Free Republic
Browse · Search
News/Activism
Topics · Post Article

Skip to comments.

Potential Biological Agents, Associated Infections, and Treatment
The Institute for Molecular Medicine ^ | 8 October, 2001 | Dr. Damian Beltran, M.D.

Posted on 10/07/2001 11:31:54 PM PDT by samantha06

There are a number of considerations that should be taken into account when undergoing therapy for Biological Contamination or immunizing your body against the possibility of infection. A few are mentioned below, and some product examples are given. The Institute for Molecular Medicine is a nonprofit institution and does not endorse commercial products. The products mentioned below are only examples of the types of substances that could be beneficial to patients.


TOPICS: Breaking News; News/Current Events
KEYWORDS:

1 posted on 10/07/2001 11:31:54 PM PDT by samantha06
[ Post Reply | Private Reply | View Replies]

Comment #2 Removed by Moderator

Barely scratches the surface. There are dozens of potential biowarfare agents.
3 posted on 10/08/2001 12:36:39 AM PDT by dbbeebs
[ Post Reply | Private Reply | To 2 | View Replies]

To: dbbeebs

 

The Biological Threat

Why should we be worried about biological terrorism?

Many of the agents used as biological weapons occur in nature and are therefore available for use among both the military and civilians. The ease with which biological weapons can be delivered also makes dissemination of these agents easy. In 1995, two members of a militia group in Minnesota were convicted of possession of ricin, a biological agent that they had produced themselves. The ease with which harmful biological agents may be obtained is probably best exemplified by the 1996 episode in which Bubonic plague cultures were obtained through the postal service, by a man in Ohio.

By dividing every 20 minutes, a single bacterium gives rise to more than a billion copies in 10 hours. A small vial of microorganisms can yield a huge number in less than a week. For some diseases, such as anthrax, inhaling a few thousand bacteria--which would cover an area smaller than the period at the end of this sentence--can be fatal. There are a number of candidate organisms terrorists could weaponize, but the Working Group identifies only a few that are widely known and feared and that would cause disease and deaths in sufficient numbers to cripple a city.

Preparing Your Family

There are a number of considerations that should be taken into account when undergoing therapy for Biological Contamination or immunizing your body against the possibility of infection. A few are mentioned below, and some product examples are given. The Institute for Molecular Medicine is a nonprofit institution and does not endorse commercial products. The products mentioned below are only examples of the types of substances that could be beneficial to patients.

 Increase your intake of fresh vegetables, fruits and grains. This will give you an increase in the level of Sterols and Sterolins, which in turn will act as a  regulator for your immune system. One such product is Vidatol.  Dosage: 3 tablets daily between meals. This tablet is not meant to treat the disease associated with Biological Infection, it is meant as a preparatory treatment. Infections such as Anthrax are fatal if your immune system is depleted.  This is child safe.  Decrease your intake of fats and eliminate simple or refined sugars that can suppress your immune system.

Potential Biological Agents, Associated Infections, and Treatment

Bacillus anthracis. Causes anthrax. If bacteria are inhaled, symptoms may develop in two to three days. High fever, vomiting, joint ache and labored breathing, and internal and external bleeding lesions follow initial symptoms resembling common respiratory infection. Exposure may be fatal. Vaccine and antibiotics provide protection unless exposure is very high. 
Precautionary: Begin with threat notification:

Adult: Vidatol: 900 mg daily

Child: Vidatol: 600 mg daily
Infected:
Adult Dosage: Doxycycline: 200-300 mg orally or intravenously as a loading dose, then 100 mg every 12 hr.
Child Dosage: Penicillin V: 25-50 mg/kg of body weight/day orally in divided doses 2 to 40 times/day

Botulinum toxin. Cause of botulism, produced by Clostridium botulinum bacteria. Symptoms appear 12 to 72 hours after ingestion or inhalation. Initial symptoms are nausea and diarrhea, followed by weakness, dizziness and respiratory paralysis, often leading to death. Antitoxin can sometimes arrest the process.

