Review Article
MEDICAL PROGRESS
Volume 341 Number 11 (September 9, 1999) New England Journal of Medicine
1999;341:815-26.
ANTHRAX
TERRY C. DIXON, B.S., MATTHEW MESELSON, PH.D., JEANNE GUILLEMIN, PH.D., AND
PHILIP C. HANNA, PH.D.
From the Department of Microbiology, Duke University Medical Center, Durham,
N.C. (T.C.D., P.C.H.); the Department of Molecular and Cellular Biology,
Harvard University, Cambridge, Mass. (M.M.); the Department of Sociology,
Boston College, Chestnut Hill, Mass. (J.G.); and the Department of
Microbiology and Immunology, University of Michigan Medical School, Ann
Arbor (P.C.H.). Address reprint requests to Dr. Hanna at 1150 W Medical,
5641 MS II, Department of Microbiology and Immunology, University of
Michigan Medical School, Ann Arbor, MI 48104, or at [log in to unmask]
<mailto:[log in to unmask]> .
Copyright©1999, Massachusetts Medical Society. All rights reserved
ANTHRAX is an often fatal bacterial infection that occurs when Bacillus
anthracis endospores enter the body through abrasions in the skin or by
inhalation or ingestion. 1 It is a zoonosis to which most mammals,
especially grazing herbivores, are considered susceptible. Human infections
result from contact with contaminated animals or animal products, and there
are no known cases of human-to-human transmission. Human anthrax is not
common, and only one of us has seen a case. Cutaneous anthrax, the most
common form, is usually curable. A small percentage of cutaneous infections
become systemic, and these can be fatal. Systemic infection resulting from
inhalation of the organism has a mortality rate approaching 100 percent,
with death usually occurring within a few days after the onset of symptoms.
2 The rate of mortality among persons with infection resulting from
ingestion is variable, depending on the outbreak, but it may also approach
100 percent. Whatever the portal of entry, systemic anthrax involves massive
bacteremia and toxemia with nondescript initial symptoms until the onset of
hypotension, shock, and sudden death. Manifestations of advanced disease,
including shock and sudden death, are believed to result from the action of
the exotoxin complex secreted by anthrax bacilli. 1,3 The efficacy of
therapy, if initiated during the incubation period, and the rapid course of
the disease once symptoms appear make early intervention an absolute
necessity. Inglesby et al. have provided a description of the policies and
strategies for dealing with anthrax as a biologic weapon. 4 The goal of this
article is to familiarize physicians with the current understanding of the
pathogenesis, diagnosis, prevention, and treatment of anthrax.
PATHOGENESIS
Anthrax infections are initiated by endospores of B. anthracis, a
gram-positive soil organism. Anthrax endospores do not divide, have no
measurable metabolism, and are resistant to drying, heat, ultraviolet light,
gamma radiation, and many disinfectants. 5 In some types of soil, anthrax
spores can remain dormant for decades. Their hardiness and dormancy have
allowed anthrax spores to be developed as biologic weapons by a number of
nations, although their only known use in war was by the Japanese army in
Manchuria in the 1940s. 6 All known anthrax virulence genes are expressed by
the vegetative form of B. anthracis that results from the germination of
spores within the body. The course of infection and clinical manifestations
are depicted in Figure 1. Endospores introduced into the body by abrasion,
inhalation, or ingestion are phagocytosed by macrophages and carried to
regional lymph nodes. Endospores germinate inside the macrophages and become
vegetative bacteria 7,8 ; the vegetative bacteria are then released from the
macrophages, multiply in the lymphatic system, and enter the bloodstream,
until there are as many as 107 to 108 organisms per milliliter of blood,
causing massive septicemia. Once they have been released from the
macrophages, there is no evidence that an immune response is initiated
against vegetative bacilli. Anthrax bacilli express virulence factors,
including toxin and capsule. 1 The resulting toxemia has systemic effects
that lead to the death of the host. The major virulence factors of B.
anthracis are encoded on two virulence plasmids, pXO1 and pXO2. The
toxin-bearing plasmid, pXO1, is 184.5 kilobase pairs (kbp) in size and codes
for the genes that make up the secreted exotoxins. The toxin-gene complex is
composed of protective antigen, lethal factor, and edema factor. 9 The three
exotoxin components combine to form two binary toxins. Edema toxin consists
of edema factor, which is a calmodulin-dependent adenylate cyclase, 10,11
and protective antigen, the binding moiety that permits entry of the toxin
into the host cell. Increased cellular levels of cyclic AMP upset water
homeostasis and are believed to be responsible for the massive edema seen in
cutaneous anthrax. Edema toxin inhibits neutrophil. function in vitro, 12
and neutrophil function is impaired in patients with cutaneous anthrax
infection. 13 Lethal toxin consists of lethal factor, which is a zinc
metalloprotease 14-16 that inactivates mitogen-activated protein kinase
kinase in vitro, 17,18 and protective antigen, which acts as the binding
domain. Lethal toxin stimulates the macrophages to release tumor necrosis
factor a and interleukin-1b, which are partly responsible for sudden death
in systemic anthrax (Fig 1, inset). The smaller capsule-bearing plasmid,
pXO2, is 95.3 kbp in size and codes for three genes (capB, capC, and capA)
involved in the synthesis of the polyglutamyl capsule. 20 The exotoxins are
thought to inhibit the immune response mounted against infection, whereas
the capsule inhibits phagocytosis of vegetative anthrax bacilli. The
expression of all known major virulence factors is regulated by
host-specific factors such as elevated temperature and carbon dioxide
concentration, and by the presence of serum components. 21,22 Regulation of
the expression of the toxin and capsule genes is mediated by the
transcriptional activator AtxA, whose activity appears to be affected by the
previously mentioned environmental conditions. 23-25 Expression of the
capsule gene is also controlled by its own transcriptional regulator, AcpA.
26 Both plasmids are required for full virulence; the loss of either one
results in an attenuated strain. Historically, bacterial strains for anthrax
vaccine were made by rendering virulent strains free of one or both
plasmids. Pasteur, an avirulent pXO2-carrying strain, is encapsulated but
does not express exotoxin components. 1 Sterne, an attenuated strain that
carries pXO1, can synthesize exotoxin components but does not have a
capsule. 1



