin on electrophoresis. It is synthesized by the 
liver and is widely distributed in body fluids. Its physi-ologic 
half-life is about four days. As with other coagulation 
factors, the exact catabolic pathway is not known. 
Furthermore, normal plasma contains more AT III than 
prothrombin. The fact that clots are able to form at all is 
due to the fact that the inhibitory reaction is much slower 
than the thrombin-catalyzed fibrinogen to fibrin reaction. 
This situation is altered in the presence of heparin, which 
greatly increases the inhibitory activity of AT III through 
a conformational change in the molecule. Heparin is 
therefore an accelerator of AT III. This appears to be the 
mode of action for this important anticoagulant. 
Clinical Significance: AT III is normally present in 
plasma. In disease, its concentration may be either 
increased or decreased. 
Increases: Damage to body tissue may produce a rise 
in plasma AT III which could result in an increased ten-dency 
to bleed. 
Decreases: Decreased AT III is of much greater clinical 
importance. A reduction in AT III leads to an increased 
tendency for blood to clot within the blood vessels (throm-bosis). 
Once formed thrombi tend to enlarge or progagate. 
Rapidly propagating thrombi may break away from the 
site where they are formed and lodge elsewhere in the 
vascular system (embolization). Serious consequences, 
even death, may follow embolization to the heart, lung 
or brain. AT III levels less than 60% of normal are asso-ciated 
with increased risk of thromboembolism. 
An inherited inability to synthesize normal AT III accounts 
for only a very small percentage of patients (2%) with 
thromboembolism. Several families with this disorder 
have been discovered. These individuals have repeated 
episodes of thromboembolism. Thromboembolism is very 
rare among young persons; when it does occur, hereditary 
AT III deficiency should be considered as a possible cause. 
Decreased AT III is more commonly an acquired condition. 
Impaired synthesis or increased utilization, or a combi-nation 
of the two, may be the basic problem. Since AT III 
is synthesized in the liver, diseases of this organ may be 
responsible for decreases in plasma AT III levels. 
AT III is a molecule about the same size as albumin and, 
consequently, may be easily lost in the urine in kidney dis-ease. 
Nephrosis, which is characterized by massive albu-minuria, 
may also be accompanied by AT III deficiency. 
AT III deficiency may result from (as well as cause) exten-sive 
intravascular coagulation (DIC). In DIC, thrombin, plas-min, 
and other serine proteases may be produced at such 
a rate as to deplete plasma AT III. 
Prolonged use of oral contraceptives is accompanied by 
some decrease in AT III levels. The exact significance of 
this is not known, but it may contribute to the slightly 
increased incidence of thromboembolism observed 
among women takin anovulatory medication. The assay 
of plasma AT III is helpful in monitoring heparin therapy. 
Patients with low AT III levels have a reduced response 
to heparin. Heparin may itself lower AT III levels. When 
this occurs, the patient has an increased tendency toward 
thrombosis after the heparin therapy is discontinued. 
Regardless of cause, low AT III signifies a hypercoagulable 
state which can be dangerous and which, with proper 
treatment, can be avoided.
Fibrin Degradation Products 
27 
Physiology: The proteolytic enzyme, plasmin, cleaves 
fibrin (and fibrinogen), producing variable-sized, soluble 
fragments called fibrin split products, or fibrin degrada-tion 
products. In normal circulating plasma there is very 
little active plasmin, hence only small amounts of FDP 
are formed and are quickly removed from the body by 
the liver. 
Plasmin activity may be increased secondary to a variety 
of disease processes. The level of FDP will increase if the 
rate of their formation exceeds the rate of removal. They 
have an inhibitory effect on the conversion of fibrinogen 
to fibrin, and may also impede platelet aggregation. Fi-brin 
degradation products are measured in serum, using 
an antibody against fibrinogen and FDP. The fragmenta-tion 
of fibrin/fibrinogen by plasmin destroys the ability of 
fibrinogen to form an insoluble clot, but only slightly 
changes the antigenic sites. Fibrinogen, which will cross-react 
with the FDP antibody, is removed from the sample 
by allowing it to clot. 
Clinical Significance: Fibrin degradation products 
are present in plasma in small amounts normally. In-creases 
indicate clot lysis is proceeding at a faster than 
normal rate. 
The FDP assay may also be clinically useful in monitoring 
patients receiving streptokinase, a plasminogen activator 
used in treatment of thromboembolic disorders such as 
deep vein thrombosis, pulmonary embolism, and myo-cardial 
infarction. Patients receiving streptokinase should 
have elevated levels of FDP. 
Increases: FDP levels are increased in a number of 
diseases such as liver disease, malignant tumors, renal 
failure, transplant rejection, and disseminated intravascu-lar 
coagulation (DIC). The highest levels occur in DIC (de-scribed 
more fully in the section dealing with fibrinogen). 
In DIC, the delicate balance between clot formation and 
clot dissolution is disturbed and widespread intravascular 
clotting occurs. In malignancy or certain obstetrical 
complications large amounts of procoagulants may be 
released into the blood stream. In massive injury or burns, 
the extrinsic clotting pathway is activated. If there is 
significant damage to vascular endothelium, plasma is 
exposed to collagen and collagen-like substances which 
can activate the intrinsic pathway. Whatever the cause 
of DIC, there is a compensatory increase in the fibrin-olytic 
activity, and an increase in levels of FDP.
Fibrinogen (Clotting Factor I) 
28 
Physiology: Fibrinogen is the soluble precursor of in-soluble 
fibrin, the major component of a blood clot. It is a 
long, rod-like, moderately large (340,000 daltons) glyco-protein. 
The molecule is constructed of polypeptide chains 
of three different types designated alpha, beta and gamma, 
The alpha, beta and gamma chains are joined together by 
disulfide bonds near their N-terminal segment, forming a 
monomer. Two such monomers are joined at their heads 
again by disulfide bonds to form a dimer of six polypeptide 
chains. When fibrinogen is attacked by the hydrolytic en-zyme 
thrombin, four segments, two alpha and two beta, 
are split off and released as fibrinopeptides A and B (FpA, 
FpB). Assay of fibrinopeptides may be used to estimate the 
presence and extent of thrombosis. Once thrombin has split 
off these terminal segments, the remaining molecules are 
free to polymerize into fibrin strands which ultimately form 
the basic structure of the blood clot. Most of the total body 
fibrinogen is intravascular. The liver synthesizes 2-5 grams 
of fibrinogen per day. Its physiologic half-life is about 4 
days. The normal catabolic path way for fibrinogen is not 
well understood. 
Fibrinogen 
FpA 
FpB 
Fibrin 
Clinical Significance: Fibrinogen is measured in 
plasma. Its concentration may be increased or decreased. 
In general, decreases are of more clinical importance 
than increases. 
Decreases: Decreases in plasma fibrinogen levels 
might be expected in liver disease since that organ is 
the site of synthesis. Although hypofibrinogenemia does 
occur in liver disease, it is not common. 
The most common cause of low plasma fibrinogen levels 
is disseminated intravascular coagulation (DIC), a condi-tion 
in which blood clots form throughout the microvascular 
system. The process may be either acute or chronic with a 
mild or fatal outcome. There are many causes of DIC. It 
accompanies many serious complications of childbirth. Pre-mature 
separation of the placenta (abruptio placenta) and 
amniotic fluid embolism (the forced injection of amniotic 
fluid into the maternal circulation during labor) are the two 
most common obstetric incidents, in which DIC occurs. 
Other problems which may produce DIC are retention of a 
dead fetus and abortion induced by the intra-amniotic in-jection 
of hypertonic saline. 
DIC can have two serious consequences: The clots may 
occlude important blood vessels and the consumption 
of fibrinogen (and factors VIII and V) may outstrip pro-duction. 
When fibrinogen levels fall to the point where 
blood is unable to clot, dangerous bleeding ensues. 
Fibrinogen levels below 100 mg/dl are associated with 
an increased risk of bleeding. 
Increases: Fibrinogen is one of several proteins which 
increase in the plasma whenever tissue is damaged. Fi-brinogen 
is not usually assayed directly for the purpose 
of assessing tissue damage. On the other hand, mea-surement 
of the rate at which red cells in anticoagulated 
blood aggregate and settle to the bottom of the tube, the 
red cell sedimentation rate, is commonly used to detect 
and monitor inflammation. Fibrinogen is more potent than 
any other plasma protein in increasing the erythrocyte 
sedimentation rate. 
A large number of inherited disorders of fibrinogen synthe-sis 
have been discovered. A total inability to synthesize this 
protein (afibrinogenemia) is known, but it is extremely rare. 
There are many genetic disorders which presumably are 
due to errors in the polypeptide structure although their exact 
nature remains to be defined. These disorders are referred 
to as dysfibrinogenemias. The quantity of plasma fibrino-gen 
is normal in such persons, but the ability to form clots 
(functional activity) is impaired, usually due to defective 
polymerization. For this reason, assays whose end point is 
clot formation are abnormal, and are interpreted as a 
decreased fibrinogen level. From a functional point of view, 
this interpretation is correct. 
Low fibrinogen levels occasionally result from the presence 
in plasma of plasmin which has not been inactivated. Plas-min 
degrades fibrinogen as it does fibrin. This process is 
known as fibrinogenolysis. It may result from the use of 
plasminogen activators such as streptokinase in the treat-ment 
of clotting disorders, or the release of activators from 
malignant tumors. It may also be due to failure of synthesis 
of adequate amounts of plasmin inhibitors.
29 
Heparin: Heparin was discovered in 1916 by a medi-cal 
student, Jay McLean, who was attempting to isolate, 
from various tissues, a substance (thromboplastin) which 
promotes coagulation. McLean found that certain extracts 
of tissues contained a substance which prevented instead 
of promoted coagulation. He and his mentor, Professor 
W. H. Howell, named this substance “heparin” because 
of its abundance in the liver (hepar). High concentrations 
were also found to be present in other tissues notably 
lung and the walls of small intestines, the two most com-mon 
sources of commercial heparin today. 
Chemistry: Heparin is a mucopolysaccharide bearing 
chemical resemblance to hyaluronic acid, chondroitin 
sulfate and keratan sulfate. It is a polymer made up of 
repeating units of sulfated and non-sulfated D-glucosamin, 
L-iduronic acid, and D-gluronic acid. Phar-macologic 
preparations of heparin are heterogeneous 
with a distribution of molecular weights between 5,000 
and 30,000 daltons. There are a large number of O- and 
N- sulfate linkages and carboxyl groups, making it the 
strongest organic acid in the body. 
Physiology: Mast cells or endothelial cells are the 
source of heparin in the body. Mast cells are medium 
sized mononuclear cells, the cytoplasm of which is stuffed 
with granules which stain intensely with basic stains. 
These granules have been found to be comprised mainly 
of heparin. Mast cells are found throughout loose con-nective 
tissue in the body but are in greatest numbers in 
the organs known to be rich in heparin, i.e., lung and 
liver. Two main functions have been ascribed to heparin. 
The first is the prevention of intravascular clotting of blood 
and the second, the clearing of fat from the blood plasma 
after the ingestion of a heavy fat meal. The anticoagulation 
function of heparin is generally believed to be the most 
important one. Heparin has no known effects except those 
on blood. 
Effects On Coagulation: Heparin inhibits coagulation 
of blood in vivo (in the body) and in vitro (in glass). This 
inhibitory action requires the presence in blood plasma of 
a naturally occurring alpha-globulin, antithrombin III. Anti-thrombin 
III (discussed elsewhere in this booklet) is syn-thesized 
in the liver. Its presence in plasma prevents the 
spontaneous intravascular coagulation of blood, i.e., it helps 
maintain blood in the fluid state. Antithrombin III is only slowly 
active in the absence of heparin but becomes very active 
in its presence. The apparent mechanism of action involves 
first the interaction of heparin with the lysine group of anti-thrombin 
III which exposes a reactive arginine group which 
in turn combines with and inactivates serine proteases. 
Coagulation occurs as a result of a series of enzyme 
reactions. At each step, an inactive proenzyme is converted 
to an active protease which in turn converts a subse-quent 
proenzyme to another active protease enzyme. 
There are several proteins involved in the coagulation of 
blood. Of these, some are activated by serine proteases 
(e.g., Prothrombin, and factors VII, IX, X, XI, and XII). 
Others are co-factors in these reactions (factors Va, VIIIa, 
kallikrein, and kinin) and one is converted to a transami-dating 
enzyme (factor XIII). The antithrombin III-heparin 
complex appears to neutralize all of the serine proteases. 
(The complex will also neutralize plasmin which is any 
enzyme active in dissolving clots). Heparin, then, acts at 
many stages of clot formation but the net effect is the 
inhibition of the formation of thrombin from prothrombin. 
Heparin, unlike the anticoagulant, Coumadin®, does not 
interfere with the synthesis of coagulation-related proteins. 
Heparin has a weak inhibitory effect on thrombin itself. 
This is probably not physiologically or pharmacologically 
important because it requires 30 to 40 times as much 
heparin to inhibit the action of thrombin than to prevents 
its formation. 
Lipoprotein Lipase Activation: Heparin appears 
to have a specific effect on the release of an enzyme, 
which catalyzes the hydrolysis of triglycerides in chylo-microns, 
releasing free fatty acids which rapidly enter 
tissues. This enzyme, lipoprotein lipase, has the effect of 
recuing the turbidity of serum. Because of this effect, it is 
also known as the “plasma clearing factor.” Uncertainty 
remains regarding the physiologic importance of heparin 
activation on this lipase. 
Absorption and Metabolism: Heparin is inactive 
when given orally; it must be administered parenterally. 
It may be given intravenously, intramuscularly or subcu-taneously. 
The biologic half-life is a function of dosage. 
High dosages are associated with relatively long half-lives. 
With standard dosage the half-life is in the range of 
one to two hours. At these dose levels, it is metabolized 
in the liver to a weakly active form which is excreted by 
the kidneys. A discovery of this derivative in urine led to its 
name - uroheparin. Following very large doses of heparin, 
especially when given intravenously, heparin itself does 
not cross the placental barrier nor does it enter the milk 
of lactating women. 
Heparin 
Coumadin® is a registered trademark of E.I. du Pont de Nemours & 
Company, Wilmington, DE 19898
30 
Clinical Uses of Heparin: Heparin is used to pre-vent 
intravascular clotting or to stop further progression 
of intravascular clotting (clot propagation) once it has 
begun. Heparin has the advantage over other antico-agulant 
drugs in that it is immediate in its action. In gen-eral, 
less heparin is required to prevent the initiation of 
clotting (prophylaxis) than to halt active clot formation 
(thrombosis). 
Physicians use heparin for clot prophylaxis in certain high 
risk patients. Among these are those of advanced age, 
with fractures of the leg or hip, with pelvic surgery, with 
congestive heart disease, with cancer, and with acute 
myocardial infarction. Common to this group of patients 
is that they may require long periods of bed rest during 
which time the blood flow in the large veins of the legs 
and pelvis is slowed, increasing the tendency of clot for-mation. 
These clots (thrombi) propagate forming fragile 
extensions which are not firmly bound to the vessel wall. 
Fragments of these clots tend to break off and move in 
the blood stream (embolize). A common and serious 
event is the movement of a large clot fragment (an em-bolus) 
from one of the deep veins of the legs or pelvis to 
the lung. The embolus is caught up in one of the major 
arteries of the lung abruptly stopping the flow of blood to 
that part of the lung. The disorder that follows, pulmonary 
embolization, is a very common cause of death in these 
patients. The use of low dose heparin in preventing deep 
vein thrombosis and high dosages in treating it once it 
has occurred has greatly reduced the mortality rate 
among these high risk patients. 
Heparin is believed to be of value in the treatment of a 
phenomenon known as disseminated intravascular coagu-lation. 
Normally blood does not clot in the blood vessels. 
There are at least two reasons for this. Circulating blood 
does not come in contact with substances which induce 
clotting and whatever activated clotting factors do appear 
are rapidly removed by body cells, particularly those of 
the liver. In many diseases, sufficient clot activation factors 
may enter the blood stream and initiate clotting throughout 
the vascular system. This state is referred to as dissemi-nated 
intravascular coagulation (DIC). DIC has many 
causes. Among these are serious infections, complications 
of pregnancy, malignant disease (cancer), and massive 
physical trauma. The successful treatment of DIC requires 
the control or elimination of the cause but until that 
can be achieved, heparin is useful in preventing further 
intravascular clot formations. 
Because of the quickness of action and the ease of 
neutralization of its anticoagulation effect by the use of 
such substances as protamine sulfate, heparin is used 
to prevent coagulation during the use of extracorporeal 
apparatus such as the heart-lung machine, which is em-ployed 
to maintain aeration and circulation of blood while 
patients are undergoing heart surgery or hemodialysis 
apparatus which are used to clear the blood of unwanted 
metabolites in patients without functioning kidneys. 
In all of these uses, it is important to monitor the plasma in 
order to make appropriate adjustments in heparin dosage. 
Tests Used to Monitor Heparin Treatment: In 
general, heparin treatment is monitored by the measure-ment 
of its effect on coagulation. The first such test was 
the measurement of the length of time required for whole 
blood to clot in a test tube (the Lee White clotting test). 
Today the most commonly used test is the activated partial 
thromboplastin test (APTT). This test is performed on 
citrated plasma in which the clotting factors have been 
activated by a surface contact activator such as kaolin. 
The APTT is the time required for clot formations after 
the plasma has been treated with the activator, and phos-pholipid 
and calcium chloride have been added. The 
APTT is sensitive to small amounts of heparin. Unfortu-nately, 
commercially available thromboplastin preparations 
vary in their sensitivity to heparin. Some are so insensitive 
as to fail to detect therapeutic levels of heparin at all. 
This variation in sensitivity means that each batch of re-agents 
should be tested and standards established. Each 
hospital laboratory, therefore, must determine its own 
therapeutic range. 
An assay system which measures heparin directly and 
depends upon the inhibition of factor Xa by heparin-activated 
antithrombin III has been introduced. In this 
procedure, a synthetic peptide which has a chromogenic 
group attached is employed as a substrate for factor Xa. 
Factor Xa, a serine protease, causes the release of the 
chromogen producing a color change. The assay is sen-sitive 
to very small quantities of heparin but even more 
important, it is reproducible, accurate, and can be stan-dardized 
over several reagent lots and heparin sources.
31 
Physiology: Plasminogen is the precursor of the fibrin-splitting 
enzyme, plasmin. It is a single chain glycoprotein 
with a molecular weight of about 90,000 daltons. It circu-lates 
in normal plasma in concentration of about 20 mg/dl. 
Plasminogen is synthesized by the liver and has a biologic 
half-life of about 2 days. The conversion of inactive plasmi-nogen 
to the active, trypsin-like protease, plasmin is brought 
about by several activators. The most important of these is 
found in the endothelial cells which line blood vessels. 
Clinical Significance: Plasminogen is measured in 
plasma. Its concentration may be increased or decreased. 
In general, only decreases are of clinical importance. 
Increases: Physiologic increases in plasminogen con-centrations 
occur in pregnancy. Certain contraceptives 
may also increase circulating plasminogen levels. In-creases 
can occur in the acute phase of inflammation. 
Decreases: Low plasminogen levels are usually due 
to liver disease with impaired synthesis, or to increased 
utilization associated with DIC (this disorder is discussed 
in detail in the section on fibrinogen). Low levels may 
occur in severe nephrosis due to loss of this relatively 
small protein molecule in the urine. 
Primary fibrinolysis is a rare cause of low plasminogen 
levels. In this disorder abnormally large quantities of plas-minogen 
activators are released into the circulation. There 
are also rare genetic disorders in which individuals syn-thesize 
a defective, inactive plasminogen molecule (dys-functional 
plasminogen). 
Plasminogen
32
Electrolytes 
33
34
35 
Carbon Dioxide 
Physiology: The major end products of the metabolism 
of most foodstuffs are water and carbon dioxide (CO2). 
Approximately 20 moles of CO2 are produced each day 
(the exact amount varies with body size and physical 
activity) and excreted by the lungs. The CO2 formed body 
cells promptly dissolve in water and combine with it to 
form carbonic acid. The latter process is a slow one in a 
simple aqueous solution in a test tube, but in the body it 
is markedly accelerated by the ubiquitous enzyme, car-bonic 
anhydrase. The carbonic acid partially dissociates 
into hydrogen ion (H+) and bicarbonate ion (HCO3). 
These reactions are summarized in the following equations. 
K1 K CO 2 + H2O 
H2CO3 
2 H+ + HCO3 Thus carbon dioxide is transported from the tissues to the 
lungs in three forms: dissolved CO2, undissociated or 
molecular H2CO3, and bicarbonate ion (HCO3). At normal 
plasma pH (7.4), the amounts of each of these are as 
follows: hydrogen ion, 4 x 10–8 M; dissolved CO2, 1 x 10–3 M; 
molecular carbonic acid, 5 x 10–6 M; bicarbonate ion, 
25 x 10–3 M. These relative amounts are determined by 
the equilibrium constants for the equations above. 
The role of bicarbonate ion as the major transport form 
of CO2 is ordinarily of only passing interest to the physician; 
of much greater importance is its role in control of plasma 
pH, which normally lies within the narrow range of 7.35 
to 7.45. The concentration of bicarbonate ion can and 
does vary more with alteration in acid-base balance than 
do chloride, sodium, or potassium ion concentrations. 
The body has control of the excretion of CO2 through 
changes in the rate and depth of respiration. Because 
the various forms of CO2 in plasma are in equilibrium, a 
change in CO2 concentration will result in a concomitant 
change in hydrogen ion and bicarbonate ion concentra-tions. 
The lungs are therefore able to compensate to some 
degree for abnormal production or loss of hydrogen ion. 
The importance of CO2 derives also from that bicarbonate 
ion buffering capacity, that is, its effectiveness in limiting 
changes in pH when acid or base enters the plasma. At 
first glance, the bicarbonate-carbonic acid buffer system 
would not appear to be particularly effective at the usual 
plasma pH of 7.4 since the effective pK1* of this system 
in plasma is 6.1 (buffers work best at pH values close to 
their pK). Buffering is in fact remarkably effective because 
the concentration of carbonic acid can be changed by 
changes in respiration. In brief, the addition of a strong 
acid to plasma (the usual direction of pH change in the 
body) results in excretion of the lungs of enough carbonic 
acid (in the form of CO2 and water) to partially correct the 
fall in pH. At the same time, the bicarbonate-carbonic acid 
buffer system becomes more effective at this lower pH. 
Clinical Significance: Knowledge of the CO2 con-centration 
permits a first approximation of the acid-base 
balance and goes at least part of the way toward eluci-dating 
the cause when an abnormality exists. Full ap-preciation 
of the acid-base status also requires knowledge 
of plasma pH, buffering capacity, hemoglobin concentra-tion, 
pO2 and pCO2. Nevertheless, at the present time, 
the determination of CO2 concentration remains the most 
commonly performed initial test in the evaluation of the 
body’s ability to control pH. 
Decreased Bicarbonate With Elevated pH - 
Respiratory Alkalosis: If the respiratory rate is in-creased, 
there is an increase in excretion of carbon diox-ide, 
and levels of plasma carbonic acid and dissolved CO2 
fall. The plasma pH rises, hence the name respiratory alka-losis. 
The causes of this condition are varied, but they have 
in common the over-stimulation of that part of the brain 
which controls breathing (the “respiratory center”). The most 
common cause is simple anxiety with increased rate and 
depth of breathing - the so called “hyperventilation syn-drome”. 
Other causes include toxic substances (e.g., 
salicylates, as in aspirin overdosage) which stimulate the 
respiratory center, and central nervous system lesions such 
as tumors located in this part of the brain. 
Decreased Bicarbonate With Decreased pH - 
Metabolic Acidosis: The condition usually results 
from the addition of excess amounts of acid to the plasma. 
Usually these acids are products of normal metabolic 
reactions, but they are being formed at a faster rate that 
they can be degraded or excreted. For example, in dia-betic 
coma, increased amounts of acetoacetic acid are 
produced from the oxidation of fatty acids. It accumu-lates 
in the plasma because of the decreased ability to 
further metabolize it via the citric acid cycle. Most of the 
acetoacetic acid is reduced to beta-hydroxybutyric acid. 
Both of these acids are increased in the plasma in dia-betic 
coma. If insulin is not given to correct the metabolic 
abnormalities, coma and death occur. Another example 
of metabolic acidosis is the accumulation of lactic acid 
in the clinical condition called “shock”. The blood pres-sure 
is low and the blood supply to muscles therefore 
decreased. The muscles received insufficient oxygen to 
completely burn glucose; it is metabolized only to the point 
The pK1 value for the net reaction [CO2 + H2O H+ + HCO3 
– ] 
in plasma (rather than in simple aqueous solution) is 6.1.
36 
of formation of pyruvic and then lactic acids. Lactic acid 
accumulates in the plasma. In both diabetic coma and shock 
the bicarbonate level may be very low, and the pH may fall 
below 7.0. Some of the hydrogen ions from these acids 
combine with bicarbonate ion to form carbonic acid and 
then CO2 and water. The CO2 is excreted by the lungs. Thus 
the level of plasma bicarbonate is decreased. There is still 
an excess of hydrogen ion in the plasma, so the pH is low. 
In fact the changes in this condition can be summarized as 
the substitution of a strong acid (lactic or betahydroxybutyric) 
for a weak acid (carbonic). Metabolic acidosis also occurs 
in patients with kidney disease: phosphoric and sulfuric acids 
(produced from the breakdown of phosphate and sulfate-containing 
proteins) can not be removed from the plasma 
at the normal rate because the kidneys are damaged. 
Increase Bicarbonate With Decreased pH-Res-piratory 
Acidosis: The condition usually results from 
diseases of the lungs which impede the excretion of 
carbon dioxide. A typical example is emphysema, a dis-ease 
in which there is widespread destruction of lung 
tissue. Carbon dioxide, carbonic acid, and hydrogen and 
bicarbonate ions accumulate in the plasma. 
Increased Bicarbonate With Elevated pH-Metabolic 
Alkalosis: This condition can result from 
excess intake of sodium bicarbonate (common baking 
soda), a commonly used remedy for abdominal pain such 
as “heartburn” or pain of peptic ulcer. Metabolic alkalosis 
can also result from prolonged vomiting or, in the hospi-talized 
patient, over-use of gastric suction with loss of 
the acid stomach contents. There is a net loss of a strong 
acid, HCl, and a rise in plasma pH. The body attempts to 
replace the lost HCl by decreasing the excretion of CO2 
by the lungs. Retention of CO2 will produce an increase 
in concentrations of hydrogen and bicarbonate ions, thus 
correcting the pH change to some extent. The net effect 
is to replace a strong acid, HCl, with a weak acid, car-bonic 
acid; therefore the pH remains abnormally high.
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book
Biochemistry clinical significance book