Yersinia pestis. Causes bubonic plague, the Black Death of the Middle Ages. If bacteria reach the lungs, symptoms--including fever and delirium--may appear in three or four days. Untreated cases are nearly always fatal. Vaccines can offer immunity, and antibiotics are usually effective if administered promptly.
Precautionary: Begin with threat notification:

Adult: Vidatol: 900 mg daily
Child: Vidatol: 600 mg daily
Infected:
Dosage: Streptomycin is the most effective, and should be used for the first five days of treatment with follow-on of Tetracycline, which should be continued for ten days. 
For new-born children kanamycin a safer drug

Ebola virus. Highly contagious and lethal. May not be desirable as a biological agent because of uncertain stability outside of animal host. Symptoms, appearing two or three days after exposure, include high fever, delirium, severe joint pain, bleeding from body orifices, and convulsions, followed by death. No known treatment.

CFIDS/GWI. Following military duties during the Gulf War, soldiers and DOD civilians based in Iraq experienced symptoms which started out as a relative benign series of flu-like illnesses becoming progressively worse with; intermittent fever, coughing, nausea, gastrointestinal problems, skin rashes, joint pain, memory loss, vision problems and severe headaches. This raises the possibility of these symptoms being the result of an unknown weapon designed by Iraq and made available to the terrorist networks. Treatment: Heavy regiment of antibiotics.
Adult Dosage: Doxycycline (200-300 milligrams/day)
Child Dosage: Doxycycline (50 milligrams/day)

More on Biological Agents


PLI Pharmaceuticals, San Jose, Costa Rica, International  506-283-4216, Toll Free 888-247-1601, www.purelife.cc

Return to Main Page


4 posted on 10/08/2001 4:17:40 AM PDT by vannrox
[ Post Reply | Private Reply | To 3 | View Replies]

To: vannrox
Plague

Plague, the disease caused by the bacteria Yersinia pestis (Y pestis), has had a profound impact on human history. In AD 541, the first great plague pandemic began in Egypt and swept over the world in the next four years.

Population losses attributable to plague during those years were between 50 and 60 percent. In 1346, the second plague pandemic, also known as the Black Death or the Great Pestilence, erupted and within 5 years had ravaged the Middle East and killed more than 13 million in China and 20-30 million in Europe, one third of the European population.

Advances in living conditions, public health and antibiotic therapy make such natural pandemics improbable, but plague outbreaks following an attack with a biological weapon do pose a serious threat.

Plague is one of very few diseases that can create widespread panic following the discovery of even a small number of cases. This was apparent in Surat, India, in 1994, when an estimated 500,000 persons fled the city in fear of a plague epidemic.

In the 1950s and 1960s, the U.S. and Soviet biological weapons programs developed techniques to directly aerosolize plague particles, a technique that leads to pneumonic plague, an otherwise uncommon, highly lethal and potentially contagious form of plague. A modern attack would most probably occur via aerosol dissemination of Y pestis, and the ensuing outbreak would be almost entirely pneumonic plague.

More than 10 institutes and thousands of scientists were reported to have worked with plague in the former Soviet Union.

Given the availability of Y pestis in microbe banks around the world, reports that techniques for mass production and aerosol dissemination of plague have been developed, the high fatality rate in untreated cases and the potential for secondary spread, a biological attack with plague is a serious concern.

An understanding of the epidemiology, clinical presentation and the recommended medical and public health response following a biological attack with plague could substantially decrease the morbidity and mortality of such an event.

A plague outbreak developing after the use of a biological weapon would follow a very different epidemiologic pattern than a naturally occurring plague epidemic.

The size of a pneumonic plague epidemic following an aerosol attack would depend on a number of factors, including the amount of agent used, the meteorological conditions and methods of aerosolization and dissemination.

A group of initial pneumonic cases would appear in about 1-2 days following the aerosol cloud exposure, with many people dying quickly after symptom onset. Human experience and animal studies suggest that the incubation period in this setting is 1 to 6 days.

A 1970 World Health Organization assessment asserted that, in a worst case scenario, a dissemination of 50 kg of Y pestis in an aerosol cloud over a city of 5 million might result in 150,000 cases of pneumonic plague, 80,000-100,000 of which would require hospitalization, and 36,000 of which would be expected to die.