CLINICAL MANIFESTATIONS
Cutaneous Anthrax
Cutaneous anthrax accounts for 95 percent of all anthrax infections in the
United States. 27-30 The name anthrax (from the Greek for coal) refers to
the typical black eschar that is seen on affected areas (Fig 2). Patients
often have a history of occupational contact with animals or animal
products. The most common areas of exposure are the head, neck, and
extremities, although any area can be involved. Pathogenic endospores are
introduced subcutaneously through a cut or abrasion. There are a few case
reports of transmission by insect bites, presumably after the insect fed on
an infected carcass. 31,32 The primary skin lesion is usually a nondescript,
painless, pruritic papule that appears three to five days after the
introduction of endospores. In 24 to 36 hours, the lesion forms a vesicle
that undergoes central necrosis and drying, leaving a characteristic black
eschar surrounded by edema and a number of purplish vesicles. The edema is
usually more extensive on the head or neck than on the trunk or extremities.
33 The common description malignant pustule is actually a misnomer, because
the cutaneous lesion is not purulent and is characteristically painless. A
painful, pustular eschar in a febrile patient indicates a secondary
infection, most often with staphylococcus or streptococcus. 34 Although
cutaneous anthrax can be self-limiting, antibiotic treatment is recommended.
Lesions resolve without complications or scarring in 80 to 90 percent of
cases. Malignant edema is a rare complication characterized by severe edema,
induration, multiple bullae, and symptoms of shock. 35,36 Malignant edema
involving the neck and thoracic region often leads to breathing difficulties
that require corticosteroid therapy or intubation. A few cases have been
reported of temporal arteritis associated with cutaneous anthrax infection
and of corneal scarring from palpebral cutaneous anthrax. 37,38 Histologic
examination of anthrax skin lesions shows necrosis and massive edema with
lymphocytic infiltrates. There is no liquefaction or abscess formation,
indicating that the lesions are not suppurative. Focal points of hemorrhage
are evident, with some thrombosis. 39 Gram's staining reveals bacilli in the
subcutaneous tissue. 39



Gastrointestinal and Oropharyngeal Anthrax
Gastrointestinal anthrax, which can be fatal, has not been reported in the
United States. The symptoms appear two to five days after the ingestion of
endospore-contaminated meat from diseased animals. 40 Therefore, multiple
cases can occur within individual households. 40,41 An unusually prolonged
outbreak was attributed to the consumption of stored meat products. 42 It is
presumed that bacterial inoculation takes place at a breach in the mucosal
lining, but exactly where the endospores germinate is unknown. On
pathological examination, bacilli can be seen microscopically in the mucosal
and submucosal lymphatic tissue, and there is gross evidence of mesenteric
lymphadenitis. 43 Ulceration is always seen. It is not known whether
ulceration occurs only at sites of bacterial infection or is distributed
more diffusely as a result of the action of anthrax toxin. 43-45
Microscopical examination of affected tissues reveals massive edema and
mucosal necrosis at infected sites. 45 Inflammatory infiltrates are seen
that are similar to those in cutaneous anthrax. Gram's staining of
peritoneal fluid may reveal numerous large gram-positive bacilli. 40,46
Although mediastinal widening is considered pathognomonic of inhalational
anthrax, it has also been reported in a case of gastrointestinal anthrax. 47
Associated symptoms include fever and diffuse abdominal pain with rebound
tenderness. There are reports of both constipation and diarrhea; the stools
are either melenic or blood-tinged. 46,48 Because of ulceration of the
gastrointestinal mucosa, patients often vomit material that is blood-tinged
or has a coffeeground appearance. Ascites develops with concomitant
reduction in abdominal pain two to four days after the onset of symptoms.
The appearance of the ascites fluid ranges from clear to purulent, and it
often yields colonies of B. anthracis when cultured. Morbidity is due to
blood loss, fluid and electrolyte imbalances, and subsequent shock. Death
results from intestinal perforation or anthrax toxemia. If the patient
survives, most of the symptoms subside in 10 to 14 days. 48 Oropharyngeal
anthrax is less common than the gastrointestinal form. It is also associated
with the ingestion of contaminated meat. Initial symptoms include cervical
edema and local lymphadenopathy, which cause dysphagia and respiratory
difficulties. Lesions can be seen in the oropharynx and usually have the
appearance of pseudomembranous ulcerations. This form is milder than the
classic gastrointestinal disease and has a more favorable prognosis. 34,48
Inhalational Anthrax
Inhalational anthrax is rare, usually occurring after the inhalation of
pathogenic endospores from contaminated animal hides or products. Before the
introduction of hygienic measures in the 1960s, including vaccination,
workers in goathair mills, for example, were regularly exposed to high
concentrations of viable anthrax spores. Nevertheless, for reasons that are
not understood, few cases of inhalational anthrax occurred among them. 49-51
When dispersed in the atmosphere as an aerosol, anthrax spores can present a
respiratory hazard even far downwind from the point of release, as
demonstrated by animal tests on Gruin-ard Island in the United Kingdom,
52-55 and by an accidental release from a military biologic facility in the
city of Sverdlovsk in the former Soviet Union. 2,56-58 Inhalational anthrax
is usually fatal, even with aggressive antimicrobial therapy. It appears
that only about one fifth of those who contracted inhalational anthrax in
Sverdlovsk recovered. 2 Anthrax spores are about 1 to 2 µm in diameter, a
size that is optimal for inhalation and deposition in the alveolar spaces.
51,59-61 Although the lung is the initial site of contact, inhalational
anthrax is not considered a true pneumonia. In most but not all cases, there
is no infection in the lungs. 58,62 Rather, the endospores are engulfed by
alveolar macrophages and transported by them to the mediastinal and
peribronchial lymph nodes, with the spores germinating en route. Anthrax
bacilli multiply in the lymph nodes, causing hemorrhagic mediastinitis, and
spread throughout the body in the blood. 43,62 Data from the Sverdlovsk
outbreak indicate a modal incubation time of approximately 10 days for
inhalational anthrax. However, the onset of symptoms occurred up to six
weeks after the reported date of exposure. 2,57 Such long incubation times
presumably reflect the ability of viable anthrax spores to remain in the
lungs for many days. 51,63,64 Longer incubation periods may be associated
with smaller inocula. The initial symptoms most often reported are fever,
nonproductive cough, myalgia, and malaise, resembling those of a viral upper
respiratory tract infection. Early in the course of the disease, chest
radiographs show a widened mediastinum, which is evidence of hemorrhagic
mediastinitis, and marked pleural effusions. After one to three days, the
disease takes a fulminant course with dyspnea, strident cough, and chills,
culminating in death. 34,59 In Sverdlovsk, the mean time between the onset
of symptoms and death was 3 days (range, 1 to 10). Although accompanying
evidence of clinical signs of pneumonia in these cases is lacking, some of
the autopsies from the Sverdlovsk outbreak showed a focus of necrotizing
hemorrhagic pneumonitis, possibly at the portal of infection. 58 Submucosal
hemorrhages occurred in the trachea and bronchi, with hemorrhage and
necrosis of peribronchial lymph nodes. Hemorrhagic mediastinal lymph nodes
represent the primary lesion; however, gastrointestinal and leptomeningeal
lesions are the result of hematogenous spread. There may be wide individual
variation in susceptibility to inhalational anthrax, as suggested by
experimental studies in nonhuman primates and by the absence of persons
younger than 24 years among the 66 deaths reported in the Sverdlovsk
outbreak. 2,51,57
Anthrax Meningitis
Involvement of the meninges by B. anthracis is a rare complication of
anthrax. 65 The most common portal of entry is the skin, from which the
organisms can spread to the central nervous system by hematogenous or
lymphatic routes. Anthrax meningitis also occurs in cases of pulmonary and
gastrointestinal anthrax. 58,66 Anthrax meningitis is almost always fatal,
with death occurring one to six days after the onset of illness, despite
intensive antibiotic therapy.In the few cases in which patients have
survived, antibiotic therapy was combined with the administration of
antitoxin, prednisone, or both. 65,67 In addition to common meningeal
symptoms and nuchal rigidity, the patient has fever, fatigue, myalgia,
headache, nausea, vomiting, and sometimes agitation, seizures, and delirium.
The initial signs are followed by rapid neurologic degeneration and death.
The pathological findings are consistent with a hemorrhagic meningitis, with
extensive edema, inflammatory infiltrates, and numerous grampositive bacilli
in the leptomeninges. 43,68 The cerebrospinal fluid is often bloody and
contains many gram-positive bacilli. 69 Gross examination at autopsy finds
extensive hemorrhage of theleptomeninges, which gives them a dark red
appearance described as cardinal's cap. 58