More Related Content

Similar to Biochemistry clinical significance book

Poin of care testing
Poin of care testingPoin of care testing
Poin of care testingLAB IDEA
 
Materia Medica of New and Old Homeopathic Medicines (David S. Riley)
Materia Medica of New and Old Homeopathic Medicines (David S. Riley)Materia Medica of New and Old Homeopathic Medicines (David S. Riley)
Materia Medica of New and Old Homeopathic Medicines (David S. Riley)SHHS Online Store & Clinic
 
Astaxanthin The world's best kept health secret Natural astaxanthin.pdf
Astaxanthin The world's best kept health secret Natural astaxanthin.pdfAstaxanthin The world's best kept health secret Natural astaxanthin.pdf
Astaxanthin The world's best kept health secret Natural astaxanthin.pdfVorawut Wongumpornpinit
 
2013 - The impact of coffee on health.pdf
2013 - The impact of coffee on health.pdf2013 - The impact of coffee on health.pdf
2013 - The impact of coffee on health.pdfJonh85
 
The vibration analysis handbook (malestrom)
The vibration analysis handbook   (malestrom)The vibration analysis handbook   (malestrom)
The vibration analysis handbook (malestrom)alfredo20152015
 
G2010ser_OnlineManual_Win_EN_V02.pdf
G2010ser_OnlineManual_Win_EN_V02.pdfG2010ser_OnlineManual_Win_EN_V02.pdf
G2010ser_OnlineManual_Win_EN_V02.pdfHassanJOudah
 
Diagnosis healthy anglais_flipbook_9-04-2014_bd
Diagnosis healthy anglais_flipbook_9-04-2014_bdDiagnosis healthy anglais_flipbook_9-04-2014_bd
Diagnosis healthy anglais_flipbook_9-04-2014_bdnatura medicatrix
 
Advanced Analytical Techniques in Dairy Chemistry.pdf
Advanced Analytical Techniques in Dairy Chemistry.pdfAdvanced Analytical Techniques in Dairy Chemistry.pdf
Advanced Analytical Techniques in Dairy Chemistry.pdfANAYNALLELYLAPON
 
Fat embolism syndrome state of-the-art review focused on
Fat embolism syndrome state of-the-art review focused onFat embolism syndrome state of-the-art review focused on
Fat embolism syndrome state of-the-art review focused oncadoc
 
Absolute Obstetric Anesthesia Review.pdf
Absolute Obstetric Anesthesia Review.pdfAbsolute Obstetric Anesthesia Review.pdf
Absolute Obstetric Anesthesia Review.pdf0r0ym1rra
 
marathi 100 greatest science
marathi 100 greatest science marathi 100 greatest science
marathi 100 greatest science dattatray godase
 
Simply keto-your-essential-21-day-full-plan-with-125-recipes
Simply keto-your-essential-21-day-full-plan-with-125-recipesSimply keto-your-essential-21-day-full-plan-with-125-recipes
Simply keto-your-essential-21-day-full-plan-with-125-recipessamking57
 
Appetizer recipes
Appetizer recipesAppetizer recipes
Appetizer recipesFree lancer
 
Anatomic_Basis_of_Neurologic_Diagnosis.pdf
Anatomic_Basis_of_Neurologic_Diagnosis.pdfAnatomic_Basis_of_Neurologic_Diagnosis.pdf
Anatomic_Basis_of_Neurologic_Diagnosis.pdfDayanaBecerra13
 
Well Teste Interpretation
Well Teste InterpretationWell Teste Interpretation
Well Teste InterpretationMeg Medeiros
 

Similar to Biochemistry clinical significance book (20)

Poin of care testing
Poin of care testingPoin of care testing
Poin of care testing
 
Materia Medica of New and Old Homeopathic Medicines (David S. Riley)
Materia Medica of New and Old Homeopathic Medicines (David S. Riley)Materia Medica of New and Old Homeopathic Medicines (David S. Riley)
Materia Medica of New and Old Homeopathic Medicines (David S. Riley)
 
Astaxanthin The world's best kept health secret Natural astaxanthin.pdf
Astaxanthin The world's best kept health secret Natural astaxanthin.pdfAstaxanthin The world's best kept health secret Natural astaxanthin.pdf
Astaxanthin The world's best kept health secret Natural astaxanthin.pdf
 
Who blood products
Who blood productsWho blood products
Who blood products
 
2013 - The impact of coffee on health.pdf
2013 - The impact of coffee on health.pdf2013 - The impact of coffee on health.pdf
2013 - The impact of coffee on health.pdf
 