There are no effective environmental warning systems to detect an aerosol cloud of plague bacilli, and there are no widely available rapid, diagnostic tests of utility. The first sign of a bioterrorist attack with plague would most likely be a sudden outbreak of patients presenting with severe symptoms.

A U.S. licensed vaccine exists and in a pre-exposure setting appears to have some efficacy in preventing or ameliorating bubonic disease. The mortality of untreated pneumonic plague approaches 100%.

Research and development efforts for a vaccine that protects against inhalationally acquired pneumonic plague are ongoing. A number of promising antibiotics and intervention strategies in the treatment and prevention of plague infection have yet to be fully explored experimentally.

Given that naturally occurring antibiotic resistance is rare and the lack of confirmation of engineered antibiotic resistance, the Working Group believes initial treatment recommendations should be based on known drug efficacy, drug availability and ease of administration.

People with household or face-to-face contacts with known pneumonic cases should immediately initiate antibiotic prophylaxis and, if exposure is ongoing, should continue it for 7 days following the last exposure.

In addition to antibiotic prophylaxis, people with established ongoing exposure to a patient with pneumonic plague should wear simple masks and should have patients do the same.

5 posted on 10/08/2001 4:20:20 AM PDT by vannrox
[ Post Reply | Private Reply | To 4 | View Replies]

To: vannrox
Anthrax

Bacillus anthracis, the organism that causes anthrax, derives its name from the Greek word for coal, anthracis, because of its ability to cause black, coal-like cutaneous eschars.

Anthrax infection is a disease acquired following contact with infected animals or contaminated animal products or following the intentional release of anthrax spores as a biological weapon.

In the second half of this century, anthrax was developed as part of a larger biological weapons program by several countries, including the Soviet Union and the U.S. The number of nations believed to have biological weapons programs has steadily risen from 10 in 1989 to 17 in 1995, but how many are working with anthrax is uncertain.

Perhaps more insidious is the specter of autonomous groups with ill intentions using anthrax in acts of terrorism. The Aum Shinrikyo religious sect, infamous for releasing sarin gas in a Tokyo subway station in 1995, developed a number of biological weapons, including anthrax.

Given appropriate weather and wind conditions, 50 kilograms of anthrax released from an aircraft along a 2 kilometer line could create a lethal cloud of anthrax spores that would extend beyond 20 kilometers downwind. The aerosol cloud would be colorless, odorless and invisible following its release. Given the small size of the spores, people indoors would receive the same amount of exposure as people on the street.

There are currently no atmospheric warning systems to detect an aerosol cloud of anthrax spores. The first sign of a bioterrorist attack would most likely be patients presenting with symptoms of inhalation anthrax.

A 1970 analysis by the World Health Organization concluded that the release of aerosolized anthrax upwind of a population of 5,000,000 could lead to an estimated 250,000 casualties, of whom as many as 100,000 could be expected to die.

A later analysis, by the Office of Technology Assessment of the U.S. Congress, estimated that 130,000 to 3 million deaths could occur following the release of 100 kilograms of aerosolized anthrax over Washington D.C., making such an attack as lethal as a hydrogen bomb. The Centers for Disease Control and Prevention estimates that such a bioterrorist attack would carry an economic burden of $26.2 billion per 100,000 people exposed to the spores.

The largest experience with inhalation anthrax occurred after the accidental release of aerosolized anthrax spores in 1979 at a military biology facility in Sverdlovsk, Russia. Some 79 cases of inhalation anthrax were reported, of which 68 were fatal.

One of the major problems with anthrax spores is the potentially long incubation period of subsequent infections. Exposure to an aerosol of anthrax spores could cause symptoms as soon as 2 days after exposure. However, illness could also develop as late as 6-8 weeks after exposure -- in Sverdlovsk, one case developed 46 days after exposure.

Further, the early presentation of anthrax disease would resemble a fever or cough and would therefore be exceedingly difficult to diagnose without a high degree of suspicion. Once symptoms begin, death follows 1-3 days later for most people. Precautionary medications include a regiment of Sterols and Sterolins (such as Vidatol) Which will boost human immune systems. If appropriate antibiotics are not started before development of symptoms, the mortality rate is estimated to be 90%. This Rate drops to 25% with precautioary care.