TABLE 1
DIFFERENTIAL DIAGNOSIS OF CLINICAL MANIFESTATIONS OF ANTHRAX
MANIFESTATION
DISEASE
CAUSATIVE ORGANISM
(IF APPLICABLE)
Cutaneous Anthrax
Ecthyma gangrenosum
Pseudomonas aeruginosa

Rat-bite fever
Streptobacillus moniliformis,
Spirillum minor




Ulceroglandular tularemia
Francisella tularensis

Plague
Yersinia pestis

Glanders
Pseudomonas pseudomallei

Rickettsialpox
Rickettsia akari

Orf
Parapoxvirus

Staphylococcal lymphadenitis
Staphylococcus aureus

Cutaneous tuberculosis
Myocbacterium tuberculosis

Leprosy
Mycobacterium leprae

Buruli ulcer
Mycobacterium ulcerans
Gastrointestinal Anthrax
Typhoid
Salmonella typhi

Intestinal tularemia
Francisella tularensis

Acute gastroenteritis


Peritonitis


Mechanical obstruction


Peptic or duodenal ulcer

Inhalational Anthrax
Acute bacterial mediastinitis


Mycoplasmal pneumonia
Mycoplasma pneumoniae

Legionnaires disease
Legionella pneumophila

Psittacosis
Chlamydia psittaci

Tularemia
Francisella tularensis

Q fever
Coxiella burnetii

Viral pneumonia
Influenzavirus, hantavirus, adenovirus,
respiratory syncytial virus, cytomega-
lovirus, varicella-zoster virus

Histoplasmosis (fibrous mediastinitis)
Histoplasma capsulatum




Coccidioidomycosis
Coccidioides immitis

Ruptured aortic aneurysm


Superior vena cava syndrome


Silicosis


Sarcoidosis

Meningeal Anthrax
Subarachnoid hemorrhage
Bacterial meningitis
Aseptic meningitis


 DIAGNOSIS
Differential Diagnosis
Table 1 summarizes the differential diagnosis of anthrax. In cutaneous
anthrax, the painless, blackened, necrotic eschar is limited to the late
stages of the infection. The ulcerative eschar of cutaneous anthrax must be
differentiated from other papular lesions that present with regional
lymphadenopathy. If the lesion is purulent and the regional lymph nodes are
palpable, staphylococcal lymphadenitis is the most likely cause, although
cutaneous anthrax lesions can be superinfected with pyogenic bacteria. 70
The initial symptoms of inhalational anthrax are nondescript or flulike and
are similar to those of atypical pneumonia from other causes. The prognosis
is improved if early treatment is implemented, so that a high level of
suspicion is necessary if there is a chance of exposure to anthrax. The
cardiopulmonary collapse associated with a history of radiographic evidence
of mediastinal widening in the late stages of inhalational anthrax must be
differentiated from cardiovascular collapse with noninfectious causes, such
as dissecting or ruptured aortic aneurysm and the superior vena cava
syndrome. Anthrax infection is unusual in that mediastinal changes can be
detected early in the course of infection by chest radiography, although
similar pictures can be seen in acute bacterial mediastinitis and fibrous
mediastinitis due to Histoplasma capsulatum. 71 Less specific findings
include pleural effusions and radiographic evidence of pulmonary edema.
Silicosis, siderosis, alveolar proteinosis, and sarcoidosis are often
alternative causes of chronic mediastinitis in patients with the relevant
occupational history and previous chest radiographs demonstrating
long-standing mediastinal widening. When ingestion of contaminated meat is
suspected, the symptoms of an acute abdomen should be considered as possible
early signs of intestinal anthrax infection. Hemorrhagic meningitis caused
by anthrax must be distinguished from subarachnoid hemorrhage by computed
tomography without contrast. To distinguish hemorrhagic meningitis caused by
B. anthracis from that caused by other bacteria, Grams staining and culture
of cerebrospinal fluid should be performed. 68 In addition to the above
indictors, the clinician should consider anthrax if there is a history of
contact with materials that may be contaminated with spores, such as
infected farm animals and imported hides, or of travel to places where
anthrax is endemic. Because of the remote possibility of an anthrax aerosol
attack, clinicians should be alert to any sudden deaths of previously
healthy persons from undiagnosed disease and report them promptly to the
Centers for Disease Control and Prevention and other appropriate public
health officials.
Bacteriologic Tests
B. anthracis is a nonmotile, gram-positive, aerobic rod 1.2 to 10 µm in
length and 0.5 to 2.5 µm in width that is capable of forming central or
terminal spores. It is part of the B. cereus group of bacilli, which
consists of B. cereus, B. anthracis, B. thuringiensis, and B. mycoides. 73
The bacteria in this group tend to be dismissed by clinical microbiology
laboratories as contaminants unless the physician specifically requests
testing. 73 Except for B. anthracis, all members of this group are resistant
to penicillin because they produce chromosomally encoded betalactamases. 74
B. anthracis is easy to differentiate from other members of the B. cereus
group by observing the morphologic features of the colony on a blood-agar
plate. Colonies of most B. anthracis isolates are nonhemolytic and are white
to gray, often looking like ground glass. 75 The unusually tenacious
colonies are able to retain their shape when manipulated. When inoculated
onto nutrient agar containing 0.7 percent bicarbonate and grown overnight at
37°C in the presence of 5 to 20 percent carbon dioxide, B. anthracis will
form its characteristic poly-D-glutamic acid capsule. 76 These colonies have
a mucoid appearance, and the capsule can be demonstrated microscopically in
a colony smear stained with McFadyean's polychrome methylene blue or India
ink. 75 Blood samples obtained from patients late in the course of infection
and stained in the same manner will reveal large numbers of encapsulated
bacilli. Bacilli can also be observed in and cultured from ascites fluid,
pleural effusions, cerebrospinal fluid (in cases of meningitis), 77 and
fluid carefully expressed from the eschar, although expressing eschar fluid
is not recommended because it can cause dissemination of the pathogen. 78
Patients with systemic disease often die before positive blood cultures can
be obtained, making early diagnosis and treatment crucial. If the samples
are likely to be contaminated with other bacillus species,
polymyxin-lysozyme-EDTA-thallous acetate agar is used as a selective medium
for B. anthracis. 79 The API 50 CH test strip (API Laboratory Products,
Plainview, N.Y.) can be used in conjunction with the API 20E test strip to
identify a number of bacillus species, including B. anthracis. 80 Blood
cultures in cases of systemic anthrax infection are almost always positive,
because of the large numbers of bacterial cells in the circulation. 1
Cultures of tissue from skin lesions, however, are not useful
diagnostically, because the rate of positive cultures does not exceed 60 to
65 percent, probably owing to the use of antimicro-bial therapy or the
microbicidal activity of local antagonistic skin flora. 81 There are reports
of clinical isolates of B. anthracis that are resistant to penicillin. 31,82
Because of the potential for drug-resistant strains, including deliberately
modified strains, antibiotic-susceptibility testing should be performed on
all isolates.
 Serologic and Immunologic Tests
The major immunogenic proteins of B. anthracis appear to be capsular
antigens and the exotoxin components. Specific enzyme-linked immunosorbent
assays (ELISAs) that show a quadrupling of the titer of antibodies against
these components are diagnostic of past infection or vaccination. The most
reliable indicators are the titers of antibody to protective antigen and to
capsular components. 73,83,84 In studies of the measurement of antibody
titers by ELISA, the sensitivity of possible indicators was as follows: 72
percent for protective antigen, 95 to 100 percent for capsule antigens, 42
percent for lethal factor, and 26 percent for edema factor. 85 Enzyme-linked
immunoelectrotransfer blotting provided a higher specificity when used in
conjunction with ELISA-based testing. 85 Indirect microhemagglutination
gives results similar to those obtained with ELISA but has certain
drawbacks, including the short shelf life of antigen-sensitized red-cell
preparations, the limited reproducibility of the test, and longer
preparation times. 86 Immunologic detection of the exotoxins in blood during
systemic infection is possible with similar tests if antibodies to anthrax
toxins are available, but those tests are unreliable for diagnosis. Thus,
although these tests are of epidemiologic value, they have little diagnostic
value in acute illness. 83 During systemic infections, antibodies to toxin
or capsular components cannot be detected until late in the course of the
disease, often when it is too late to initiate treatment. 73 In treated
infections, no increase in the antitoxin antibody titer is seen. The
anthraxin skin test, consisting of subdermal injection of a commercially
produced chemical extract of an attenuated strain of B. anthracis, is
available for the diagnosis of acute and previous cases of anthrax. 81,87,88
In one study the skin test diagnosed 82 percent of cases one to three days
after the onset of symptoms and 99 percent of cases by the end of the fourth
week. 81 The skin test may be suitable for both rapid diagnosis of acute
cases and the retrospective analysis of anthrax infections.
New Molecular Diagnostic Methods
New diagnostic techniques have focused on the use of the polymerase chain
reaction to amplify markers specific to B. anthracis or the B. cereus group.
Two markers, vrrA 89 and Ba813, 90-92 have been the subject of extensive
study. Other methods using the polymerase chain reaction to amplify specific
virulence plasmid markers harbored by different anthrax strains may soon
become available. 56,93-96 These new rapid methods may become useful in the
clinical setting, where early diagnosis is crucial.