The vibration analysis handbook (malestrom)
The vibration analysis handbook   (malestrom)The vibration analysis handbook   (malestrom)
The vibration analysis handbook (malestrom)
 
G2010ser_OnlineManual_Win_EN_V02.pdf
G2010ser_OnlineManual_Win_EN_V02.pdfG2010ser_OnlineManual_Win_EN_V02.pdf
G2010ser_OnlineManual_Win_EN_V02.pdf
 
Diagnosis healthy anglais_flipbook_9-04-2014_bd
Diagnosis healthy anglais_flipbook_9-04-2014_bdDiagnosis healthy anglais_flipbook_9-04-2014_bd
Diagnosis healthy anglais_flipbook_9-04-2014_bd
 
2020 K9 Carnivore product catalog pdf
2020 K9 Carnivore product catalog pdf2020 K9 Carnivore product catalog pdf
2020 K9 Carnivore product catalog pdf
 
Advanced Analytical Techniques in Dairy Chemistry.pdf
Advanced Analytical Techniques in Dairy Chemistry.pdfAdvanced Analytical Techniques in Dairy Chemistry.pdf
Advanced Analytical Techniques in Dairy Chemistry.pdf
 
Fat embolism syndrome state of-the-art review focused on
Fat embolism syndrome state of-the-art review focused onFat embolism syndrome state of-the-art review focused on
Fat embolism syndrome state of-the-art review focused on
 
Absolute Obstetric Anesthesia Review.pdf
Absolute Obstetric Anesthesia Review.pdfAbsolute Obstetric Anesthesia Review.pdf
Absolute Obstetric Anesthesia Review.pdf
 
Biology book1
Biology book1Biology book1
Biology book1
 
marathi 100 greatest science
marathi 100 greatest science marathi 100 greatest science
marathi 100 greatest science
 
Simply keto-your-essential-21-day-full-plan-with-125-recipes
Simply keto-your-essential-21-day-full-plan-with-125-recipesSimply keto-your-essential-21-day-full-plan-with-125-recipes
Simply keto-your-essential-21-day-full-plan-with-125-recipes
 
Appetizer recipes
Appetizer recipesAppetizer recipes
Appetizer recipes
 
Appetizer recipes
Appetizer recipesAppetizer recipes
Appetizer recipes
 
EPILEPSY A public health imperative
EPILEPSY A public health imperativeEPILEPSY A public health imperative
EPILEPSY A public health imperative
 
Anatomic_Basis_of_Neurologic_Diagnosis.pdf
Anatomic_Basis_of_Neurologic_Diagnosis.pdfAnatomic_Basis_of_Neurologic_Diagnosis.pdf
Anatomic_Basis_of_Neurologic_Diagnosis.pdf
 
Well Teste Interpretation
Well Teste InterpretationWell Teste Interpretation
Well Teste Interpretation
 

Recently uploaded

Report Back from SGO: What’s the Latest in Ovarian Cancer?
Report Back from SGO: What’s the Latest in Ovarian Cancer?Report Back from SGO: What’s the Latest in Ovarian Cancer?
Report Back from SGO: What’s the Latest in Ovarian Cancer?bkling
 
Quality control tests of suppository ...
Quality control tests  of suppository ...Quality control tests  of suppository ...
Quality control tests of suppository ...Hasnat Tariq
 
ESC HF 2024 Spotlights Day-2.pptx heart failure
ESC HF 2024 Spotlights Day-2.pptx heart failureESC HF 2024 Spotlights Day-2.pptx heart failure
ESC HF 2024 Spotlights Day-2.pptx heart failuremahiavy26
 
ROSE CASE SPINAL SBRT BY DR KANHU CHARAN PATRO
ROSE  CASE SPINAL SBRT BY DR KANHU CHARAN PATROROSE  CASE SPINAL SBRT BY DR KANHU CHARAN PATRO
ROSE CASE SPINAL SBRT BY DR KANHU CHARAN PATROKanhu Charan
 
Histology of Epithelium - Dr Muhammad Ali Rabbani - Medicose Academics
Histology of Epithelium - Dr Muhammad Ali Rabbani - Medicose AcademicsHistology of Epithelium - Dr Muhammad Ali Rabbani - Medicose Academics
Histology of Epithelium - Dr Muhammad Ali Rabbani - Medicose AcademicsMedicoseAcademics
 
TEST BANK For Huether and McCance's Understanding Pathophysiology, Canadian 2...
TEST BANK For Huether and McCance's Understanding Pathophysiology, Canadian 2...TEST BANK For Huether and McCance's Understanding Pathophysiology, Canadian 2...
TEST BANK For Huether and McCance's Understanding Pathophysiology, Canadian 2...marcuskenyatta275
 
^In Pietermaritzburg Hager Werken Embalming +27789155305 Compound Powder in ...
^In Pietermaritzburg  Hager Werken Embalming +27789155305 Compound Powder in ...^In Pietermaritzburg  Hager Werken Embalming +27789155305 Compound Powder in ...
^In Pietermaritzburg Hager Werken Embalming +27789155305 Compound Powder in ...pinkpowder997723
 
Unlocking Holistic Wellness: Addressing Depression, Mental Well-Being, and St...
Unlocking Holistic Wellness: Addressing Depression, Mental Well-Being, and St...Unlocking Holistic Wellness: Addressing Depression, Mental Well-Being, and St...
Unlocking Holistic Wellness: Addressing Depression, Mental Well-Being, and St...Health Kinesiology Natural Bioenergetics
 
Sonia Journal club presentation (2).pptx
Sonia Journal club presentation (2).pptxSonia Journal club presentation (2).pptx
Sonia Journal club presentation (2).pptxpalsonia139
 
Signs It’s Time for Physiotherapy Sessions Prioritizing Wellness
Signs It’s Time for Physiotherapy Sessions Prioritizing WellnessSigns It’s Time for Physiotherapy Sessions Prioritizing Wellness
Signs It’s Time for Physiotherapy Sessions Prioritizing WellnessGokuldas Hospital
 
Hemodialysis: Chapter 1, Physiological Principles of Hemodialysis - Dr.Gawad
Hemodialysis: Chapter 1, Physiological Principles of Hemodialysis - Dr.GawadHemodialysis: Chapter 1, Physiological Principles of Hemodialysis - Dr.Gawad
Hemodialysis: Chapter 1, Physiological Principles of Hemodialysis - Dr.GawadNephroTube - Dr.Gawad
 
VIII.1 Nursing Interventions to Promote Healthy Psychological responses, SELF...
VIII.1 Nursing Interventions to Promote Healthy Psychological responses, SELF...VIII.1 Nursing Interventions to Promote Healthy Psychological responses, SELF...
VIII.1 Nursing Interventions to Promote Healthy Psychological responses, SELF...JRRolfNeuqelet
 
Mgr university bsc nursing adult health previous question paper with answers
Mgr university  bsc nursing adult health previous question paper with answersMgr university  bsc nursing adult health previous question paper with answers
Mgr university bsc nursing adult health previous question paper with answersShafnaP5
 
Sell pmk powder cas 28578-16-7 from pmk supplier Telegram +85297504341
Sell pmk powder cas 28578-16-7 from pmk supplier Telegram +85297504341Sell pmk powder cas 28578-16-7 from pmk supplier Telegram +85297504341
Sell pmk powder cas 28578-16-7 from pmk supplier Telegram +85297504341Sherrylee83
 
The Orbit & its contents by Dr. Rabia I. Gandapore.pptx
The Orbit & its contents by Dr. Rabia I. Gandapore.pptxThe Orbit & its contents by Dr. Rabia I. Gandapore.pptx
The Orbit & its contents by Dr. Rabia I. Gandapore.pptxDr. Rabia Inam Gandapore
 
DR. Neha Mehta Best Psychologist.in India
DR. Neha Mehta Best Psychologist.in IndiaDR. Neha Mehta Best Psychologist.in India
DR. Neha Mehta Best Psychologist.in IndiaNehamehta128467
 
Overview on the Automatic pill identifier
Overview on the Automatic pill identifierOverview on the Automatic pill identifier
Overview on the Automatic pill identifierNidhi Joshi
 
VVIP Hadapsar ℂall Girls 6350482085 Scorching { Pune } Excellent Girl Serviℂe...
VVIP Hadapsar ℂall Girls 6350482085 Scorching { Pune } Excellent Girl Serviℂe...VVIP Hadapsar ℂall Girls 6350482085 Scorching { Pune } Excellent Girl Serviℂe...
VVIP Hadapsar ℂall Girls 6350482085 Scorching { Pune } Excellent Girl Serviℂe...dhyaansingh0898#S07
 
Tips to Choose the Best Psychiatrists in Indore
Tips to Choose the Best Psychiatrists in IndoreTips to Choose the Best Psychiatrists in Indore
Tips to Choose the Best Psychiatrists in IndoreGokuldas Hospital
 

Recently uploaded (20)

Report Back from SGO: What’s the Latest in Ovarian Cancer?
Report Back from SGO: What’s the Latest in Ovarian Cancer?Report Back from SGO: What’s the Latest in Ovarian Cancer?
Report Back from SGO: What’s the Latest in Ovarian Cancer?
 
Quality control tests of suppository ...
Quality control tests  of suppository ...Quality control tests  of suppository ...
Quality control tests of suppository ...
 
ESC HF 2024 Spotlights Day-2.pptx heart failure
ESC HF 2024 Spotlights Day-2.pptx heart failureESC HF 2024 Spotlights Day-2.pptx heart failure
ESC HF 2024 Spotlights Day-2.pptx heart failure
 
ROSE CASE SPINAL SBRT BY DR KANHU CHARAN PATRO
ROSE  CASE SPINAL SBRT BY DR KANHU CHARAN PATROROSE  CASE SPINAL SBRT BY DR KANHU CHARAN PATRO
ROSE CASE SPINAL SBRT BY DR KANHU CHARAN PATRO
 
Histology of Epithelium - Dr Muhammad Ali Rabbani - Medicose Academics
Histology of Epithelium - Dr Muhammad Ali Rabbani - Medicose AcademicsHistology of Epithelium - Dr Muhammad Ali Rabbani - Medicose Academics
Histology of Epithelium - Dr Muhammad Ali Rabbani - Medicose Academics
 
TEST BANK For Huether and McCance's Understanding Pathophysiology, Canadian 2...
TEST BANK For Huether and McCance's Understanding Pathophysiology, Canadian 2...TEST BANK For Huether and McCance's Understanding Pathophysiology, Canadian 2...
TEST BANK For Huether and McCance's Understanding Pathophysiology, Canadian 2...
 
Best medicine 100% Effective&Safe Mifepristion ௵+918133066128௹Abortion pills ...
Best medicine 100% Effective&Safe Mifepristion ௵+918133066128௹Abortion pills ...Best medicine 100% Effective&Safe Mifepristion ௵+918133066128௹Abortion pills ...
Best medicine 100% Effective&Safe Mifepristion ௵+918133066128௹Abortion pills ...
 
^In Pietermaritzburg Hager Werken Embalming +27789155305 Compound Powder in ...
^In Pietermaritzburg  Hager Werken Embalming +27789155305 Compound Powder in ...^In Pietermaritzburg  Hager Werken Embalming +27789155305 Compound Powder in ...
^In Pietermaritzburg Hager Werken Embalming +27789155305 Compound Powder in ...
 
Unlocking Holistic Wellness: Addressing Depression, Mental Well-Being, and St...
Unlocking Holistic Wellness: Addressing Depression, Mental Well-Being, and St...Unlocking Holistic Wellness: Addressing Depression, Mental Well-Being, and St...
Unlocking Holistic Wellness: Addressing Depression, Mental Well-Being, and St...
 
Sonia Journal club presentation (2).pptx
Sonia Journal club presentation (2).pptxSonia Journal club presentation (2).pptx
Sonia Journal club presentation (2).pptx
 
Signs It’s Time for Physiotherapy Sessions Prioritizing Wellness
Signs It’s Time for Physiotherapy Sessions Prioritizing WellnessSigns It’s Time for Physiotherapy Sessions Prioritizing Wellness
Signs It’s Time for Physiotherapy Sessions Prioritizing Wellness
 
Hemodialysis: Chapter 1, Physiological Principles of Hemodialysis - Dr.Gawad
Hemodialysis: Chapter 1, Physiological Principles of Hemodialysis - Dr.GawadHemodialysis: Chapter 1, Physiological Principles of Hemodialysis - Dr.Gawad
Hemodialysis: Chapter 1, Physiological Principles of Hemodialysis - Dr.Gawad
 
VIII.1 Nursing Interventions to Promote Healthy Psychological responses, SELF...
VIII.1 Nursing Interventions to Promote Healthy Psychological responses, SELF...VIII.1 Nursing Interventions to Promote Healthy Psychological responses, SELF...
VIII.1 Nursing Interventions to Promote Healthy Psychological responses, SELF...
 
Mgr university bsc nursing adult health previous question paper with answers
Mgr university  bsc nursing adult health previous question paper with answersMgr university  bsc nursing adult health previous question paper with answers
Mgr university bsc nursing adult health previous question paper with answers
 
Sell pmk powder cas 28578-16-7 from pmk supplier Telegram +85297504341
Sell pmk powder cas 28578-16-7 from pmk supplier Telegram +85297504341Sell pmk powder cas 28578-16-7 from pmk supplier Telegram +85297504341
Sell pmk powder cas 28578-16-7 from pmk supplier Telegram +85297504341
 
The Orbit & its contents by Dr. Rabia I. Gandapore.pptx
The Orbit & its contents by Dr. Rabia I. Gandapore.pptxThe Orbit & its contents by Dr. Rabia I. Gandapore.pptx
The Orbit & its contents by Dr. Rabia I. Gandapore.pptx
 
DR. Neha Mehta Best Psychologist.in India
DR. Neha Mehta Best Psychologist.in IndiaDR. Neha Mehta Best Psychologist.in India
DR. Neha Mehta Best Psychologist.in India
 
Overview on the Automatic pill identifier
Overview on the Automatic pill identifierOverview on the Automatic pill identifier
Overview on the Automatic pill identifier
 
VVIP Hadapsar ℂall Girls 6350482085 Scorching { Pune } Excellent Girl Serviℂe...
VVIP Hadapsar ℂall Girls 6350482085 Scorching { Pune } Excellent Girl Serviℂe...VVIP Hadapsar ℂall Girls 6350482085 Scorching { Pune } Excellent Girl Serviℂe...
VVIP Hadapsar ℂall Girls 6350482085 Scorching { Pune } Excellent Girl Serviℂe...
 