There are a number of rapid diagnostic tests for identifying anthrax at national reference laboratories, but none is widely available.

If anthrax is suspected on clinical, laboratory or pathology grounds, then the Working Group recommends that hospital epidemiologists contact local and state health officials immediately so that the proper reference tests can be performed.

The U.S. has a sterile protein-based human anthrax vaccine that was licensed in 1970 and has been mandated for use in all U.S. military personnel. In studies with monkeys, inoculation with this vaccine at 0 and 2 weeks was completely protective against infection from an aerosol challenge at 8 and 38 weeks, and 88% effective at 100 weeks.

However, U.S. vaccine supplies are limited and U.S. production capacity is modest. There is no vaccine available for civilian use.

Anthrax Treatment

6 posted on 10/08/2001 4:21:54 AM PDT by vannrox
[ Post Reply | Private Reply | To 5 | View Replies]

To: crystal55t
Smallpox

Smallpox, because of its high case-fatality rates and transmissibility, now represents one of the most serious bioterrorist threats to the civilian population. Over the centuries, naturally occurring smallpox, with its case-fatality rate of 30 percent or more and its ability to spread in any climate and season, has been universally feared as the most devastating of all the infectious diseases.

Smallpox was once worldwide in scope; before vaccination was practiced almost everyone eventually contracted the disease. In 1980, the World Health Assembly announced that smallpox had been eradicated and recommended that all countries cease vaccination. That same year, the Soviet government embarked on an ambitious program to grow smallpox in large quantities and adapt it for use in bombs and intercontinental ballistic missiles. That initiative succeeded.

Russia still possesses an industrial facility that is capable of producing tons of smallpox virus annually and also maintains a research program that is thought to be seeking to produce more virulent and contagious strains.

An aerosol release of smallpox virus would disseminate readily given its considerable stability in aerosol form and epidemiological evidence suggesting the infectious dose is very small. Even as few as 50-100 cases would likely generate widespread concern or panic and a need to invoke large-scale, perhaps national emergency control measures.

Several factors fuel the concern: the disease has historically been feared as one of the most serious of all pestilential diseases; it is physically disfiguring; it bears a 30 percent case-fatality rate; there is no treatment; it is communicable from person to person; and no one in the U.S. has been vaccinated during the past 25 years. Vaccination ceased in this country in 1972, and vaccination immunity acquired before that time has undoubtedly waned.

Smallpox spreads directly from person to person, primarily by droplet nuclei expelled from the oropharynx of the infected person or by aerosol. Natural infection occurs following implantation of the virus on the oropharyngeal or respiratory mucosa.

Contaminated clothing or bed linen could also spread the virus. Special precautions need to be taken to insure that all bedding and clothing of patients are autoclaved. Disinfectants such as hypochlorite and quaternary ammonia should be used for washing contaminated surfaces.

A smallpox outbreak poses difficult problems because of the ability of the virus to continue to spread throughout the population unless checked by vaccination and/or isolation of patients and their close contacts.

Between the time of an aerosol release of smallpox and diagnosis of the first cases, an interval of as much as two weeks is apt to occur. This is because there is an average incubation period of 12 to 14 days.

After the incubation period, the patient experiences high fever, malaise, and prostration with headache and backache. Severe abdominal pain and delirium are sometimes present. A mascopapular rash then appears, first on the mucosa of the mouth and pharynx, face and forearms, spreading to the trunk and legs. Within one or two days, the rash becomes vesicular and later pustular. The pustules are characteristically round, tense and deeply embedded in the dermis; crusts begin to form about the eighth or ninth day. When the scabs separate, pigment-free skin remains, and eventually pitted scars form.

Approximately 140,000 vials of vaccine are in storage at the Centers for Disease Control and Prevention, each with doses for 50-60 people, and an additional 50-100 million doses are estimated to exist worldwide. This stock cannot be immediately replenished, since all vaccine production facilities were dismantled after 1980, and renewed vaccine production is estimated to require at least 24-36 months.