PREVENTION AND TREATMENT
Prophylaxis, Vaccination, and Decontamination
Prophylaxis for asymptomatic patients with suspected exposure to anthrax
spores can be achieved with a six week course of doxycycline or
ciprofloxacin. If the suspected dose of spores is high, a longer course of
antibiotics is warranted. Extended treatment is needed for total pulmonary
clearance of spores, which are not affected by the presence of antibiotics.
63,97 The standard anthrax vaccine in the United States is approved by the
Food and Drug Administration and is routinely administered to persons at
risk for exposure to anthrax spores. The existing supplies are currently
being used to immunize all military personnel. Designated anthrax vaccine
adsorbed (AVA), it is an aluminum hydroxide-precipitated preparation of
protective antigen from attenuated, nonencapsulated B. anthracis cultures of
the Sterne strain. 98,99 Two inoculations with AVA afforded substantial
protection against inhalational anthrax in rhesus monkeys, 100 and a limited
trial of a similar vaccine in humans indicated that it afforded considerable
protection against cutaneous anthrax. 101 AVA is administered subcutaneously
in a 0.5-ml dose that is repeated at 2 and 4 weeks and at 6, 12, and 18
months. 102 Boosters are then given annually. For those receiving antibiotic
prophylaxis for suspected exposure, AVA may be given concurrently. There is
a need for vaccines with better protection and a simpler schedule. Vaccines
now being tested include preparations of protective antigen subunits with
different adjuvants, protective antigen purified from recombinant sources,
and live vaccines based on anthrax strains with auxotrophic mutations.
103-113 Live attenuated endospore-based vaccines were widely used in the
Soviet Union for both humans and livestock and remain in use in the Russian
Federation today. 103 The ability of any vaccine to protect humans in the
event of aerosol attack, as in biologic terrorism or warfare, cannot be
tested directly and therefore must remain a concern. 114 A textile mill
contaminated with anthrax spores was decontaminated with vaporized
formaldehyde, 115 and soil decontamination at Gruinard Island was achieved
with formaldehyde in seawater. 116 Although decontamination is desirable,
the risk that resuspension of a deposited aerosol will lead to inhalational
anthrax is much less than the risk due to a primary aerosol. 117,118
Autoclaving and incineration are acceptable procedures for the
decontamination of laboratory materials.



TABLE 2
Pharmacologic Therapy for Bacillus anthracis Infection and Its Sequelae*
Therapy
Dosage for Adults
Dosage for Children
Treatment of Infection†


Penicillin V
200-500 mg orally 4 times/day
25-50 mg/kg of body weight/day orally in divided doses 2 or 4 times/day
Penicillin G
8 million-12 million U total, intravenously in divided doses every 4-6 hr,
100,000-150,000 U/kg/day in divided doses every 4-6 hr
Streptomycin
30 mg/kg intramuscularly or intravenously per day - gentamicin can also be
used (in conjunction with penicillin)

Tetracycline
250-500 mg orally or intravenously 4 times/day
Tetracycline is not approved for children
Doxycycline
200 mg orally or intravenously as a loading dose, then 50-100 mg every 12 hr
Doxycycline is not approved for children <9 yr old For children Ç45 kg: 2.5
mg/kg every 12 hr For children 45 kg: use adult dosage
Erythromycin
250 mg orally every 6 hr
40/mg/kg/day orally in divided doses every 6 hr
Erythromycin lactobionate
15-20 mg/kg (maximum, 4 g) intravenously per day
20-40 mg/kg/day intravenously in divided doses every 6 hr (1- to 2-hr
infusion)
Chloramphenicol
50-100 mg/kg/day orally or intravenously in divided doses every 6 hr
50-75 mg/kg/day in divided doses every 6 hr
Ciprofloxacin
250-750 mg orally twice/day 200-400 mg intravenously every 12 hr
20-30 mg/kg/day in divided doses every 12 hr Oral or intravenous dosing is
not approved for patients <18 yr old
Prophylaxis‡


Doxycycline
100 mg orally twice/day for 4 wk

Ciprofloxacin
500 mg orally twice/day for 4 wk

Corticosteroid therapy for severe edema


Dexamethasone
0.75-0.90 mg/kg/day
orally, intravenously, or intramuscularly in divided doses every 6 hr
0.25-0.5 mg/kg every 6 hr
Prednisone
1-2 mg/kg or 5-60 mg orally/day
0.5-2 mg/kg/day
*Most B. anthracis strains are resistant to cefuroxime in vitro.
†For inhalational, gastrointestinal, or meningeal anthrax infection in
adults, the intravenous regimen is used with peni- cillin G, streptomycin,
tetracycline, doxycycline, erythromycin lactobionate, chloramphenicol, and
ciprofloxacin; for these infections in children, the intravenous regimen is
used with penicillin G, doxycycline, erythromycin lactobionate, and
chloramphenicol.
‡If patient is unvaccinated, begin initial doses of vaccine.