Tips to Choose the Best Psychiatrists in Indore
Tips to Choose the Best Psychiatrists in IndoreTips to Choose the Best Psychiatrists in Indore
Tips to Choose the Best Psychiatrists in Indore
 

Biochemistry clinical significance book

  • 1. aca® discrete clinical analyzer Dimension® clinical chemistry system OPUS™/OPUS™ PLUS/ OPUS Magnum® Immunoassay System Stratus® CS STAT Fluorometric Analyzer 1 Clinical Significance
  • 2. 2 Here’s How to Order Additional Copies Copies of “Clinical Significance” may be purchased for $20 each, including shipping and handling charges. To order, please write to us on your letterhead, specifying the num-ber of copies, your ship-to address and phone number. Mail to: Dade Behring Inc. Glasgow Business Community Mailbox 531 P.O. Box 6101 Newark, Delaware 19714 U.S.A. Enclose a check or money order in the amount for the total number of copies ordered and make payable to Dade Behring.
  • 3. 3 Table of Contents Page Introduction .........................................................................5 Cancer Markers Carcinoembryonic Antigen ...................................................9 Prostate Specific Antigen .................................................. 10 Cardiac Markers Creatine Kinase-MB .......................................................... 15 Myoglobin ......................................................................... 16 Troponin-I ......................................................................... 18 Coagulation General Information .......................................................... 23 Antithrombin III ................................................................ 26 Fibrin Degradation Products ............................................. 27 Fibrinogen ......................................................................... 28 Heparin ............................................................................. 29 Plasminogen ..................................................................... 31 Electrolytes Carbon Dioxide ................................................................. 35 Chloride ............................................................................ 37 Potassium ......................................................................... 38 Sodium ............................................................................. 40 Endocrinology Thyroid Function Tests: ..................................................... 45 Free Thyroxine Index ...................................................... 45 Thyroxine ........................................................................ 45 Thyronine Uptake ........................................................... 45 Thyroid Stimulating Hormone ........................................ 47 Enzymes Acid Phosphatase ............................................................. 53 Alanine Aminotransferase ................................................. 54 Alkaline Phosphatase ........................................................ 55 Amylase ............................................................................ 57 Aspartate Aminotransferase .............................................. 58 Total Creatine Kinase ........................................................ 60 g-Glutamyl Transferase ..................................................... 61 Lactic Dehydrogenase ....................................................... 63 Lactate Dehydrogenase Isoenzyme 1 ............................... 63 Liver Lactic Dehydrogenase .............................................. 63 Lipase ............................................................................... 65 Pseudocholinesterase ....................................................... 66 Fertility Hormones Follitropin .......................................................................... 69 Human Chorionic Gonadotropin (Choriogonadotropin) .... 72 Luteinizing Hormone ......................................................... 76 Prolactin ............................................................................ 80 Page General Chemistry Bilirubin (Conjugated and Total) ....................................... 85 Calcium ............................................................................. 87 Cerebrospinal Fluid Protein ............................................... 89 Cholesterol ........................................................................ 90 Creatinine .......................................................................... 92 Glucose ............................................................................. 93 High Density Lipoprotein Cholesterol ............................... 96 Iron ................................................................................... 98 Magnesium ..................................................................... 100 Neonatal Bilirubin ........................................................... 101 Phosphorus .................................................................... 103 Total Protein and Albumin ............................................... 105 Triglycerides .................................................................... 107 Urea (Blood Urea Nitrogen) ............................................ 109 Urinary Protein ............................................................... 110 Uric Acid ......................................................................... 113 Immunology/Serology General Information ........................................................ 117 Hypergammaglobulinemias ............................................ 119 Hypogammaglobulinemias (Decreases in Immunoglobulins) ... 120 C-Reactive Protein .......................................................... 122 Therapeutic Drug Monitoring Aminoglycoside Antibiotics ............................................ 127 Amikacin ....................................................................... 127 Gentamicin ................................................................... 127 Tobramycin ................................................................... 127 Antiarrhythmic Drugs ..................................................... 129 Digitoxin ....................................................................... 129 Digoxin ......................................................................... 129 Lidocaine ...................................................................... 130 N-Acetylprocainamide .................................................. 130 Procainamide................................................................ 130 Quinidine ...................................................................... 131 Antiasthmatic Drugs ....................................................... 132 Theophylline ................................................................. 132 Anti-convulsant Drugs: ................................................... 134 Carbamazepine ............................................................. 134 Ethosuximide ................................................................ 135 Phenobarbital ............................................................... 135 Primidone ..................................................................... 136 Phenytoin ..................................................................... 137 Valproic Acid ................................................................ 138 Chemotherapeutic Drug .................................................. 139 Methotrexate ................................................................ 139 Vancomycin .................................................................... 141
  • 4. 4 Table of Contents (cont'd) Page Toxicology Acetaminophen ............................................................... 145 Ethanol ("alcohol," ethyl alcohol) .................................... 146 Drug Screen Tests: .......................................................... 148 Barbiturate Screen ........................................................ 148 Benzodiazepine Screen ................................................. 148 Tricyclic Antidepressant Screen ................................... 148 Salicylate ......................................................................... 150 Urine Drugs of Abuse Screen Tests Introduction ............ 151 Urine Amphetamines Screen .......................................... 152 Urine Barbiturates Screen ............................................... 153 Urine Benzodiazepines Screen ........................................ 155 Urine Cannabinoids Screen ............................................. 156 Urine Cocaine Metabolite Screen .................................... 158 Urine Methadone Screen ................................................ 159 Urine Opiates Screen ...................................................... 160 Urine Phencyclidine Screen ............................................ 161 Specialty Ammonia ........................................................................ 165 b2 Microglobulin ............................................................ 167 Ferritin ............................................................................ 168 Lactic Acid ...................................................................... 170 Urinary Albumin for the detection of microalbuminuria ................ 171
  • 5. 5 Introduction This collection of papers is intended to accompany the technical descriptions of the procedures performed by the Dade Behring aca® discrete clinical analyzer, Dimension® clinical chemistry system, OPUS™/OPUS™ Plus/OPUS Magnum® Immunoassay System and the Stratus® CS STAT Fluorometric Analyzer. The purpose is to explain in a very general way why the physician is interested in the substances measured. The approach has been to de-scribe briefly the normal physiology of the substance and then to list some of the diseases in which abnormal val-ues might be expected to occur. These are by no means complete lists; only the most common diseases have been included. Those wishing to know more about the clinical relevance of these tests are referred to any one of the standard textbooks of clinical pathology. Some comments may be useful relative to the nature of the specimen examined. Most of the tests are done on serum. Blood, serum, and plasma are words which are sometimes used interchangeably, yet they refer in fact to different substances. Blood needs no explanation except to say that it ordinarily refers to whole blood drawn from the veins, that is, venous blood. There are differences between arterial, capillary, and venous blood, but none of these are of much importance with respect to the tests now performed. Plasma is the fluid part of the blood. It is obtained by removing the cellular components (red cells, white cells, and platelets) from whole blood by centrifugation. In order to prevent clotting, an anticoagulant is added when the blood is drawn. Plasma must be used when coagulation factors are assayed, but it may also be used for the usual chemistry procedures when a very rapid answer is re-quired. For this purpose, heparin may be used as the anticoagulant. Serum is used in the measurement of most chemical substances in blood. It is the fluid part of the blood which remains after whole blood has been allowed to clot. Except for the absence in serum of some of the coagulation fac-tors, the differences between serum and plasma are of little consequence. Serum has the advantage of remaining fluid while plasma tends to clot eventually. Many of the substances measured are referred to erro-neously as though they were measured in a whole blood sample when in fact it is serum (or plasma) which is as-sayed. Thus physicians (and clinical chemists) refer to “blood sugar”, “blood urea nitrogen”, etc., when the more accurate reference would be to serum glucose or serum urea nitrogen. Frank C. Larson, M.D. Myrna Traver, MD Professor Emeritus Pathologist Department of Medicine Madison, WI Medical School University of Wisconsin-Madison
  • 6. 6
  • 8. 8
  • 9. Carcinoembryonic Antigen 9 Structure and Function: CEA is one of approxi-mately 20 related complex glycoproteins that are mem-bers of the immunoglobulin superfamily1. It’s molecular mass varies between 150 and 300 kd2. The alteration in molecular mass is due to extensive, but differing glycosylation of the individual CEA molecules. CEA is one of a number of proteins that are expressed during normal differentiation of fetal tissue but are partially or completely suppressed in the adult3. It resides on the surface of epithelial cells of the gastrointestinal tract and a variety of other tissues and functions to mediate inter-cellular adhesion. In the normal adult, CEA is expressed in only trace amounts and is attached to the cell surface by way of a GPI (glycosylphosphatidylinositol) anchor4. In cancerous states, surface CEA can be shed in larger amounts and detected in the circulation and other body fluids. No clear picture has emerged regarding the role of CEA in cancer, but several studies suggest that over expression in carcinomas interferes with cell differentiation. Clinical Application: CEA is the most commonly used tumor marker for gastrointestinal tract tumors. Once thought to be specific for bowel cancer, elevated levels are also detected in association with a number of other cancers such as colorectal (70%), lung (45%), gastric (50%), breast (40%), pancreatic (55%), ovarian (25%), and uterine (40%). However, an abnormal serum level is not specific for cancer or unique to malignancy. CEA lev-els may also be elevated in some patients with benign conditions such as cirrhosis (45%), pulmonary emphy-sema (30%), rectal polyps (5%), benign breast disease (15%), and ulcerative colitis (15%)2. Up to 19% of smok-ers with no evidence of malignancy and 3% of a healthy population may also demonstrate elevated CEA levels1. Expected normal range values for nonsmokers are < 3.0 ng/ml and < 5.0 ng/ml for smokers. Age, sex, and race have no significant effect on serum CEA levels. Caution should be exercised when interpreting patient CEA values. Quantitation is method dependent and there-fore values should only be compared when using the same method. Measurement of CEA is not recommended as a screening tool given that false-positives arise from non-cancerous conditions and false negatives may oc-cur since many tumors do not produce CEA. It’s quantitation can be used as an aid in the prognosis and management of cancer patients in whom changing con-centrations of CEA are observed. The American Society of Clinical Oncology (ASCO) recommends the use of CEA in colorectal cancer for preoperative evaluation, postoperative follow-up, and monitoring of treatment for metastatic disease.5 Preoperative Evaluation: CEA may be ordered pre-operatively in patients with colorectal carcinoma if it would assist in staging and surgical treatment planning. Al-though elevated preoperative CEA (>5 ng/ml) may cor-relate with poorer prognosis, data are insufficient to sup-port the use of CEA to determine whether to treat a pa-tient with adjunct therapy.5 Postoperative Follow-up: If resection of liver me-tastases would be clinically indicated, it is recommended that postoperative serum CEA testing may be performed every 2 to 3 months in patients with stage II or III dis-ease for 2 or more years after diagnosis. An elevated CEA, if confirmed by retesting, warrants further evalua-tion for metastatic disease, but does not justify the insti-tution of adjunct therapy or systemic therapy for presumed metastatic disease.5 Monitoring During Treatment for Metastatic Disease: Present data are sufficient to recommend rou-tine use of the serum CEA alone for monitoring response to treatment. If no other test is available to indicate a response, CEA should be measured at the start of treat-ment for metastatic disease, and every 2 to 3 months during active treatment. Two values above baseline are adequate to document progressive disease, even in the absence of corroborating radiographs. CEA is regarded as the marker of choice for monitoring colorectal cancer.5 References: 1. E. P. Mitchell: Role of carcinoembryonic antigen in the manage-ment of advanced colorectal cancer. Seminars in Oncol-ogy. 25:12-20, 1998. 2. D. W. Chan and S. Sell. Tumor Markers. In: C.A. Burtis and E.R. Ashwood, eds. Textbook of Clinical Chemistry. Philadelphia: W.B. Saunders, 1994:897-927. 3. J. Mendelsohn: Principles of Neoplasia. In: E. Braunwald, et al, eds. Principles of Internal Medicine. New York: McGraw-Hill, 1987:412-431. 4. B. Obrink: CEA adhesion molecules: multifunctional proteins with signal-regulatory properties. Current Opinion in Cell Biology. 9:616-626, 1997. 5. 1997 Update of recommendations for the use of tumor markers in breast and colorectal cancer. J. Clinical Oncology. 16:793-795, 1998.