In 2000, CDC awarded a contract to Oravax of Cambridge, Massachusetts to produce smallpox vaccine. Initially producing 40 million doses, Oravax anticipates delivery of the first full scale production lots in 2004.

Treatment of smallpox is limited to supportive therapy and antibiotics as required for treating secondary bacterial infections. There are no proven antiviral agents effective in treating smallpox.

Recommendations of the Working Group include testing and ultimate consideration for FDA approval of a vaccinia strain grown in tissue culture rather than on calves, finding a rapid diagnostic test for smallpox virus in the asymptomatic early stages, and developing a more attenuated strain of vaccine.


Tularemia

Francisella tularensis, the organism that causes tularemia, is one of the most infectious pathogenic bacteria known, requiring inoculation or inhalation of as few as 10 organisms to cause disease. It is considered to be a dangerous potential biological weapon because of its extreme infectivity, ease of dissemination, and substantial capacity to cause illness and death.

During World War II, the potential of F. tularensis as a biological weapon, was studied by the Japanese as well as by the US and its allies.

Tularemia was one of several biological weapons that were stockpiled by the US military in the late 1960's, all of which were destroyed by 1973. The Soviet Union continued weapons production of antibiotic and vaccine resistant strains into the early 1990s.

Francisella tularensis is a hardy non-spore forming organism that is capable of surviving for weeks at low temperatures in water, moist soil, hay, straw or decaying animal carcasses.

F. tularensis has been divided into two subspecies: F. tularensis biovar tularensis (type A), which is the most common biovar isolated in North America and may be highly virulent in humans and animals; F. tularensis biovar palaearctica (type B) which is relatively avirulent and thought to the cause of all human tularemia in Europe and Asia.

Tularemia is a zoonosis. Natural reservoirs include small mammals such as voles, mice, water rats, squirrels, rabbits and hares. Naturally acquired human infection occurs through a variety of mechanisms such as: bites of infected arthropods; handling infectious animal tissues or fluids; direct contact or ingestion of contaminated water, food, or soil; and inhalation of infective aerosols. F. tularensis is so infective that examining an open culture plate can cause infection.

Human to human transmission has not been documented.

In the natural setting, tularemia is noted to be a predominately rural disease with clinical presentations including ulceroglandular, glandular, oculoglandular, oropharyngeal, pneumonic, typhoidal and septic forms.

The Working Group on Civilian Biodefense believes that of the various possible ways that F. tularensis could be used as a weapon, an aerosol release would cause the greatest adverse medical and public health consequences.

A World Health Organization (WHO) expert committee reported in 1970 that if 50 kg of virulent F. tularensis was dispersed as an aerosol over a metropolitan area with a population of 5 million there would an estimated 250,000 incapacitating casualties, including 19,000 deaths.

Aerosol dissemination of F. tularensis in a populated area would be expected to result in the abrupt onset of large numbers of cases of acute, non-specific febrile illness beginning 3 to 5 days later (incubation range, 1-14 days), with pleuropneumonitis developing in a significant proportion of cases over the ensuing days and weeks. Without antibiotic treatment, the clinical course could progress to respiratory failure, shock and death.

The overall mortality rate for severe Type A strains has been 5-15%, but in pulmonic or septicemic cases of tularemia without antibiotics treatment the mortality rate has been as high as 30-60%. With treatment, the most recent mortality rates in the US have been 2%. Aminoglycosides, macrolides, chloramphenicol and fluoroquinolones have each been with used with success in the treatment of tularemia.

In the United States, a live-attenuated vaccine derived from the avirulent Live Vaccine Strain (LVS) has been used to protect laboratory personnel routinely working with F. tularensis. Given the short incubation period of tularemia and incomplete protection of current vaccines against inhalational tularemia, vaccination is not recommended for post-exposure prophylaxis.

Given the lack of human-to-human transmission, isolation is not recommended for tularemia patients.

The Working Group lacks information on survival of intentionally-dispersed particles, but would expect a short half-life due to dessication, solar radiation, oxidation and other environmental factors, and a very limited risk from secondary dispersal.