Treatment
Antibiotics
Table 2 summarizes pharmacologic therapy for anthrax. Penicillin and
doxycycline are used for the treatment of anthrax. Intravenous
administration is recommended in cases of inhalational, gastrointestinal,
and meningeal anthrax. Cutaneous anthrax with signs of systemic involvement,
extensive edema, or lesions on the head and neck also requires intravenous
therapy. Streptomycin had a synergistic effect with penicillin in
experiments and may also be given for inhalational anthrax. Despite early
and vigorous treatment, the prognosis of patients with inhalational,
gastrointestinal, or meningeal anthrax remains poor. Antibiotic therapy
should be continued for at least 14 days after symptoms abate. 67,78 In
cutaneous anthrax, treatment with oral penicillin renders lesions sterile
after 24 hours, although they still progress to eschar formation.
Chloramphenicol, erythromycin, tetracycline, or ciprofloxacin can be
administered to patients who are allergic to penicillin. If resistance to
penicillin and doxycycline is suspected and antibiotic-susceptibility data
are not available, ciprofloxacin may be administered empirically.
Doxycycline and tetracycline are not recommended for pregnant women or
children, and the effects of ciprofloxacin in pregnant women have not been
determined. 4 For culturing cutaneous lesions, gentle sampling with a moist,
sterile applicator is preferred. Excision of the eschar is contraindicated
and might hasten systemic dissemination. Lesions should be covered with
sterile dressings that are changed regularly. Soiled dressings should be
autoclaved and properly disposed of. In cases of extensive edema,
meningitis, or swelling in the head-and-neck region, corticosteroid therapy
should be initiated. 119,120 Supportive therapy should be initiated to
prevent septic shock and fluid and electrolyte imbalance, and to maintain
airway patency.
Potential New Treatments
The current understanding that anthrax is a toxigenic condition suggests the
potential of antitoxin
therapy. The central importance of lethal toxin is supported by much
research. Early experiments in which antibiotics were administered to
animals at different stages of infection found a principle of no return;
once the infection had reached a certain point, the animal was doomed, even
after removal of the microbes. Test animals injected intravenously with
purified lethal toxin died in a manner very similar to that of animals that
died of the natural infection. 3,15,19 Lethal-toxin-deficient strains are
highly attenuated. 121,122 Prior immunity (passive or active) to the
lethal-toxin proteins protects animals from endospore challenge. 63,123
Finally, toxin-affected macrophages produce the proinflammatory cytokines
that mediate the shock and sudden death that occur in anthrax. 3,15,19
Unfortunately, antitoxin preparations are not currently available in the
United States. In addition, the recent discovery that lethal toxin acts as a
zinc metalloprotease inside target cells and the identification of potential
target substrates may provide new insights for use in designing drugs that
directly inhibit the toxicity of lethal factor in vivo. 14,17,18824 á
September 9, 1999
FUTURE CHALLENGES
Anthrax holds an important place in the development of modern medicine and
has long been intertwined with human history. Anthrax is believed to have
been one of the Egyptian plagues at the time of Moses, and cases were
clearly recorded by the ancient Romans. 124 The anthrax bacillus was the
model first used in the development of Koch's postulates and is considered
the first germ proved to cause human disease. 125 Pasteur later generated a
capsule-null anthrax strain that was the first vaccine made from live
attenuated bacteria for use in humans. 126 At the birth of cellular
immunology, Metchnikoff used the anthrax bacillus to examine the ability of
his newly discovered macrophages to kill microbes. 127 Today, investigators
are using B. anthracis and its toxins in an attempt to understand early
events in the infectious process and the molecular basis of inflammation.
3,15,19 Unfortunately, new issues have arisen beyond those related to
scientific inquiry. No casualty-producing terrorist use of anthrax has
occurred, and the Federal Bureau of Investigation has stated that it has no
intelligence that state sponsors of terrorism, international terrorist
groups, or domestic terrorist groups are currently planning to use these
deadly weapons in the United States. 128 However, the incidence of hoaxes
has greatly increased with recent publicity about anthrax, providing a
challenge to law enforcement. 129 Recent revelations regarding the
development of anthrax weapons by the former Soviet Union and by Iraq, and
of attempts to develop such weapons by the Aum Shinrikyo cult in Japan, make
the potential use of B. anthracis in biologic terrorism a legitimate
concern. 4,129 New strains resistant to antibiotics or containing additional
virulence factors could be misused with the intent of confounding treatment
or prophylaxis. 114,130 Whether our medical system would be able to provide
appropriate prophylaxis and therapy in the event of a large-scale exposure
to pathogenic endospores remains uncertain, even doubtful. It has now become
relevant for physicians to re-familiarize themselves with clinical anthrax.
Supported in part by grants (AI-08649 and AI-40644) and a Medical Scientist
Training award from the National Institutes of Health, by a grant (IRG-158
K) from the American Cancer Society, and by Duke University Medical Center.
We are indebted to Arthur Friedlander, M.D., Julia Chosy, Tanya Dixon, John
Ireland, Matthew Weiner, and Kenneth Alexander, M.D., Ph.D., for their
reading and critical discussion of the manuscript.
REFERENCES
1. Hanna P. Anthrax pathogenesis and host response. Curr Top Microbiol
Immunol 1998;225:13-35.
2. Meselson M, Guillemin J, Hugh-Jones M, et al. The Sverdlovsk anthrax
outbreak of 1979. Science 1994;266:1202-8.
3. Hanna PC, Acosta D, Collier RJ. On the role of macrophages in anthrax.
Proc Natl Acad Sci U S A 1993;90:10198-201.