  • 10. Prostate Specific Antigen 10 Structure and Function Prostate specific antigen (PSA) is a single chain protein consisting of 237 amino acids and one N-linked carbo-hydrate chain. The molecular weight of 28.5 kDa includes the 7% carbohydrate content. PSA is a serine protease member of the kallikrein family and shares amino acid sequence homology with at least two other family mem-bers, human glandular kallikrein and pancreatic renal kallikrein. PSA is produced mainly by the epithelial and ductal cells of the prostate gland, but is also present in the urethra, periurethral and anal glands of the male. Low level PSA production has also been observed in the female salivary glands and breast. PSA is a normal con-stituent of seminal fluid where its role is liquefaction of the coagulum to increase sperm motility. PSA may also function as a growth factor modulator via proteolytic cleavage of growth factor, as is the case with pro-EGF (epidermal growth factor) or the transport protein for the growth factor as is the case for the binding protein-3 for insulin-like growth factor. PTH-rp (parathyroid hormone related peptide) has also been shown to be a substrate for PSA suggesting a role for PSA in intracellular cal-cium regulation. The serum PSA test has become an important aid in the clinical management of patients with prostate cancer. Serum PSA Molecular Forms: In serum, PSA circulates as a “com-plex” with proteolytic enzyme inhibitors and also in a “free” form. The majority of measurable PSA consists of PSA complexed in a 1:1 ratio with a-1 antichymotrypsin (ACT). Smaller amounts of PSA are bound to a-1 antitrypsin and protein C inhibitor. PSA complexed with a-2 macro-globulin (2:1) ratio is not measurable with conventional immunoassays, as the PSA is engulfed by the larger a-2 macroglobulin. In some healthy subjects, the free form of PSA in serum may constitute a significant frac-tion of the total PSA (up to 40% for PSA values in the range of 1.0 - 4.0 ng/mL). The free form may consist of the proenzyme that is inappropriately released by the prostate cell, or “nicked” PSA which is enzymatically in-activated as a result of peptide bond cleavage at the lysine-lysine 145-146 position and possibly at other sites as well. The complexed form of PSA is cleared by the liver with an apparent half-life of 2.7 days, while free PSA has a half-life of approximately 1.5 hours. Expected Ranges for Total Serum PSA: Normal values for serum PSA are age related, reflecting the effects of increasing prostate size as a man ages. The age-referenced upper limits of normal (95th percentile) for Caucasian males are: 40-49 years, 2.5 ng/mL; 50-59 years, 3.5 ng/mL; 60-69 years, 4.5 ng/mL and 70-79, 6.5 ng/mL. Asian men may have lower PSA values and African American men may have higher values than those given for Caucasian males. The value of 4.0 ng/mL has become the established cut-off value for diagnostic purposes. There have been no reports of significant diurnal or circadian variation for serum PSA, but some men may demonstrate significant biological variation, showing coefficients of variation as high as 50% for mean PSA concentrations below 4.0 ng/mL. Pre-Analytical Considerations: In some men, se-rum PSA values may be increased by prostatic massage as occurs with the digital rectal exam (DRE) or transrectal ultrasonography (TRUS). Thus, it is recommended that serum PSA values be determined prior to any of these activities in order to minimize false positive PSA results. Drug therapy with finasteride for benign prostatic hyper-trophic (BPH) disease may cause serum PSA values to decrease. Biopsy or surgery of the prostate will cause significant false elevation of serum PSA values and it is recommended that PSA measurements be delayed for at least six weeks after these procedures are performed. Human glandular kallikrein (HgK) is a potential source of false positive error in PSA immunoassays due to its high homology with PSA. However, careful selection of immunoreagents and structural differences between PSA and HgK appears to have minimized this potential problem. Non-prostatic sources of PSA in males are secreted into the urine tract and do not affect serum measurements. Clinical Application Patient Monitoring: Patients with cancer confined to the prostate are usually treated by prostatectomy or ra-diation, or may be offered “watchful waiting.” The “watch-ful waiting” approach consists of close monitoring to as-certain the rate of tumor growth. It is considered by many to be most useful for older men who have less aggres-sive tumors, that is, tumors with small volumes (less than 1.0 cc) of low Gleason grade.
  • 11. 11 After radical prostatectomy, serum PSA values should decline to values of less than 0.2 ng/mL after 2-3 days. In many urologic practices, post-surgical PSA values which rise above 0.5 ng/mL are strongly suggestive of recumbinant tumor or metastasis and will signal additional work-up or treatment. The pattern of PSA change for patients undergoing ra-diation therapy reflects the slower elimination of PSA producing tumor cells compared to the more immediate and complete removal of the tumor by surgery. Serum PSA values may decrease for 6-12 months before stabi-lization. The level to which the PSA must drop to reflect effective radiation therapy is controversial. However, it is apparent that the probability of relapse decreases with the extent to which the PSA concentration drops. The lowest relapse rate for radiation treated patients has been achieved when the serum PSA value drops to levels below 0.2 ng/mL as demonstrated for patients treated by radical prostatectomy. Rising PSA values in patients treated by radiation therapy signals tumor recurrence and progres-sion of disease. It is hoped that the early treatment of these localized cancers will improve the overall cure-rate for pros-tate cancer. Hormonal treatments are commonly used to treat advanced prostate cancer. The primary strategy is to reduce the con-centration of testosterone by surgical castration (bilateral orchiectomy), estrogen therapy, or androgen blockage with the use of an LH-RH analogue and an anti-androgen. The response to hormonal therapy and the resulting decline in serum PSA concentration can be dramatic. However, the response to hormone therapy as well as the PSA decline is related to the composition of the tumor which consists of both hormonally sensitive and insensitive cell clones. Ulti-mately, the tumor will progress with the proliferation of the androgen resistant clone. Serum PSA may not increase to reflect the tumor burden in some of these cases, as the hormonally insensitive clone is less well-differentiated and may lack the ability to produce PSA to the same extent as hormonally sensitive clones. Staging and Prognosis: In routine practice, serum PSA provides little discrimination between patients with localized disease and those with extracapsular spread of cancer. While serum PSA varies directly with tumor volume, there is wide variation of PSA values and also considerable overlap of PSA ranges for the various dis-ease stages. Approximately 80% of prostate cancer pa-tients have an elevated PSA value defined by the cut-off value of 4.0 ng/mL. The majority of patients with PSA values of less than 4.0 ng/mL will have disease confined to the prostate, and most patients with PSA values greater than 10.0 ng/mL will have metastatic disease to lymph nodes or bone. Bone scans are used to confirm metasta-sis, however, bone scans may not be necessary in the routine workup of patients who present with no skeletal symptoms and have PSA values of less than 20.0 ng/ mL. Since serum PSA cannot accurately identify patients who have disease confined to the prostate, pretreatment serum PSA measurement provides no prognostic infor-mation for the individual patient. Other Prostate Diseases: The primary cause of false positive PSA values is BPH disease. While PSA production in BPH tissue is estimated to be only one-tenth that of prostate cancer tissue, the larger volume of BPH tissue leads to serum PSA values greater than 4.0 ng/mL in 20-30% of patients. Most of these elevated BPH values are in the range of 4.0-10.0 ng/mL. Acute urinary retention, prostatic infarction and prostatitis may also cause elevated serum PSA values. Conclusion In the management of prostatic carcinoma, serum PSA values provide accurate assessment of the patient’s re-sponse to therapy. Prepared by: Herbert A. Fritsche, Ph.D. M. D. Anderson Cancer Center Houston TX
  • 12. 12
  • 14. 14
  • 15. Creatine Kinase MB (CK Isoenzyme) 15 Physiology: Creatine phosphate and creatine kinase, the enzyme which synthesizes and degrades it, are important compounds in the storage and transfer of chemical energy within cells. When food from the diet is oxidized, its poten-tial energy is trapped as a high-energy compound which may be utilized immediately or stored in the cell until it is needed to perform mechanical work or to drive some en-ergy- requiring metabolic reaction. The most common of the “energy transfer” compounds is adenosine triphosphate, or ATP. For example, the energy for muscle contraction (and for many other metabolic reactions) is supplied by the hy-drolysis of the terminal pyrophosphate bond of ATP to yield adenosine diphosphate (ADP) and inorganic phosphate ion. ATP is normally produced by the oxidation of foods through a long series of reactions - a relatively slow process. There is only a small amount of ATP in muscle cells - about 6 mmol/g. This is sufficient to sustain muscle contraction for only a short period of time. Some additional ATP can be produced fairly quickly by the partial degradation of glu-cose liberated by hydrolysis of stored muscle glycogen. However, at the onset of a sudden burst of intense muscle contraction, muscle cells require some form of “stored” energy which can be quickly converted to ATP. The high-energy compound which serves to “store” energy in the muscle cell is creatine phosphate (also called phosphocre-atine) which has a high-energy guanidophosphate bond. ATP can be quickly resynthesized by the one-step trans-fer of the phosphate group of creatine phosphate to ADP. This reaction is catalyzed by the enzyme creatine kinase (CK - previously known as creatine phosphokinase, or CPK). All body cells probably contain some CK, but the largest amounts are found in tissues in which a large amount of energy is stored as creatine phosphate. Skeletal muscle contains 2000-3000 units of this enzyme per gram of tissue, heart muscle 400 U/g, and the smooth muscle of the intestinal wall about 150 U/g. In contrast, there are only 3 U/g in liver. As is true of many other enzymes, several molecular forms (isoenzymes) of CK exist. They differ in their Michaelis constants and pH optima, but the physiologi-cal significance of this is unknown. Each of the three isoenzymes of CK is composed of two polypeptide chains. In the case of skeletal muscle CK, the chains are identical and are called M chains. The enzyme is called type MM. Brain CK is also composed of two identical chains, but these are different from the chains in muscle CK. Brain CK is type BB, composed of two type B chains. The third isoenzyme is a hybrid molecule, called type MB, consisting of one M chain and one B chain. Heart muscle, although it contains primarily type MM CK, also contains some type MB. This is of major clinical significance since CKMB is usually found only in heart muscle. Clinical Significance: Normal serum contains a small amount of CKMM and practically no CKBB or CKMB. When any tissue is damaged some of the intracellular enzymes may leak into the blood. Thus injury to skeletal muscle or heart muscle may be followed by a rise in serum CK. Measurement of serum total CK has been widely used in the diagnosis of acute myocardial infarction (AMI).* This disease is the most common cause of death in the western world. A portion of the heart muscle is de-prived of its blood supply, usually because of occlusions of a coronary artery by fatty plaques and/or by thrombus formation. The heart muscle cells die, and CK is released into the blood along with other enzymes such as AST and LDH. Serum CK levels correlate with the volume of damaged heart muscle and have been used as an index of the extent of the infarction. The usefulness of serum to-tal CK in MI has been compromised by the fact that some injury to skeletal muscle may accompany AMI, and skel-etal muscle contains 10 times as much CK as does heart muscle. For example, injections of pain medication into muscle (a common event in the treatment of a patient with suspected AMI) may cause enough inflammation of the muscle to give a spurious elevation in total serum CK. This problem can be circumvented by measuring only serum CKMB. This is a remarkable, sensitive and spe-cific test for heart muscle damage. As with total CK, in-creases in serum CKMB can usually be seen at 4 hours after the onset of chest pain. The enzyme level peaks soon after and returns to normal within 72 hours. *CK is also used in the diagnosis of some diseases of muscle and brain. These are discussed in the section on CK.
  • 16. 16 Physiology Structure and Function: Myoglobin is a 17,800 dalton porphyrin-containing protein consisting of 153 amino acids. Myoglobin is almost exclusively found in the cytoplasm of skeletal and cardiac striated muscle. It constitutes 2% of the total muscle protein, and there are approximately 2.8 mg of myoglobin per gram of heart tissue. No organ-specific isoforms of myoglobin have been demonstrated in humans thus far. One of the primary roles of myoglobin in muscle tissue is to bind oxygen and release it when the partial pressure (pO2) within the cells becomes very low. Clinical Significance Myoglobin Release from Cells: Clinical studies suggest that myoglobin, owing to its small size and cytoplasmic location, is one of the first tissue specific markers to be released from the compromised skeletal or cardiac myocyte. It is this characteristic on which its clinical sig-nificance rests. Serum and plasma levels of myoglobin increase within one hour of the onset of chest pain in acute myocardial infarction (AMI), thus most clinical labo-ratories today measure myoglobin levels. Leakage by Molecular Size When the myocyte experiences ischemia, one of the first responses is swelling. This swelling results in a sequen-tial loss of cytoplasmic constituents into the interstitial spaces in a manner that is roughly dependent on the molecular weight of each constituent. In this leakage model, very small molecular weight molecules like elec-trolytes are lost first. Then the macromolecules of increas-ing size like myoglobin (17,800 daltons) and enzymes like creatine kinase and its MB isoenzyme (CKMB) at 86,000 daltons are lost from the cytoplasm. Myoglobin at 17,800 daltons is lost from the myocyte cytoplasm before cell death in ischemic conditions such as unstable angina; thus, circulating levels of myoglobin have been found to increase in ischemia. In addition, myoglobin seems to move directly to the coronary blood flow, while CKMB and lactate dehydrogenase isoenzyme 1 (LD1) are likely carried by lymphatic drainage from the injured myocardium. Myoglobin in Acute Myocardial Infarction It has been known since the first reports by Kagen and Stone in 1975 that serum and plasma myoglobin levels increase with AMI. Information has begun to accumu-late suggesting patients with AMI show a rise in myoglobin levels before either mass CKMB or cardiac troponin I (cTnI). The rapid clearance of myoglobin from the circulation in uncomplicated AMI, makes this marker suitable for de-tection of reinfarction in patients with recurrent chest pain. Myoglobin in Emergency Department Diagnosis of AMI Until the early 1980’s most patients presenting with chest pain were routinely being admitted to the hospital for observation. However, two parallel events helped to change how laboratory tests to detect AMI would be used in the Emergency Department (ED) setting. The first of these events was the adoption of the Diagnostic Related Group (DRG) method of payment for in-patient treatment of Medicare and Medicaid patients in 1983 and the rise of managed care in the private sector. The payment change introduced for the first time significant monetary deincentives for inappropriate admission of patients who turned out to not have myocardial events. Next, in 1985 the first reports began to appear on the use of tissue-type plasminogen activator (TPA) and then subsequently streptokinase as alternatives to angioplasty by their ability to dissolve coronary occlusions. The common link be-tween all the clot busting drugs was that the sooner they were administered after the arrival in the ED, the better the patient outcome. Therefore, the detection of reperfusion by lab tests could significantly alter subse-quent patient management. These two events contrib-uted to the increased interest in finding better methods to correctly identify patients actually having myocardial events and to detect refusion as soon as possible after administration of thrombolytic agents. One of the difficulties in trying to make the diagnosis of AMI in the ED is that patients present at widely variable times after onset of chest pain depending on personal, societal and logistical considerations. Some locations may show 90% of patients presenting within 6 hours of chest pain onset, while others in different parts of the United States commonly see over 50% of patients pre-senting more than 12 hours after onset of chest pain. A further complication is that only 10% of those with chest pain will have ST-segment elevations and AMI, the clas-sic indicator of coronary artery occlusion. Another 10% will have non-diagnostic ECG changes (e.g., ST-segment decreases) and AMI. About 30% of those presenting with chest pain will have acute coronary syndromes and no AMI, while the remaining 50% of those presenting with chest pain will have no cardiac etiology. Myoglobin
  • 17. 17 Laboratory testing is not involved in the diagnostic deci-sion to use thrombolytic therapy since, with occlusion, serum and plasma markers may not reflect the extent of the myocardial damage due to their accumulation on the other side of the blockage. The correct clinical diagno-sis of the remaining AMI presenting population, which have no ST-segment increase, relies increasingly on laboratory tests. Since patients may present within less than an hour to over 24 hours after onset of chest pain, laboratory diagnostic markers of AMI need to be selected based on when the levels in serum and plasma become diagnostic. Myoglobin rises quickly after AMI and becomes diagnostic within 2-4 hours post-infarct, peaking at 9-12 hours and returning to baseline within 24-36 hours. Myo-globin tends to rise and fall sooner than other cardiac mark-ers with AMI. Myoglobin Utility on ED Presentation The time of patient presentation in the Emergency De-partment (ED) with possible AMI is nearly always more reliably known than the time, based on patient recollec-tion, of chest pain onset. Therefore, many reports relate the clinical utility of the various markers for AMI to the time of initial patient presentation in the ED. Generally myoglobin is found to have a better sensitivity for AMI in the first 4 hours after chest pain onset than mass CKMB, cTnI and cardiac troponin-T (cTnT), but has a lower speci-ficity than these other markers. A number of reports sug-gest the measurement of myoglobin as a diagnostic aid in “ruling-out” AMI with negative predictive values of up to 100% reported at certain times after onset of symptoms. A study in 1996 by Gornall and Levinott using serial sam-pling at 1, 2, and 6 hours after presentation showed myo-globin had the highest sensitivity, specificity, and nega-tive predictive value for AMI in the first 2 hours of ED presentation. They found myoglobin’s sensitivity to be 77% at 1 hour, and 93% at 2 hours, thus the serial pre-dictive efficiency at presentation was 96% vs. Baseline ECG at 70% and CKMB at 62%. Another study also based on collecting blood at presentation and 1 and 2 hours later, using a criteria of myoglobin above 100 ng/ mL or a change (+/-) from baseline of more than 50% found similar results. Sensitivity in the 0-2 hour period was 93%, with a 95% confidence interval of 59-92%. The literature suggests that by using the change from baseline presentation, myoglobin may yield the most sensitive and specific assessments of AMI detection if myoglobin mea-surements alone must be employed. Myoglobin with Perioperative Myocardial Infarction The same characteristics which make myoglobin useful for the detection of AMI in the ED have been found in perioperative MI in both cardiac and noncardiac surgical patients. For example, one study of coronary artery by-pass grafting (CABG) surgical patients found that myo-globin levels peaked within 1 hour after aortic unclamping, and returned to baseline within 4 hours. When a perioperative MI occurred, myoglobin levels were elevated 1 hour after aortic unclamping, and continued to be significantly elevated at least 3 hours post-surgery compared with levels observed in non-MI patients. Myo-globin levels in patients who suffered perioperative MIs were found to be greater than 400 ng/mL 4 hours after the procedure. Cautions with Use of Myoglobin There are several clinical situations where myoglobin results should be used with caution in assessing AMI. The most significant source of false myoglobin eleva-tions is skeletal muscle injury. It has been found that myoglobin and mass CKMB may be elevated in some patients after electrical cardioversion while cardiac tropo-nin I remains normal. Individuals running marathons have also been shown to have elevations of myoglobin, mass CKMB and CK isoforms but normal cTnI levels. The specificity of myoglobin for AMI detection was found to fall from 82% in non-cocaine users to 50% in individu-als who recently used cocaine. Other clinical situations, such as severe burn victims, Rhabdomyolysis patients or other disease states where skeletal muscle injury is involved could cause increased levels of myoglobin.