Simple, rapid and reliable diagnostic tests that could be used to identify persons infected with F. tularensis in the mass exposure setting need to be developed. Research is also needed to develop accurate and reliable proced-ures to rapidly detect F. tularensis in environmental samples.


Botulinum Toxin

Botulinum toxin poses a major bioweapons threat because of its extreme potency and lethality; its ease of production, transport and misuse; and the potential need for prolonged intensive care in affected persons. Botulinum toxin is the single most poisonous substance known.

A number of states named by the U.S. State Department as "state sponsors of terrorism" have developed or are developing botulinum toxin as a biological weapon. Aum Shinrikyo tried but failed to use botulinum toxin as a biological weapon.

Botulinum toxin is derived from the genus of anaerobic bacteria named Clostridia. Seven antigenic types of botulinum toxin exist, designated from A through G. They can be identified based on antibody cross reactivity studies - i.e., anti-A toxin antibodies do not neutralize the B through G toxins.

Naturally occurring botulism is the disease that results from the absorption of botulinum toxin into the circulation from a mucosal surface (gut, lung) or a wound. It does not penetrate intact skin. The toxin irreversibly binds to peripheral cholinergic synapses, preventing the release of the neurotransmitter acetylcholine from the terminal end of motor neurons. This leads to muscle paralysis, and in severe cases, can lead to a need for mechanical respiration.

The incubation period for food-borne botulism can be from 2 hours to 8 days after ingestion, depending on the dose of the bacteria or the toxin. The average incubation period is 12-72 hours after ingestion. Patients with botulism typically present with difficulty speaking, seeing and/or swallowing. Prominent neurologic findings in all forms of botulism include ptsosis, diplopia, blurred vision, dysarthria and dysphagia. Patients typically are afebrile and do not have an altered level of consciousness. Patients may initially present with gastrointestinal distress, nausea, and vomiting preceding neurological symptoms. Symptoms are similar for all toxin types, but the severity of illness can vary widely, in part depending on the amount of toxin absorbed. Recovery from paralysis can take from weeks to months and requires the growth of new motor nerve endings. In the event botulism is suspected, the hospital epidemiologist and local and state health departments should be contacted immediately.

Natural cases of botulism are rare and typically result from food contamination. Many types of food have been associated in outbreaks in the past, with the common factor being that implicated food items were not heated or were incompletely heated. Heat > 85oC inactivates the toxin. The largest botulism outbreak in the U.S. in the past century occurred in 1977, when 59 people became ill from poorly preserved jalapeño peppers.

No cases of waterborne botulism have ever been reported. This is likely due to the large amount of toxin needed, and the fact that the toxin is easily neutralized by common water treatment techniques.

A deliberate aerosol or food-borne release of botulinum toxin could be detected by several features including: a large number of acute cases presenting all at once; cases involving an uncommon toxin type (C, D, F, G, or non-aquatic food associated E); patients with a common geographic factor but without a common dietary exposure; and, multiple simultaneous outbreaks without a common source.

Diagnosis and testing are available at the CDC and some local and state laboratories. The standard test for the toxin is the mouse bioassay. Unfortunately, this assay is time consuming. Future development is focused on rapid diagnosis/detection. Polymerase Chain Reaction (PCR) assays that can detect the Clostridia spp. bacterial DNA toxin sequences are currently under development. Enzyme Linked ImmunoSorbent Assays (ELISAs) are being developed to detect functionally active toxins.

In the event that there is a clinical suspicion of botulinum toxin, treatment with antitoxin should not be delayed for microbiological testing. In the U.S., licensed botulinum antitoxin is available from the CDC via state and local health departments. An investigational heptavalent antitoxin is held by the U.S. Army. Optimal therapy for botulism requires early suspicion of the disease and prompt administration of antitoxin in conjunction with supportive care. Supportive care for patients with botulism may include mechanical ventilators in the intensive care unit, parenteral nutrition, and treatment of secondary infections.

An investigational botulinum toxoid is used to provide immunity for laboratory workers. It has been used to provide immunity against botulinum toxin over the past 30 years. However, supply of the toxoid is limited, and use of it would eliminate possible beneficial uses of toxoid for medical purposes. The toxoid induces immunity over several months and so would not be effective for rapid, post-exposure prophylaxis.