4. Inglesby TV, Henderson DA, Bartlett JG, et al. Anthrax as a biological
weapon: medical and public health management. JAMA 1999;281:1735-45.
5. Watson A, Keir D. Information on which to base assessments of risk from
environments contaminated with anthrax spores. Epidemiol Infect
1994;113:479-90.
6. Harris SH. Factories of death: Japanese secret biological warfare,
1932-1945, and the American cover-up. London: Routledge, 1994.
7. Ross JM. The pathogenesis of anthrax following the administration of
spores by the respiratory route. J Pathol Bacteriol 1957;73:485-94.
8. Guidi-Rontani C, Weber-Levy M, Labruyere E, Mock M. Germination of
Bacillus anthracis spores within alveolar macrophages. Mol Microbiol
1999;31:9-17.
9. Leppla SH. The anthrax toxin complex. In: Alouf J, Freer JH,
eds.Sourcebook of bacterial protein toxins. London: Academic Press,
1991:277-302.
10. Idem. Anthrax toxin edema factor: a bacterial adenylate cyclase that
increases cyclic AMP concentrations of eukaryotic cells. Proc Natl Acad Sci
U S A 1982;79:3162-6.
11. Idem. Bacillus anthracis calmodulin-dependent adenylate cyclase:
chemical and enzymatic properties and interactions with eucaryotic cells.
Adv Cyclic Nucleotide Protein Phosphorylation Res 1984;17:189-98.
12. O'Brien J, Friedlander A, Dreier T, Ezzell J, Leppla S. Effects of
anthrax toxin components on human neutrophils. Infect Immun 1985;47:306-10.
13. Alexeyev OA, Morozov VG, Suzdaltseva TV, Mishukov AS, Steinberg LA.
Impaired neutrophil function in the cutaneous form of anthrax. Infection
1994;22:281-2.
14. Hammond SE, Hanna PC. Lethal factor active-site mutations affect
catalytic activity in vitro. Infect Immun 1998;66:2374-8.
15. Hanna PC, Kruskal BA, Ezekowitz RA, Bloom BR, Collier RJ. Role of
macrophage oxidative burst in the action of anthrax lethal toxin. Mol Med
1994;1:7-18.
16. Klimpel KR, Arora N, Leppla SH. Anthrax toxin lethal factor contains a
zinc metalloprotease consensus sequence which is required for lethal toxin
activity. Mol Microbiol 1994;13:1093-100.
17. Duesbery NS, Webb CP, Leppla SH, et al. Proteolytic inactivation of
MAP-kinase-kinase by anthrax lethal factor. Science 1998;280:734-7.
18. Vitale G, Pellizzari R, Recchi C, Napolitani G, Mock M, Montecucco C.
Anthrax lethal factor cleaves the N-terminus of MAPKKs and induces
tyrosine/threonine phosphorylation of MAPKs in cultured macrophages. Biochem
Biophys Res Commun 1998;248:706-11.
19. Hanna PC, Kochi S, Collier RJ. Biochemical and physiological changes
induced by anthrax lethal toxin in J774 macrophage-like cells. Mol Biol Cell
1992;3:1269-77.
20. Makino S-I, Uchida I, Terakado N, Sasakawa C, Yoshikawa M. Molecular
characterization and protein analysis of the cap region, which is essential
for encapsulation in Bacillus anthracis. J Bacteriol 1989;171:722-30.
21. Makino S, Sasakawa C, Uchida I, Terakado N, Yoshikawa M. Cloning and
CO2 -dependent expression of the genetic region for encapsulation from
Bacillus anthracis. Mol Microbiol 1988;2:371-6.
22. Dai Z, Sirard J-C, Mock M, Koehler TM. The atxA gene product activates
transcription of the anthrax toxin genes and is essential for virulence. Mol
Microbiol 1995;16:1171-81.
23. Dai Z, Koehler TM. Regulation of anthrax toxin activator gene (atxA)
expression in Bacillus anthracis: temperature, not CO2/bicarbonate, affects
AtxA synthesis. Infect Immun 1997;65:2576-82.
24. Uchida I, Hornung JM, Thorne CB, Klimpel KR, Leppla SH. Cloning and
characterization of a gene whose product is a transactivator of anthrax
toxin synthesis. J Bacteriol 1993;175:5329-38.
25. Uchida I, Makino S, Sekizaki T, Terakado N. Cross-talk to the genes for
Bacillus anthracis capsule synthesis by atxA, the gene encoding the
transactivator of anthrax toxin synthesis. Mol Microbiol 1997;23:1229-40.
26. Vietri NJ, Marrero R, Hoover TA, Welkos SL. Identification and
characterization of a transactivator involved in the regulation of
encapsulation by Bacillus anthracis. Gene 1995;152:1-9.
27. Taylor JP, Dimmitt DC, Ezzell JW, Whitford H. Indigenous human cutaneous
anthrax in Texas. South Med J 1993;86:1-4.
28. Human cutaneous anthrax North Carolina, 1987. Arch Dermatol
1988;124:1324.
29. Leads from the MMWR: human cutaneous anthrax -- North Carolina, 1987.
JAMA 1988;260:616.
30. Human cutaneous anthrax -- North Carolina, 1987. MMWR Morb Mortal Wkly
Rep 1988;37:413-4.
31. Bradaric N, Punda-Polic V. Cutaneous anthrax due to penicillin-resistant
Bacillus anthracis transmitted by an insect bite. Lancet 1992;340:306-7.
32. Turell MJ, Knudson GB. Mechanical transmission of Bacillus anthracis by
stable flies (Stomoxys calcitrans) and mosquitoes (Aedes aegypti and Aedes
taeniorhynchus). Infect Immun 1987;55:1859-61.
33. Smego RA Jr, Gebrian B, Desmangels G. Cutaneous manifestations of
anthrax in rural Haiti. Clin Infect Dis 1998;26:97-102.
34. Edwards MS. Anthrax. In: Feigin RD, Cherry JD, eds. Textbook of
pediatric infectious diseases. 3rd ed. Vol. 1. Philadelphia: W.B. Saunders,
1992:1053-6.
35. Doganay M, Bakir M, Dokmetas I. A case of cutaneous anthrax with
toxaemic shock. Br J Dermatol 1987;117:659-62.
36. Kutluk MT, Secmeer G, Kanra G, Celiker A, Aksoyek H. Cutaneous anthrax.
Cutis 1987;40:117-8.
37. Doganay M, Aygen B, Inan M, Kandemir O, Turnbull P. Temporal artery
inflammation as a complication of anthrax. J Infect 1994;28:311-4.
38. Yorston D, Foster A. Cutaneous anthrax leading to corneal scarring from
cicatricial ectropion. Br J Ophthalmol 1989;73:809-11.
39. Mallon E, McKee PH. Extraordinary case report: cutaneous anthrax.Am J
Dermatopathol 1997;19:79-82.
40. LaForce FM. Anthrax. Clin Infect Dis 1994;19:1009-14.
41. de Lalla F, Ezzell JW, Pellizzer G, et al. Familial outbreak of
agriculture anthrax in an area of northern Italy. Eur J Clin Microbiol
Infect Dis 1992;11:839-42.
42. Batykin V, Vygodchikov G, Sazshina Y. Outbreak of intestinal anthrax in
Yarolslavl. J Hyg Epidemiol 1929;1:25-30.
43. Dutz W, Kohout E. Anthrax. Pathol Annu 1971;6:209-48.
44. Dutz W, Saidi F, Kohout E. Gastric anthrax with massive ascites. Gut
1970;11:352-4.
45. Dutz W, Kohout-Dutz E. Anthrax. Int J Dermatol 1981;20:203-6.