  • 18. 18 Physiology Structure and Function: Troponin C, I and T are polypep-tides that combine to form a CIT complex on the thin fila-ment (actin) portion of the contractile apparatus of cardiac and skeletal striated muscle. This troponin complex is not present in smooth muscle. Troponin C (TnC) can bind up to four calcium ions and is responsible for regulating thin fila-ment activation; troponin I (TnI) binds and inhibits acto-myosin ATPase thereby blocking myosin movement, and troponin T (TnT) binds TnI and TnC to the actin-tropomyo-sin filament positioning the complex on the filament. The size of the troponins vary from 23,500 daltons for TnI to 37,000 daltons for TnT. TnC has a variable molecular weight depending on the number of calcium molecules bound. The binding affinity of TnC for TnI is very high at about 108 L/mol if calcium is present, this enables TnI to wrap around TnC contacting both N- and C-terminal globular domain Ca2+ binding sites on TnC. Removal of Ca2+ with EDTA decreases the TnC - TnI binding affinity 1,000 fold. TnI has three striated muscle isoforms. Cardiac troponin I (cTnI) is found only in heart striated muscle. Slow twitch Troponin I (stTnI) is the isoform found in “red” striated muscle such as in the soleus and diaphragm. Lastly, fast twitch troponin I (ftTnI) is the isoform associated with the “white” striated muscle in skeletal muscles such as the biceps and triceps. Production of each isoform is controlled by a different gene. Because the amino acid sequence of each isoform is different, e. g., cTnI has 31 unique amino acids at its N-terminal end, antibodies can be developed which can specifically detect cTnI, even in the presence of high levels of the stTnI and ftTnI striated muscle TnI isoforms. cTnI is not seen in fetal skeletal muscle, in regenerating skeletal muscle or in chronic muscle disease. TnT also shows differences between cardiac and skeletal muscle isoforms; however, the pres-ence of cardiac TnT in fetal skeletal muscle raises con-cern that regenerating skeletal muscle may also produce the cardiac TnT isoform. TnC has the identical amino acid sequence in cardiac and skeletal muscle so it is not use-ful as a marker for cardiac tissue damage. Clinical Significance Troponin Release from Cells: Most troponin is bound into CIT complexes as part of the myofibril structure within the muscle myocytes. cTnI and cTnT are present in stoichio-metrically equal amounts in the contractile apparatus of striated muscle. A smaller amount of cTnI, cTnT and TnC, approximately 6% to 8% of cTnT and 2.5% of total cTnI, resides in the myocyte’s cytoplasm. When the myocyte experiences ischemia, one of the first responses is swelling. The cell swelling is believed to result in a sequential loss of cytoplasmic constituents into the in-terstitial spaces. This cytoplasmic leakage occurs in a manner that is roughly dependent on the molecular weight of each constituent. In this leakage model, very small molecular weight molecules such as electrolytes are lost first. Then macromolecules of increasing size such as myoglobin (17,800 daltons) and enzymes such as creatine kinase (both MM and MB) at 86,000 daltons and lactate dehydrogenase (LD) at 135,000 daltons are lost from the cytoplasm. The macromolecule size, which defines the threshold between leaking and ruptured cells, is not well defined. However, it is likely that molecules the size of CKMB and LD may not be able to escape from an intact living cell. However, there is evidence that cTnI, at approximately 23,500 daltons, is lost from myo-cyte cytoplasm before cell death in conditions such as unstable angina. cTnI seems to be released from the cytoplasm of myocytes in patients with acute myocardial infarction (AMI) at about the same time as CKMB based on the timing of cTnI’s appearance in serum and plasma. Both mass CKMB and cTnI first attain optimal diagnostic effi-ciencies for AMI in the range of 80% at about 6 hours after onset of chest pain. However, cTnI is about 13 times higher in concentration in the myocardium than CKMB, offering the possibility of greater sensitivity to myocar-dial damage than CKMB. If the ischemia is not reversed the myocyte will die releasing structural elements, includ-ing the myofibril troponin CIT complexes into the circula-tion. Support for this sequential release model comes from the well known sequence of appearance of bio-chemical markers in serum and plasma. The bimodal rise of serum and plasma cTnT is also believed to be due to an initial cytoplasmic release followed by a second cTnT rise due to the slower myofibrillar degradation occuring with cell death. Free vs Complexed cTnI Little free cTnI is measurable in serum and plasma after acute myocardial infarction. Because of the high binding affinity of cTnI for TnC in the presence of Ca2+, nearly all measurable serum and plasma cTnI exists as a cTnI-TnC complex. The use of blood collection tubes containing strong Ca2+ chelators like EDTA (purple top tubes) dra-matically lowers the cTnI-TnC binding affinity, resulting in more free cTnI in the sample. Studies using highly selective antibodies for free and complexed cTnI show that nearly all of the measurable cTnI in acute myocar-dial infarction (AMI) is due to an increase of the cTnI-Troponin- I
  • 19. 19 TnC complexes. Peak concentrations of total cTnI lev-els with AMI were found to be 5 to 12 times higher than the corresponding free cTnI levels. This information sug-gests that measurement of free cTnI should not contrib-ute significantly to the diagnostic efficiency of serum and plasma cTnI assays for myocardial infarction detection. Cardiac Troponin I (cTnI) Measurement cTnI’s unique 31 amino acid N-terminal sequence allows antibodies to be developed to highly discrete epitope sites on the polypeptide molecule. A properly selected anti-body permits highly specific cTnI measurement in the presence of severe skeletal muscle trauma and the high levels of serum skeletal muscle troponin isoforms that would result. Whether an immunoassay measures complexed or free cTnI depends on the cTnI epitopes to which the antibody is directed. AMI is generally associ-ated with a serum rise of the cTnI-TnC complex; there-fore, assays must measure this form of cTnI to be clini-cally useful in AMI assessment. There is also evidence that oxidation of cysteine-SH groups on cTnI may alter the exposed epitopes and the ability of some assays to detect cTnI. Since serum and plasma cTnI levels are normally low to non-detectable, clinical laboratory mea-surement of cTnI requires a highly specific antibody de-tection system and use of a final measurement system that can generate a detectable signal at low analyte con-centrations. One of the biggest problems faced in the clinical appli-cation of cTnI tests is that assays frequently employ an-tibodies with different cTnI epitope specificities, and each commercial method employs a different calibrator mate-rial. The result is that commercial cTnI methods from dif-ferent companies do not produce the same analytical results and may vary as much as 30 fold on the same sample. As a practical matter this means that diagnostic cutoff values and clinical utility decision points presented in the literature are highly method specific and should not be exported to another cTnI method without repeating the clinical studies needed to establish new numerical decision points associated with, for example, unstable an-gina and myocardial infarction. Therefore, the method used (e. g., Dade Behring Stratus®) will be indicated as relevant clinical literature findings are discussed in this article. cTnI in Acute Myocardial Infarction Several lines of evidence suggest cTnI may be a good marker for myocardial injury. cTnI concentration is about 13 times higher in heart muscle than CKMB; therefore, small amounts of injury should generate a large increase from normal background levels. In practice, some clini-cal decision points may be near the assay’s analytical sensitivity since there is no measurable cTnI in serum or plasma of individuals without cardiac involvement. Indeed, cTnI (as measured by Dade Behring Stratus®) has not been found to increase in serum or plasma in conditions with acute skeletal muscle damage like trauma or rhabdomyolysis, or with chronic skeletal muscle dam-age such as polymyositis or Duchenne’s muscular dystropy, or after running a marathon. Because those same acute and chronic skeletal muscle pathologies can result in increases in CKMB that are not associated with AMI, the cTnI offers significantly improved specificity for myocardial damage. Chronic renal failure with hemo-dialysis or peritoneal dialysis also shows no increase of cTnI. Elevations in cTnI that correlate closely with myo-cardial injury have been verified by echocardiography. These characteristics all suggest that cTnI should be a better overall serum and plasma marker for routine as-sessment of myocardial infarction than CKMB. cTnI in Emergency Department Diagnosis of AMI One of the difficulties associated with making the diagno-sis of acute myocardial infarction (AMI) in the emergency department is that patients present at widely variable times after the onset of chest pain depending on per-sonal, societal and logistical considerations. Some loca-tions may show 90% of patients presenting within 6 hours of chest pain onset, while others (in different parts of the United States) commonly see over 50% of patients pre-senting more than 12 hours after the onset of chest pain. To further complicate matters, currently only 50% of pa-tients actually having AMI and 10% of individuals who present with chest pain, show the ST-segment eleva-tions associated with occlusion of coronary arteries and require emergency thrombolytic therapy or angioplasty. Another 10% of individuals who present with chest pain will have nondiagnostic electrocardiogram (ECG) changes (e. g., ST-segment decreases), and AMI. About 30% of those presenting with chest pain will have acute coronary syndromes and no AMI, and in the remaining 50% of their chest pain will have no cardiac etiology. Laboratory testing is often not involved in the diagnostic decision to use thrombolytic therapy since, with occlusion, cardiac markers may not reflect the extent of myocardial damage due to their accumulation on the other side of a blockage. The correct clinical diagnosis of patients pre-senting with AMI who have no ST-segment increase relies increasingly on laboratory tests. Since patients may present within less than an hour to over 24 hours after onset of chest pain, laboratory diagnostic markers of AMI need to be selected based on when the levels in serum and plasma become diagnostic. Myoglobin rises quickly after AMI and becomes diagnostic at about 1 to 3 hours after chest pain onset and remains elevated for 12 to 24 hours. It is a useful marker for “ruleout” AMI within 24 hours. CKMB begins to rise at 3 to 4 hours after onset of chest pain, becomes diagnostic at about 6 hours, peaks at 12 to 24 hours, and remains elevated about 24 to 36 hours. LD isoenzyme 1(LD1) becomes diagnostic at about 10 hours after chest pain onset and remains el-
  • 20. 20 evated for at least 72 hours. cTnI begins to rise 4 to 6 hours after chest pain onset, becomes diagnostic at about 6 hours, and will remain elevated for from 4 to 14 days. Consequently, cTnI tends to rise like CKMB in serum but stays increased for as long as LD1. This release pattern enables the use of cTnI to detect AMI over a longer period of time from the onset of chest pain than the other three traditional markers of AMI. Well controlled studies wherein serum or plasma levels of multiple markers of myocardial injury were examined in the same sample and related to time of chest pain onset show cTnI rises in serum at or before the time of CKMB. These studies consistently show that CKMB and cTnI have poor diagnostic sensitivity (e. g., less than 50%) for myocardial injury before 6 hours after onset of chest pain. In one typical study, in the 7 to 36 hour pe-riod after chest pain onset cTnI sensitivity was 88 - 100% compared to CKMB sensitivity of 61% to 81%. Studies using serial testing of patients with possible acute myocardial events, but no ST-segment elevation, have consistently shown that absence of cTnI rise indicates a low risk of near term coronary death or infarction. In one study, 773 consecutive patients presenting with chest pain of less than 12 hours duration and no ST-segment elevation were tested on admission and again 4 hours later, or at a second time corresponding to at least 6 hours after chest pain onset. The risk of coronary death or infarct in those with negative cTnI levels on both tests was only 0.3%. In another study, cTnI values less than 0.6 ng/mL showed negative predictive values (NPV-%) results, to be 85% at presentation to the ED, 85.5 at 1 hour later, 87.4 at 2 hours, 96.7 at 6 hours, and 98% at 12-24 hours. The positive predictive value (NPV+%) at time of ED presentation was 25%, 40% 1 hour later, 60% at 2 hours, 88% at 6 hours, and 88.9% at 12-24 hours. This and other studies strongly suggest serial cTnI testing assists in the clinical decision to discharge or ad-mit a patient presenting at the ED with possible AMI. Available information suggests that cTnI or CKMB results from serum or plasma collected less than 4 hours after onset of chest pain may yield results still within the normal range that could change to positive in samples collected later. cTnI in Acute Coronary Syndromes Acute coronary syndromes (ACS) share a common pathophysiologic mechanism which is initiated when a coronary atherosclerotic plaque ruptures and a throm-bus forms that interrupts perfusion of myocardial tissue. Patients presenting with resting chest pain, but with no ST-segment elevation on the electrocardiogram (ECG), may have an ACS such as unstable angina or non-Q wave AMI. Detection of ACS frequently cannot be done clinically or by angiography. Consequently, analysis of serum markers of myocardial damage such as CKMB and cTnI have been routinely ordered to assist in detection of ACS. Making the diagnosis at the time the patient presents with the first ACS chest pain event is critical since, without intervention, 40% of those patients will have an AMI within 3 months. About 17% of those pre-senting with ACS will die within 3 months. Early inter-vention can reduce the risk of AMI to 8% and death to 3% within the same 3 month period. Evidence now suggests that increased serum and plasma cTnI level in the first 24 hours from onset of chest pain is a much more sensitive and specific marker for ACS than CKMB. The prognostic value (predictive value of positive results) of cTnI increase is highest in patients presenting more than six hours after onset of chest pain. One study used samples collected from 1,404 patients presenting with unstable angina or non-Q wave AMI as part of the Thrombolysis in Myocardial Ischemia Phase IIIB (TIMI IIIB) study. It was found that 2.6% of patients presenting within 6 to 24 hours after chest pain onset, having cTnI (Dade Behring Stratus®) levels above 0.4 ng/mL but negative mass CKMB levels, died from car-diac causes within 42 days. If cTnI was less than 0.4 ng/ mL, mortality within the 42 days was 5 fold less, at 0.5%. This and other studies suggest that cTnI is a useful di-agnostic marker for ACS, the utility of which will likely improve as detection methods for cTnI evolve. cTnI to Assess Cardiac Contusion Detection of cardiac injury in patients with chest trauma can be difficult because the serum and plasma level of mass CKMB could be elevated due to collateral skeletal muscle injury. Studies using transthoracic echocardiograms as the criteria for myocardial damage show that serum and plasma cTnI levels are much more specific than mass CKMB lev-els for detecting cardiac contusion. In one study (using Dade Behring Stratus® II) of 44 patients without cardiac contu-sion, none had serum cTnI levels above 0.35 ng/mL, sug-gesting no cardiac damage. In contrast, 26 patients had serum CKMB mass levels above 6.7 ng/mL, suggesting cardiac damage was present. That the commonly employed CKMB/Total CK ratio percent does not correct for all skel-etal muscle damage is suggested by the finding that 3% of the patients in this study had levels above 2.5%, a range generally attributed to cardiac damage. Several studies also suggest that cTnI may be useful for detection of unsus-pected perioperative myocardial injury or infarction. cTnI to Assess Myocardial Injury in Critically Ill Patients Critically ill patients are exposed to stresses that increase tissue oxygen demand at a time when oxygen supply to the myocardium could decrease due to a combination of variables such as hypoxia, fall in blood pressure, tachycar-dia, anemia and septicemia. Available evidence suggests that myocardial injury may go undetected in about 66% of critically ill patients in the absence of cTnI surveillance.
  • 22. 22
  • 23. 23 General Information Hemostasis, the process which prevents the loss of blood from a blood vessel, is accomplished through three inter-related events: vascular constriction, platelet aggregation, and coagulation. The initial vasoconstriction is short-lived. It enables the second event, platelet aggregation, to pro-duce a fragile, unstable platelet plug. Prolonged hemosta-sis depends upon the third phenomenon, the formation of a blood clot. To be effective a clot must form promptly and yet be closely confined to the damaged portion of the vessel. Coagulation is a highly complicated process in which the factors which prevent clotting are delicately balanced against those which promote it. Understanding the complex coagulation process is made more difficult by the lack of standard nomenclature. In some cases, the clotting factors are given their chemical names (i.e. calcium, prothrombin, fibrinogen). Other factors have also been named for their functions (proaccelerin, proconver-tin, antihemophilic factor). In still other cases factors were given the surname of the person in whom hereditary deficiency of that factor was first detected (Hageman, Fletcher, and Stuart). In an attempt to reduce confusion, Roman numerals have been assigned to most of the clot-ting factors. The numbers reflect the order of their dis-covery, and not the sequence in which they participate in the clotting reactions. Factors may exist in plasma as the inactive protein or precursor which is converted to the active form during the clotting process. The active form is identified by appending a lower case ‘a’ to the Roman numeral, e.g. factor Xa. Even in normal plasma the stage is constantly set for clotting. It can be triggered by events which take place either within the blood itself, in the blood vessel wall, or in the surrounding tissues. Regardless of how clotting is initiated, the last several steps are identical. These final steps result in the formation of a three-dimensional fibrin lattice in which platelets, red cells, and white cells are trapped. Fibrin is an insoluble protein which polymerizes spontane-ously. The initial polymers aggregate due to electrostatic and hydrophobic bonds. The immature fibrin aggregate is therefore gelatinous and easily broken up. As the clot matures, covalent (peptide) bonds are formed between the fibrin strands, strengthening and contracting the clot. This process is catalyzed by a transglutaminase, factor XIIIa. Fibrin cannot exist per se in plasma. It must neverthe-less be readily available in abundant quantity. Plasma therefore contains large amounts of a fibrin precursor, fibrinogen. Fibrinogen is converted to fibrin by the catalytic action of the enzyme thrombin. Figure 1 Xa Prothrombin Thrombin Fibrinogen Fibrin Monomer Soluble Fibrin Stabilized Fibrin (Fibrin Clot) Ca++, PF3 Va Ca++ XIII XIIIa Thrombin also is responsible for the activation of factor XIII (i.e. the conversion of protransglutaminase to trans-glutaminase) in the final step of clot formation. Thrombin is a key enzyme in coagulation. If thrombin existed in the active form in plasma, uncontrolled clotting would occur. It, too, circulates as an inactive precursor, prothrombin. The conversion of prothrombin to thrombin is catalyzed by factor Xa. This has proved to be a com-plex enzymatic reaction requiring the presence of factor Va, calcium, and a phospholipid-Platelet Factor 3 (PF3) (See figure 1). The activation of factor X is pivotal in clotting. It can come about in two different ways, referred to as the intrinsic and extrinsic pathways; the distinction is based upon the source of the activators. The intrinsic pathway is acti-vated by trauma within the vascular system such as ex-posed endothelium. In contrast, the extrinsic pathway is activated by external trauma such as a cut. Of the two systems, the intrinsic is slower but more important. The intrinsic system involves a series of reactions in which the product of one acts as a catalyst or cofactor for the succeeding reaction. This reaction chain is often referred to as a “cascade” (See figure 2). The intrinsic pathway is triggered when blood comes in contact with an unfamiliar surface such as a damaged blood vessel wall, causing a conformational change in factor XII and converting it to factor XIIa. Factor XIIa is a weak initiator of several reactions, including the activation of factor XI to XIa and the conversion of prekallikrein (Fletcher factor) to kallikrein. Factor XIa activates factor IX. Factor IXa is an endopeptidase which, in the presence of factor VIIIa converts factor X to factor Xa. The concomitant presence of factor VIIIa, platelet phospholipids (PF3), and calcium is required for the activation of factor X to proceed rapidly.
  • 24. 24 Figure 2 XII Activator Chemicals Collagen Exposed Endothelium Platelets XIIa kallikrein prekallikrein Intrinsic Pathway XI XIa IX IXa Ca++, PF3 VIIIa X Xa Clotting is initiated through the extrinsic system when blood escapes from the vascular system and contacts damaged tissues. A poorly understood tissue factor called thromboplastin is released from the damaged cells. It forms a complex with calcium and factor VII which is able to activate factor X directly (See figure 3). Thus several of the early steps which must occur in the intrinsic system are bypassed in the extrinsic system. Clotting occurs much more quickly by this route. Figure 3 Extrinsic Pathway VII Tissue Factor VIIa X Xa Control of Clotting Once clotting has started it is important to control its rate and, when hemostasis is achieved, to stop it altogether. This is accomplished by several mechanisms. The flow of blood past the site of the developing clot washes away the activated clotting factors and delivers them to the liver where they are removed from the blood. In addition there are several circulating clotting inhibitors, including antithrombin III (AT III), alpha-1-antitrypsin, C1 inhibitor, and alpha-2-macroglobulin. Of these, AT III is the most important. As its name implies, its primary role is to com-bine with and thereby inactivate thrombin. This inhibition proceeds relatively slowly but is greatly accelerated in the presence of heparin. AT III also inactivates factors Xa, and to a lesser extent, inactivates IXa, XIa, and XIIa. Inhibition of these factors is also accelerated by heparin (See figure 4). Figure 4 Antithrombin III + Heparin Inactive Xa Complex Inactive Thrombin Complex Xa Thrombin
  • 25. Inactive Xa Complex Activators Kallikrein (Intrinsic) Tissue Activator (Extrinsic) Urokinase Streptokinase Antithrombin III + Heparin Prothrombin Thrombin 25 Clot Dissolution Blood clots are not intended to be permanent. Their pur-pose is to stop the flow of blood until the damaged blood vessel can be repaired. Normally all but the largest clots will be completely dissolved. Dissolution, which begins almost as soon as the clot is formed, is accomplished by the hydrolytic action of the enzyme plasmin. Plasmin also hydrolyzes fibrinogen and other clotting factors. Therefore, it cannot exist in its active form in plasma. An elaborate control system exists which allows clot lysis to proceed while protecting clotting factors against destructive hydrolysis. Normal plasma contains an inactive precursor of plasmin called plasminogen. Plasminogen has an affinity for fibrin molecules and is incorporated within the fibrin strands as the clot forms. Plasminogen remains dormant inside the clot until activated by specific enzymes called plas-minogen activators. Such activators have been found in tissues, in urine, in plasma, and, most importantly, in the endothelial cells which line blood vessels. Factor XIIa also accelerates the activation of plasminogen. Thus, the factor which triggers the clotting process also contributes to clot dissolution. The fact that a clot forms at all is ex-plained by the relative rates of the two processes. Clot for-mation occurs rapidly until it is slowed by specific limiting mechanisms. Clot dissolution, on the other hand, is slow in starting, but is a continuous process (See figure 5). Hemostasis XII XIIa Activator Chemicals Collagen Exposed Endothelium Platelets Kallikrein Prekallikrein Kallikrein Extrinsic – Intrinsic Interaction Extrinsic System Intrinsic System Common Pathway Fibrinolytic Pathway Inhibition VIIa XIa Tissue Factor XI VII IX IXa X Xa XIII XIIIa Ca++, PF3 VIIIa Ca++, PF3 Va Activators Kallikrein (Intrinsic) Tissue Activator (Extrinsic) Urokinase Streptokinase Inactive Thrombin Complex inactive plasmin complex Fibrin (ogen) Degradation Product Stabilized Fibrin Plasminogen a2antiplasmin Plasmin Fibrinogen FpA FpB Fibrin Monomer Soluble Fibrin Figure 5 inactive plasmin complex Fibrin (ogen) Degradation Product Fibrinogen Fibrin Monomer Soluble Fibrin Stabilized Fibrin Plasminogen a2antiplasmin Plasmin Active plasmin sometimes finds its way into the plasma; it may escape from a dissolving clot or be formed if plas-minogen activators gain access to the blood stream. When this happens, fibrinogen and other essential clotting proteins, including factors V and VIII, are destroyed. To prevent this, plasmin inhibitors, the most important of which is alpha-2-antiplasmin, quickly inactivate any plasmin in the circulation.
  • 26. 26 Antithrombin III Physiology: Antithrombin III (AT III) is a plasma pro-tein which is an inhibitor of serine proteases. As the name implies, its main physiologic function is the inactivation of the key clotting enzyme, thrombin. It also inactivates other clotting and clot-lysing enzymes, including factors Xa, IXa, XIa, XIIa, kallikrein, urokinase, and plasmin, all of which are serine proteases. AT III is a glycoprotein with a molecular weight of 65,000. It migrates as an al-pha- globulin on electrophoresis. It is synthesized by the liver and is widely distributed in body fluids. Its physi-ologic half-life is about four days. As with other coagulation factors, the exact catabolic pathway is not known. Furthermore, normal plasma contains more AT III than prothrombin. The fact that clots are able to form at all is due to the fact that the inhibitory reaction is much slower than the thrombin-catalyzed fibrinogen to fibrin reaction. This situation is altered in the presence of heparin, which greatly increases the inhibitory activity of AT III through a conformational change in the molecule. Heparin is therefore an accelerator of AT III. This appears to be the mode of action for this important anticoagulant. Clinical Significance: AT III is normally present in plasma. In disease, its concentration may be either increased or decreased. Increases: Damage to body tissue may produce a rise in plasma AT III which could result in an increased ten-dency to bleed. Decreases: Decreased AT III is of much greater clinical importance. A reduction in AT III leads to an increased tendency for blood to clot within the blood vessels (throm-bosis). Once formed thrombi tend to enlarge or progagate. Rapidly propagating thrombi may break away from the site where they are formed and lodge elsewhere in the vascular system (embolization). Serious consequences, even death, may follow embolization to the heart, lung or brain. AT III levels less than 60% of normal are asso-ciated with increased risk of thromboembolism. An inherited inability to synthesize normal AT III accounts for only a very small percentage of patients (2%) with thromboembolism. Several families with this disorder have been discovered. These individuals have repeated episodes of thromboembolism. Thromboembolism is very rare among young persons; when it does occur, hereditary AT III deficiency should be considered as a possible cause. Decreased AT III is more commonly an acquired condition. Impaired synthesis or increased utilization, or a combi-nation of the two, may be the basic problem. Since AT III is synthesized in the liver, diseases of this organ may be responsible for decreases in plasma AT III levels. AT III is a molecule about the same size as albumin and, consequently, may be easily lost in the urine in kidney dis-ease. Nephrosis, which is characterized by massive albu-minuria, may also be accompanied by AT III deficiency. AT III deficiency may result from (as well as cause) exten-sive intravascular coagulation (DIC). In DIC, thrombin, plas-min, and other serine proteases may be produced at such a rate as to deplete plasma AT III. Prolonged use of oral contraceptives is accompanied by some decrease in AT III levels. The exact significance of this is not known, but it may contribute to the slightly increased incidence of thromboembolism observed among women takin anovulatory medication. The assay of plasma AT III is helpful in monitoring heparin therapy. Patients with low AT III levels have a reduced response to heparin. Heparin may itself lower AT III levels. When this occurs, the patient has an increased tendency toward thrombosis after the heparin therapy is discontinued. Regardless of cause, low AT III signifies a hypercoagulable state which can be dangerous and which, with proper treatment, can be avoided.
  • 27. Fibrin Degradation Products 27 Physiology: The proteolytic enzyme, plasmin, cleaves fibrin (and fibrinogen), producing variable-sized, soluble fragments called fibrin split products, or fibrin degrada-tion products. In normal circulating plasma there is very little active plasmin, hence only small amounts of FDP are formed and are quickly removed from the body by the liver. Plasmin activity may be increased secondary to a variety of disease processes. The level of FDP will increase if the rate of their formation exceeds the rate of removal. They have an inhibitory effect on the conversion of fibrinogen to fibrin, and may also impede platelet aggregation. Fi-brin degradation products are measured in serum, using an antibody against fibrinogen and FDP. The fragmenta-tion of fibrin/fibrinogen by plasmin destroys the ability of fibrinogen to form an insoluble clot, but only slightly changes the antigenic sites. Fibrinogen, which will cross-react with the FDP antibody, is removed from the sample by allowing it to clot. Clinical Significance: Fibrin degradation products are present in plasma in small amounts normally. In-creases indicate clot lysis is proceeding at a faster than normal rate. The FDP assay may also be clinically useful in monitoring patients receiving streptokinase, a plasminogen activator used in treatment of thromboembolic disorders such as deep vein thrombosis, pulmonary embolism, and myo-cardial infarction. Patients receiving streptokinase should have elevated levels of FDP. Increases: FDP levels are increased in a number of diseases such as liver disease, malignant tumors, renal failure, transplant rejection, and disseminated intravascu-lar coagulation (DIC). The highest levels occur in DIC (de-scribed more fully in the section dealing with fibrinogen). In DIC, the delicate balance between clot formation and clot dissolution is disturbed and widespread intravascular clotting occurs. In malignancy or certain obstetrical complications large amounts of procoagulants may be released into the blood stream. In massive injury or burns, the extrinsic clotting pathway is activated. If there is significant damage to vascular endothelium, plasma is exposed to collagen and collagen-like substances which can activate the intrinsic pathway. Whatever the cause of DIC, there is a compensatory increase in the fibrin-olytic activity, and an increase in levels of FDP.
  • 28. Fibrinogen (Clotting Factor I) 28 Physiology: Fibrinogen is the soluble precursor of in-soluble fibrin, the major component of a blood clot. It is a long, rod-like, moderately large (340,000 daltons) glyco-protein. The molecule is constructed of polypeptide chains of three different types designated alpha, beta and gamma, The alpha, beta and gamma chains are joined together by disulfide bonds near their N-terminal segment, forming a monomer. Two such monomers are joined at their heads again by disulfide bonds to form a dimer of six polypeptide chains. When fibrinogen is attacked by the hydrolytic en-zyme thrombin, four segments, two alpha and two beta, are split off and released as fibrinopeptides A and B (FpA, FpB). Assay of fibrinopeptides may be used to estimate the presence and extent of thrombosis. Once thrombin has split off these terminal segments, the remaining molecules are free to polymerize into fibrin strands which ultimately form the basic structure of the blood clot. Most of the total body fibrinogen is intravascular. The liver synthesizes 2-5 grams of fibrinogen per day. Its physiologic half-life is about 4 days. The normal catabolic path way for fibrinogen is not well understood. Fibrinogen FpA FpB Fibrin Clinical Significance: Fibrinogen is measured in plasma. Its concentration may be increased or decreased. In general, decreases are of more clinical importance than increases. Decreases: Decreases in plasma fibrinogen levels might be expected in liver disease since that organ is the site of synthesis. Although hypofibrinogenemia does occur in liver disease, it is not common. The most common cause of low plasma fibrinogen levels is disseminated intravascular coagulation (DIC), a condi-tion in which blood clots form throughout the microvascular system. The process may be either acute or chronic with a mild or fatal outcome. There are many causes of DIC. It accompanies many serious complications of childbirth. Pre-mature separation of the placenta (abruptio placenta) and amniotic fluid embolism (the forced injection of amniotic fluid into the maternal circulation during labor) are the two most common obstetric incidents, in which DIC occurs. Other problems which may produce DIC are retention of a dead fetus and abortion induced by the intra-amniotic in-jection of hypertonic saline. DIC can have two serious consequences: The clots may occlude important blood vessels and the consumption of fibrinogen (and factors VIII and V) may outstrip pro-duction. When fibrinogen levels fall to the point where blood is unable to clot, dangerous bleeding ensues. Fibrinogen levels below 100 mg/dl are associated with an increased risk of bleeding. Increases: Fibrinogen is one of several proteins which increase in the plasma whenever tissue is damaged. Fi-brinogen is not usually assayed directly for the purpose of assessing tissue damage. On the other hand, mea-surement of the rate at which red cells in anticoagulated blood aggregate and settle to the bottom of the tube, the red cell sedimentation rate, is commonly used to detect and monitor inflammation. Fibrinogen is more potent than any other plasma protein in increasing the erythrocyte sedimentation rate. A large number of inherited disorders of fibrinogen synthe-sis have been discovered. A total inability to synthesize this protein (afibrinogenemia) is known, but it is extremely rare. There are many genetic disorders which presumably are due to errors in the polypeptide structure although their exact nature remains to be defined. These disorders are referred to as dysfibrinogenemias. The quantity of plasma fibrino-gen is normal in such persons, but the ability to form clots (functional activity) is impaired, usually due to defective polymerization. For this reason, assays whose end point is clot formation are abnormal, and are interpreted as a decreased fibrinogen level. From a functional point of view, this interpretation is correct. Low fibrinogen levels occasionally result from the presence in plasma of plasmin which has not been inactivated. Plas-min degrades fibrinogen as it does fibrin. This process is known as fibrinogenolysis. It may result from the use of plasminogen activators such as streptokinase in the treat-ment of clotting disorders, or the release of activators from malignant tumors. It may also be due to failure of synthesis of adequate amounts of plasmin inhibitors.