46. Nalin DR, Sultana B, Sahunja R, et al. Survival of a patient with
intestinal anthrax. Am J Med 1977;62:130-2.
47. Paulet R, Caussin C, Coudray JM, Selcer D, de Rohan Chabot P. Forme
viscerale de charbon humain importee d'Africa. Presse Med 1994; 23:477-8.
48. Alizad A, Ayoub EM, Makki N. Intestinal anthrax in a two-year-old child.
Pediatr Infect Dis J 1995;14:394-5.
49. Dahlgren CM, Buchanan LM, Decker HM, Freed SW, Phillips CR, Brachman PS.
Bacillus anthracis aerosols in goat hair processing mills. Am J Hyg
1960;72:24-31.
50. Albrink WS, Brooks SM, Biron RE, Kopel M. Human inhalation anthrax: a
report of three fatal cases. Am J Pathol 1960;36:457-71.
51. Brachman PS, Kaufman AF, Dalldorf FG. Industrial inhalation anthrax.
Bacteriol Rev 1966;30:646-59.
52. Manchee RJ, Broster MG, Henstridge RM, Stagg AJ, Melling J. Anthrax
island. Nature 1982;296:598.
53. Manchee RJ. Bacillus anthracis on Gruinard Island. Nature 1981;294:
254-5.
54. Sterne M. Anthrax island. Nature 1982;295:362.
55. Wynn J. Anthrax island: why worry? Nature 1982;298:506-7.
56. Jackson PJ, Hugh-Jones ME, Adair DM, et al. PCR analysis of tissue
samples from the 1979 Sverdlovsk anthrax victims: the presence of multiple
Bacillus anthracis strains in different victims. Proc Natl Acad Sci U S A
1998;95:1224-9.
57. Guillemin J. Anthrax: the investigation of a deadly outbreak. Berkeley:
University of California Press (in press).
58. Abramova FA, Grinberg LM, Yampolskaya OV, Walker DH. Pathology of
inhalational anthrax in 42 cases from the Sverdlovsk outbreak of 1979. Proc
Natl Acad Sci U S A 1993;90:2291-4.
59. Penn CC, Klotz SA. Anthrax pneumonia. Semin Respir Infect 1997;
12:28-30.
60. Brachman PS. Inhalation anthrax. Ann N Y Acad Sci 1980;353:83-93.
61. Idem. Anthrax. Ann N Y Acad Sci 1970;174:577-82.
62. Albrink WS. Pathogenesis of inhalation anthrax. Bacteriol Rev 1961;
25:268-73.
63. Friedlander AM, Welkos SL, Pitt ML, et al. Postexposure prophylaxis
against experimental inhalation anthrax. J Infect Dis 1993;167:1239-43.
64. Henderson DW, Peacock S, Belton FC. Observations on the prophylaxis of
experimental pulmonary anthrax in the monkey. J Hyg 1956;54: 28-36.
65. Tahernia AC, Hashemi G. Survival in anthrax meningitis. Pediatrics
1972;50:329-33.
66. Tabatabaie P, Syadati A. Bacillus anthracis as a cause of bacterial
meningitis. Pediatr Infect Dis J 1993;12:1035-7.
67. Tahernia AC. Treatment of anthrax in children. Arch Dis Child 1967;
42:181-2.
68. Rangel RA, Gonzalez DA. Bacillus anthracic meningitis. Neurology
1975;25:525-30.
69. Pluot M, Vital C, Aubertin J, Croix JC, Pire JC, Poisot D. Anthrax
meningitis: report of two cases with autopsies. Acta Neuropathol (Berl)
1976;36:339-45.
70. Aksaray N, Cinaz P, Coskun U, Serbest M, Koksal F. Cutaneous anthrax.
Trop Geogr Med 1990;42:168-71.
71. Wheat J. Histoplasma. In: Gorbach SL, Bartlett JG, Blacklow NR,
eds.Infectious diseases. 2nd ed. Philadelphia: W.B. Saunders, 1998:2335-44.
72. Claus D, Berkeley RCW. Genus Bacillus. In: Sneath PHA, ed. Bergey's
manual of systematic bacteriology. Vol. 2. Baltimore: Williams & Wilkins,
1986:1105-39.
73. Turnbull PCB, Kramer JM. Bacillus. In: Balows A, ed. Manual of clinical
microbiology. 5th ed. Washington, D.C.: American Society for Microbiology,
1991:296-303.
74. Penn CC, Klotz SA. Bacillus anthracis and other aerobic spore formers.
In: Gorbach SL, Bartlett JG, Blacklow NR, eds. Infectious diseases. 2nd ed.
Philadelphia: W.B. Saunders, 1998:1747-50.
75. Parry JM, Turnbull PCB, Gibson JR. A color atlas of Bacillus species.
London: Wolfe Medical, 1983:272.
76. Green BD, Battisti L, Koehler TM, Thorne CB, Ivins BE. Demonstration of
a capsule plasmid in Bacillus anthracis. Infect Immun 1985;49:291-7.
77. Penn CC, Klotz SA. Anthrax. In: Gorbach SL, Bartlett JG, Blacklow NR,
eds. Infectious diseases. 2nd ed. Philadelphia: W.B. Saunders, 1998: 1575-8.
78. Gold H. Treatment of anthrax. Fed Proc 1967;26:1563-8.
79. Knisely RF. Selective medium for Bacillus anthracis. J Bacteriol 1966;
92:784-6.
80. Logan NA, Carman JA, Melling J, Berkeley RCW. Identification of Bacillus
anthracis by API tests. J Med Microbiol 1985;20:75-85.
81. Shlyakhov E, Rubinstein E. Evaluation of the anthraxin skin test for
diagnosis of acute and past human anthrax. Eur J Clin Microbiol Infect Dis
1996;15:242-5.
82. Lalitha MK, Thomas MK. Penicillin resistance in Bacillus anthracis.
Lancet 1997;349:1522.
83. Harrison LH, Ezzell JW, Abshire TG, Kidd S, Kaufmann AF. Evaluation of
serologic tests for diagnosis of anthrax after an outbreak of cutaneous
anthrax in Paraguay. J Infect Dis 1989;160:706-10.
84. Turnbull PC, Doganay M, Lindeque PM, Aygen B, McLaughlin J. Serology and
anthrax in humans, livestock and Etosha National Park wildlife. Epidemiol
Infect 1992;108:299-313.
85. Sirisanthana T, Nelson KE, Ezzell JW, Abshire TG. Serological studies of
patients with cutaneous and oraloropharyngeal anthrax from northern
Thailand. Am J Trop Med Hyg 1988;39:575-81.
86. Johnson-Winegar A. Comparison of enzyme-linked immunosorbent and
indirect hemagglutination assays for determining anthrax antibodies. J Clin
Microbiol 1984;20:357-61.
87. Pfisterer RM. Eine Milzbrandepidemie in der Schweiz: Klinische,
diagnostische und epidemiologische Aspekte einer weitgehend vergessenen
Krankheit. Schweiz Med Wochenschr 1991;121:813-25.
88. Shlyakhov E, Rubinstein E, Novikov I. Anthrax post-vaccinal
cell-mediated immunity in humans: kinetics pattern. Vaccine 1997;15:631-6.
89. Andersen GL, Simchock JM, Wilson KH. Identification of a region of
genetic variability among Bacillus anthracis strains and related species. J
Bacteriol 1996;178:377-84.
90. Patra G, Vaissaire J, Weber-Levy M, Le Doujet C, Mock M. Molecular
characterization of Bacillus strains involved in outbreaks of anthrax in
France in 1997. J Clin Microbiol 1998;36:3412-4.
91. Patra G, Sylvestre P, Ramisse V, Therasse J, Guesdon JL. Isolation of a
specific chromosomic DNA sequence of Bacillus anthracis and its possible use
in diagnosis. FEMS Immunol Med Microbiol 1996;15:223-31.
92. Ramisse V, Patra G, Garrigue H, Guesdon JL, Mock M. Identification and
characterization of Bacillus anthracis by multiplex PCR analysis of