  • 29. 29 Heparin: Heparin was discovered in 1916 by a medi-cal student, Jay McLean, who was attempting to isolate, from various tissues, a substance (thromboplastin) which promotes coagulation. McLean found that certain extracts of tissues contained a substance which prevented instead of promoted coagulation. He and his mentor, Professor W. H. Howell, named this substance “heparin” because of its abundance in the liver (hepar). High concentrations were also found to be present in other tissues notably lung and the walls of small intestines, the two most com-mon sources of commercial heparin today. Chemistry: Heparin is a mucopolysaccharide bearing chemical resemblance to hyaluronic acid, chondroitin sulfate and keratan sulfate. It is a polymer made up of repeating units of sulfated and non-sulfated D-glucosamin, L-iduronic acid, and D-gluronic acid. Phar-macologic preparations of heparin are heterogeneous with a distribution of molecular weights between 5,000 and 30,000 daltons. There are a large number of O- and N- sulfate linkages and carboxyl groups, making it the strongest organic acid in the body. Physiology: Mast cells or endothelial cells are the source of heparin in the body. Mast cells are medium sized mononuclear cells, the cytoplasm of which is stuffed with granules which stain intensely with basic stains. These granules have been found to be comprised mainly of heparin. Mast cells are found throughout loose con-nective tissue in the body but are in greatest numbers in the organs known to be rich in heparin, i.e., lung and liver. Two main functions have been ascribed to heparin. The first is the prevention of intravascular clotting of blood and the second, the clearing of fat from the blood plasma after the ingestion of a heavy fat meal. The anticoagulation function of heparin is generally believed to be the most important one. Heparin has no known effects except those on blood. Effects On Coagulation: Heparin inhibits coagulation of blood in vivo (in the body) and in vitro (in glass). This inhibitory action requires the presence in blood plasma of a naturally occurring alpha-globulin, antithrombin III. Anti-thrombin III (discussed elsewhere in this booklet) is syn-thesized in the liver. Its presence in plasma prevents the spontaneous intravascular coagulation of blood, i.e., it helps maintain blood in the fluid state. Antithrombin III is only slowly active in the absence of heparin but becomes very active in its presence. The apparent mechanism of action involves first the interaction of heparin with the lysine group of anti-thrombin III which exposes a reactive arginine group which in turn combines with and inactivates serine proteases. Coagulation occurs as a result of a series of enzyme reactions. At each step, an inactive proenzyme is converted to an active protease which in turn converts a subse-quent proenzyme to another active protease enzyme. There are several proteins involved in the coagulation of blood. Of these, some are activated by serine proteases (e.g., Prothrombin, and factors VII, IX, X, XI, and XII). Others are co-factors in these reactions (factors Va, VIIIa, kallikrein, and kinin) and one is converted to a transami-dating enzyme (factor XIII). The antithrombin III-heparin complex appears to neutralize all of the serine proteases. (The complex will also neutralize plasmin which is any enzyme active in dissolving clots). Heparin, then, acts at many stages of clot formation but the net effect is the inhibition of the formation of thrombin from prothrombin. Heparin, unlike the anticoagulant, Coumadin®, does not interfere with the synthesis of coagulation-related proteins. Heparin has a weak inhibitory effect on thrombin itself. This is probably not physiologically or pharmacologically important because it requires 30 to 40 times as much heparin to inhibit the action of thrombin than to prevents its formation. Lipoprotein Lipase Activation: Heparin appears to have a specific effect on the release of an enzyme, which catalyzes the hydrolysis of triglycerides in chylo-microns, releasing free fatty acids which rapidly enter tissues. This enzyme, lipoprotein lipase, has the effect of recuing the turbidity of serum. Because of this effect, it is also known as the “plasma clearing factor.” Uncertainty remains regarding the physiologic importance of heparin activation on this lipase. Absorption and Metabolism: Heparin is inactive when given orally; it must be administered parenterally. It may be given intravenously, intramuscularly or subcu-taneously. The biologic half-life is a function of dosage. High dosages are associated with relatively long half-lives. With standard dosage the half-life is in the range of one to two hours. At these dose levels, it is metabolized in the liver to a weakly active form which is excreted by the kidneys. A discovery of this derivative in urine led to its name - uroheparin. Following very large doses of heparin, especially when given intravenously, heparin itself does not cross the placental barrier nor does it enter the milk of lactating women. Heparin Coumadin® is a registered trademark of E.I. du Pont de Nemours & Company, Wilmington, DE 19898
  • 30. 30 Clinical Uses of Heparin: Heparin is used to pre-vent intravascular clotting or to stop further progression of intravascular clotting (clot propagation) once it has begun. Heparin has the advantage over other antico-agulant drugs in that it is immediate in its action. In gen-eral, less heparin is required to prevent the initiation of clotting (prophylaxis) than to halt active clot formation (thrombosis). Physicians use heparin for clot prophylaxis in certain high risk patients. Among these are those of advanced age, with fractures of the leg or hip, with pelvic surgery, with congestive heart disease, with cancer, and with acute myocardial infarction. Common to this group of patients is that they may require long periods of bed rest during which time the blood flow in the large veins of the legs and pelvis is slowed, increasing the tendency of clot for-mation. These clots (thrombi) propagate forming fragile extensions which are not firmly bound to the vessel wall. Fragments of these clots tend to break off and move in the blood stream (embolize). A common and serious event is the movement of a large clot fragment (an em-bolus) from one of the deep veins of the legs or pelvis to the lung. The embolus is caught up in one of the major arteries of the lung abruptly stopping the flow of blood to that part of the lung. The disorder that follows, pulmonary embolization, is a very common cause of death in these patients. The use of low dose heparin in preventing deep vein thrombosis and high dosages in treating it once it has occurred has greatly reduced the mortality rate among these high risk patients. Heparin is believed to be of value in the treatment of a phenomenon known as disseminated intravascular coagu-lation. Normally blood does not clot in the blood vessels. There are at least two reasons for this. Circulating blood does not come in contact with substances which induce clotting and whatever activated clotting factors do appear are rapidly removed by body cells, particularly those of the liver. In many diseases, sufficient clot activation factors may enter the blood stream and initiate clotting throughout the vascular system. This state is referred to as dissemi-nated intravascular coagulation (DIC). DIC has many causes. Among these are serious infections, complications of pregnancy, malignant disease (cancer), and massive physical trauma. The successful treatment of DIC requires the control or elimination of the cause but until that can be achieved, heparin is useful in preventing further intravascular clot formations. Because of the quickness of action and the ease of neutralization of its anticoagulation effect by the use of such substances as protamine sulfate, heparin is used to prevent coagulation during the use of extracorporeal apparatus such as the heart-lung machine, which is em-ployed to maintain aeration and circulation of blood while patients are undergoing heart surgery or hemodialysis apparatus which are used to clear the blood of unwanted metabolites in patients without functioning kidneys. In all of these uses, it is important to monitor the plasma in order to make appropriate adjustments in heparin dosage. Tests Used to Monitor Heparin Treatment: In general, heparin treatment is monitored by the measure-ment of its effect on coagulation. The first such test was the measurement of the length of time required for whole blood to clot in a test tube (the Lee White clotting test). Today the most commonly used test is the activated partial thromboplastin test (APTT). This test is performed on citrated plasma in which the clotting factors have been activated by a surface contact activator such as kaolin. The APTT is the time required for clot formations after the plasma has been treated with the activator, and phos-pholipid and calcium chloride have been added. The APTT is sensitive to small amounts of heparin. Unfortu-nately, commercially available thromboplastin preparations vary in their sensitivity to heparin. Some are so insensitive as to fail to detect therapeutic levels of heparin at all. This variation in sensitivity means that each batch of re-agents should be tested and standards established. Each hospital laboratory, therefore, must determine its own therapeutic range. An assay system which measures heparin directly and depends upon the inhibition of factor Xa by heparin-activated antithrombin III has been introduced. In this procedure, a synthetic peptide which has a chromogenic group attached is employed as a substrate for factor Xa. Factor Xa, a serine protease, causes the release of the chromogen producing a color change. The assay is sen-sitive to very small quantities of heparin but even more important, it is reproducible, accurate, and can be stan-dardized over several reagent lots and heparin sources.
  • 31. 31 Physiology: Plasminogen is the precursor of the fibrin-splitting enzyme, plasmin. It is a single chain glycoprotein with a molecular weight of about 90,000 daltons. It circu-lates in normal plasma in concentration of about 20 mg/dl. Plasminogen is synthesized by the liver and has a biologic half-life of about 2 days. The conversion of inactive plasmi-nogen to the active, trypsin-like protease, plasmin is brought about by several activators. The most important of these is found in the endothelial cells which line blood vessels. Clinical Significance: Plasminogen is measured in plasma. Its concentration may be increased or decreased. In general, only decreases are of clinical importance. Increases: Physiologic increases in plasminogen con-centrations occur in pregnancy. Certain contraceptives may also increase circulating plasminogen levels. In-creases can occur in the acute phase of inflammation. Decreases: Low plasminogen levels are usually due to liver disease with impaired synthesis, or to increased utilization associated with DIC (this disorder is discussed in detail in the section on fibrinogen). Low levels may occur in severe nephrosis due to loss of this relatively small protein molecule in the urine. Primary fibrinolysis is a rare cause of low plasminogen levels. In this disorder abnormally large quantities of plas-minogen activators are released into the circulation. There are also rare genetic disorders in which individuals syn-thesize a defective, inactive plasminogen molecule (dys-functional plasminogen). Plasminogen
  • 32. 32
  • 34. 34
  • 35. 35 Carbon Dioxide Physiology: The major end products of the metabolism of most foodstuffs are water and carbon dioxide (CO2). Approximately 20 moles of CO2 are produced each day (the exact amount varies with body size and physical activity) and excreted by the lungs. The CO2 formed body cells promptly dissolve in water and combine with it to form carbonic acid. The latter process is a slow one in a simple aqueous solution in a test tube, but in the body it is markedly accelerated by the ubiquitous enzyme, car-bonic anhydrase. The carbonic acid partially dissociates into hydrogen ion (H+) and bicarbonate ion (HCO3). These reactions are summarized in the following equations. K1 K CO 2 + H2O H2CO3 2 H+ + HCO3 Thus carbon dioxide is transported from the tissues to the lungs in three forms: dissolved CO2, undissociated or molecular H2CO3, and bicarbonate ion (HCO3). At normal plasma pH (7.4), the amounts of each of these are as follows: hydrogen ion, 4 x 10–8 M; dissolved CO2, 1 x 10–3 M; molecular carbonic acid, 5 x 10–6 M; bicarbonate ion, 25 x 10–3 M. These relative amounts are determined by the equilibrium constants for the equations above. The role of bicarbonate ion as the major transport form of CO2 is ordinarily of only passing interest to the physician; of much greater importance is its role in control of plasma pH, which normally lies within the narrow range of 7.35 to 7.45. The concentration of bicarbonate ion can and does vary more with alteration in acid-base balance than do chloride, sodium, or potassium ion concentrations. The body has control of the excretion of CO2 through changes in the rate and depth of respiration. Because the various forms of CO2 in plasma are in equilibrium, a change in CO2 concentration will result in a concomitant change in hydrogen ion and bicarbonate ion concentra-tions. The lungs are therefore able to compensate to some degree for abnormal production or loss of hydrogen ion. The importance of CO2 derives also from that bicarbonate ion buffering capacity, that is, its effectiveness in limiting changes in pH when acid or base enters the plasma. At first glance, the bicarbonate-carbonic acid buffer system would not appear to be particularly effective at the usual plasma pH of 7.4 since the effective pK1* of this system in plasma is 6.1 (buffers work best at pH values close to their pK). Buffering is in fact remarkably effective because the concentration of carbonic acid can be changed by changes in respiration. In brief, the addition of a strong acid to plasma (the usual direction of pH change in the body) results in excretion of the lungs of enough carbonic acid (in the form of CO2 and water) to partially correct the fall in pH. At the same time, the bicarbonate-carbonic acid buffer system becomes more effective at this lower pH. Clinical Significance: Knowledge of the CO2 con-centration permits a first approximation of the acid-base balance and goes at least part of the way toward eluci-dating the cause when an abnormality exists. Full ap-preciation of the acid-base status also requires knowledge of plasma pH, buffering capacity, hemoglobin concentra-tion, pO2 and pCO2. Nevertheless, at the present time, the determination of CO2 concentration remains the most commonly performed initial test in the evaluation of the body’s ability to control pH. Decreased Bicarbonate With Elevated pH - Respiratory Alkalosis: If the respiratory rate is in-creased, there is an increase in excretion of carbon diox-ide, and levels of plasma carbonic acid and dissolved CO2 fall. The plasma pH rises, hence the name respiratory alka-losis. The causes of this condition are varied, but they have in common the over-stimulation of that part of the brain which controls breathing (the “respiratory center”). The most common cause is simple anxiety with increased rate and depth of breathing - the so called “hyperventilation syn-drome”. Other causes include toxic substances (e.g., salicylates, as in aspirin overdosage) which stimulate the respiratory center, and central nervous system lesions such as tumors located in this part of the brain. Decreased Bicarbonate With Decreased pH - Metabolic Acidosis: The condition usually results from the addition of excess amounts of acid to the plasma. Usually these acids are products of normal metabolic reactions, but they are being formed at a faster rate that they can be degraded or excreted. For example, in dia-betic coma, increased amounts of acetoacetic acid are produced from the oxidation of fatty acids. It accumu-lates in the plasma because of the decreased ability to further metabolize it via the citric acid cycle. Most of the acetoacetic acid is reduced to beta-hydroxybutyric acid. Both of these acids are increased in the plasma in dia-betic coma. If insulin is not given to correct the metabolic abnormalities, coma and death occur. Another example of metabolic acidosis is the accumulation of lactic acid in the clinical condition called “shock”. The blood pres-sure is low and the blood supply to muscles therefore decreased. The muscles received insufficient oxygen to completely burn glucose; it is metabolized only to the point The pK1 value for the net reaction [CO2 + H2O H+ + HCO3 – ] in plasma (rather than in simple aqueous solution) is 6.1.
  • 36. 36 of formation of pyruvic and then lactic acids. Lactic acid accumulates in the plasma. In both diabetic coma and shock the bicarbonate level may be very low, and the pH may fall below 7.0. Some of the hydrogen ions from these acids combine with bicarbonate ion to form carbonic acid and then CO2 and water. The CO2 is excreted by the lungs. Thus the level of plasma bicarbonate is decreased. There is still an excess of hydrogen ion in the plasma, so the pH is low. In fact the changes in this condition can be summarized as the substitution of a strong acid (lactic or betahydroxybutyric) for a weak acid (carbonic). Metabolic acidosis also occurs in patients with kidney disease: phosphoric and sulfuric acids (produced from the breakdown of phosphate and sulfate-containing proteins) can not be removed from the plasma at the normal rate because the kidneys are damaged. Increase Bicarbonate With Decreased pH-Res-piratory Acidosis: The condition usually results from diseases of the lungs which impede the excretion of carbon dioxide. A typical example is emphysema, a dis-ease in which there is widespread destruction of lung tissue. Carbon dioxide, carbonic acid, and hydrogen and bicarbonate ions accumulate in the plasma. Increased Bicarbonate With Elevated pH-Metabolic Alkalosis: This condition can result from excess intake of sodium bicarbonate (common baking soda), a commonly used remedy for abdominal pain such as “heartburn” or pain of peptic ulcer. Metabolic alkalosis can also result from prolonged vomiting or, in the hospi-talized patient, over-use of gastric suction with loss of the acid stomach contents. There is a net loss of a strong acid, HCl, and a rise in plasma pH. The body attempts to replace the lost HCl by decreasing the excretion of CO2 by the lungs. Retention of CO2 will produce an increase in concentrations of hydrogen and bicarbonate ions, thus correcting the pH change to some extent. The net effect is to replace a strong acid, HCl, with a weak acid, car-bonic acid; therefore the pH remains abnormally high.