sequences on plasmids pXO1 and pXO2 and chromosomal DNA. FEMS Microbiol Lett
1996;145:9-16.
93. Keim P, Kalif A, Schupp J, et al. Molecular evolution and diversity in
Bacillus anthracis as detected by amplified fragment length polymorphism
markers. J Bacteriol 1997;179:818-24.
94. Klevytskya A, Schupp J, Price LB, et al. A multiplexed fluorescent PCR
genotyping system based on ten highly informative single-locus markers for
B. anthracis strain identification. Presented at the Third International
Conference on Anthrax, Plymouth, England, 1998. abstract.
95. Price LB, Hugh-Jones M, Jackson PJ, Keim P. Genetic diversity in the
protective antigen gene of Bacillus anthracis. J Bacteriol 1999;181:2358-62.
96. Jackson PJ, Walthers EA, Kalif AS, et al. Characterization of the
variable-number tandem repeats in vrrA from different Bacillus anthracis
isolates. Appl Environ Microbiol 1997;63:1400-5.
97. Vancurik J. Causes of the failure of antibiotic prophylaxis of
inhalation anthrax and clearance of the spores from the lungs. Folia
Microbiol (Praha) 1966;11:459-64.
98. Vaccine against anthrax. BMJ 1965;5464:717-8.
99. Anthrax vaccine. Med Lett Drugs Ther 1998;40:52-3.
100. Ivins BE, Fellows P, Pitt ML, et al. Efficacy of standard human anthrax
vaccine against Bacillus anthracis aerosol spore challenge in rhesus
monkeys. Salisbury Med Bull 1996;87:125-6.
101. Brachman PS, Gold H, Plotkin SA, Fekety FR, Werrin M, Ingraham NR.
Field evaluation of a human anthrax vaccine. Am J Public Health
1962;52:632-45.
102. Franz DR, Jahrling PB, Friedlander AM, et al. Clinical recognition and
management of patients exposed to biological warfare agents. JAMA
1997;278:399-411.
103. Shlyakhov EN, Rubinstein E. Human live anthrax vaccine in the former
USSR. Vaccine 1994;12:727-30.
104. Pezard C, Weber M, Sirard JC, Berche P, Mock M. Protective immunity
induced by Bacillus anthracis toxin-deficient strains. Infect Immun
1995;63:1369-72.
105. Pezard C, Sirard JC, Mock M. Protective immunity induced by Bacillus
anthracis toxin mutant strains. Adv Exp Med Biol 1996;397:69-72.
106. Miller J, McBride BW, Manchee RJ, Moore P, Baillie LW. Production and
purification of recombinant protective antigen and protective efficacy
against Bacillus anthracis. Lett Appl Microbiol 1998;26:56-60.
107. Little SF, Knudson GB. Comparative efficacy of Bacillus anthracis live
spore vaccine and protective antigen vaccine against anthrax in the guinea
pig. Infect Immun 1986;52:509-12.
108. Ivins BE, Welkos SL, Knudson GB, Little SF. Immunization against
anthrax with aromatic compound-dependent (Aro-) mutants of Bacillus
anthracis and with recombinant strains of Bacillus subtilis that produce
anthrax protective antigen. Infect Immun 1990;58:303-8.
109. Ivins BE, Welkos SL, Little SF, Crumrine MH, Nelson GO. Immunization
against anthrax with Bacillus anthracis protective antigen combined with
adjuvants. Infect Immun 1992;60:662-8.
110. Ivins BE, Fellows PF, Nelson GO. Efficacy of a standard human anthrax
vaccine against Bacillus anthracis spore challenge in guinea-pigs. Vaccine
1994;12:872-4.
111. Coulson NM, Fulop M, Titball RW. Bacillus anthracis protective antigen,
expressed in Salmonella typhimurium SL 3261, affords protection against
anthrax spore challenge. Vaccine 1994;12:1395-401.
112. Baillie L, Moir A, Manchee R. The expression of the protective antigen
of Bacillus anthracis in Bacillus subtilis. J Appl Microbiol 1998;84: 741-6.
113. Baillie LW, Moore P, McBride BW. A heat-inducible Bacillus subtilis
bacteriophage phi 105 expression system for the production of the protective
antigen of Bacillus anthracis. FEMS Microbiol Lett 1998;163:43-7.
114. Hanna P. How anthrax kills. Science 1998;280:1671, 1673-4.
115. Young LS, Feeley JC, Brachman PS. Vaporized formaldehyde treatment of a
textile mill contaminated with Bacillus anthracis. Arch Environ Health
1970;20:400-3.
116. Manchee RJ, Broster MG, Stagg AJ, Hibbs SE. Formaldehyde dilution
effectively inactivates spores of Bacillus anthracis on the Scottish island
of Gruinard. Appl Environ Microbiol 1994;60:4167-71.
117. Birensvige A. Inhalation hazard from reaerosolized biological agents: a
review. Aberdeen, Md.: Army Chemical Research, Development and Engineering
Center, 1992.
118. Davids DE, Lejeune AR. Secondary aerosol hazard in the field. Raulston,
Alta.: Suffield Defence Research Establishment, 1981.
119. Knudson GB. Treatment of anthrax in man: history and current concepts.
Mil Med 1986;151:71-7.
120. Doust JY, Sarkarzadeh A, Kavoossi K. Corticosteroid in treatment of
malignant edema of chest wall and neck (anthrax). Dis Chest 1968;53:773-4.
121. Cataldi A, Labruyere E, Mock M. Construction and characterization of a
protective antigen-deficient Bacillus anthracis strain. Mol Microbiol
1990;4:1111-7.
122. Pezard C, Berche P, Mock M. Contribution of individual toxin components
to virulence of Bacillus anthracis. Infect Immun 1991;59:3472-7.
123. Turnbull PC, Leppla SH, Broster MG, Quinn CP, Melling J. Antibodies to
anthrax toxin in humans and guinea pigs and their relevance to protective
immunity. Med Microbiol Immunol (Berl) 1988;177:293-303.
124. Dirckx JH. Virgil on anthrax. Am J Dermatopathol 1981;3:191-5.
125. Koch R. The aetiology of anthrax based on the ontogeny of the an-thrax
bacillus. Beitr Biol Pflanz 1877;2:277-82.
126. Pasteur L. De l'attenuation des virus et de leur retour ˆ la virulence.
C R Acad Sci III 1881;92:429-35.
127. Metchnikoff E. Immunity in infective diseases. Cambridge, England:
Cambridge University Press, 1905.
128. Ember LR. Bioterrorism: combating the threat. Chem Eng News
1999;77:8-17.
129. Bioterrorism alleging use of anthrax and interim guidelines for
management -- United States, 1998. MMWR Morb Mortal Wkly Rep 1999; 48:69-74.
130. Pomerantsev AP, Staritsin NA, Mockov Yu V, Marinin LI. Expression of
cereolysine AB genes in Bacillus anthracis vaccine strain ensures protection
against experimental hemolytic anthrax infection. Vaccine 1997;15: 1846-50.


Edward E. Rylander, M.D.
Diplomat American Board of Family Practice.
Diplomat American Board of Palliative Medicine.