Existing technologies could produce large reserves of human antibody against the botulinum toxin. Administration of such a therapy could provide immunity of up to a month or greater and obviate the need for rationing the equine antitoxin. The development of such a human antibody reserve would require sufficient resources be dedicated to this problem.



7 posted on 10/08/2001 4:27:54 AM PDT by vannrox
[ Post Reply | Private Reply | To 2 | View Replies]

To: dbbeebs
CFIDS Chronicle 9(3): 66-69 (1996)

MYCOPLASMAL INFECTIONS--

Diagnosis and Treatment of Gulf War Illness/CFIDS Patients

Garth L. Nicolson, Ph.D.

Nancy L. Nicolson, Ph.D.

Introduction--The problem

Returning U.S. and coalition military forces from Operation Desert Storm/Desert Shield have reported a variety of health problems, including a collection of signs and symptoms characterized by disabling fatigue, intermittent fever, muscle and joint pain, impairments in short-term memory, headaches, skin rashes, gastrointestinal and respiratory problems and a collection of additional symptoms that has defied a careful case definition.1 This disorder has been called Persian Gulf War Syndrome, Gulf War Illness (GWI) or Desert Storm Illness, and it has afflicted approximately 100,000 Desert Storm veterans and their immediate family members. Since the signs and symptoms of GWI were not well established as criteria for particular illnesses and they did not readily fit into common military or Veterans Administration diagnosis categories, this has resulted in unknown diagnoses, or they have been diagnosed with psychological problems, such as Post Traumatic Stress Disorder.1 Military personnel that we interviewed were particularly disdainful of this explanation for GWI. Although the concept of a distinct syndrome peculiar to the Persian Gulf Theater of Operations has been advanced, it has not been proven,2 and we prefer to use the term Gulf War Illness instead of Gulf War Syndrome.

Recently Major General Ronald Blanck, commanding officer of Walter Reed Army Medical Center in Washington DC, stated at a recent CFIDS Association meeting that the symptomology of GWI is analogous to Chronic Fatigue-Immune Dysfunction Syndrome (CFIDS).3 At about this time we were in the process of analyzing our data on approximately 650 GWI patients,4,5 and we found that the signs and symptoms of these GWI patients fit quite closely with the literature signs and symptoms of CFIDS6,7 (Figure 1).5 The classical working case definition of CFIDS is that of Holmes et al.8, who proposed that CFIDS is primarily characterized by persistent or relapsing, debilitating fatigue or easy fatigability in a person who has no previous history of similar symptoms, that does not resolve with rest and is severe enough to reduce or impair average daily activity below 50% of the patient's premorbid activity level. In addition to the absence of clinical conditions that could easily explain the symptoms, such as malignancies or autoimmune diseases, patients present with mild fever, sore throat, joint and muscle pain, generalized muscle weakness, headaches, painful lymph nodes, sleep difficulties, and neuropsychologic complaints, such as memory loss, light sensitivity, confusion, transient visual problems, irritability and depression.8 These CFIDS signs and symptoms closely parallel those found in GWI (Figure 1).4,5

A few of the patients that have some of the multiple chronic symptoms shown in Figure 1 may eventually have their diagnoses linked to chemical exposures in the Persian Gulf, such as oil spills and fires, smoke from military operations, chemicals on clothing, pesticides, chemoprophylactic agents, chemical weapons and others. In some cases, such exposure may have resulted in Multiple Chemical Sensitivities (MCS). MCS shares some but certainly not all of the symptoms in Figure 1. In many of the soldiers with GWI the spread of the illness to immediate family members was not consistent with a diagnosis of MCS. The U.S. Senate Committee on Banking, Housing and Urban Affairs found that 77% of the spouses and 65% of the children of GWI patients have developed the same or similar signs and symptoms. Thus we feel that the most appropriate syndrome that characterizes GWI is CFIDS, and this type of GWI/CFIDS is apparently being transmitted to close family members.5

Hypotheses on the origin of Gulf War Illness

Several hypotheses have been advanced to explain the pathophysiologic origins of CFIDS, but the model of Cheney10 is particularly useful in discussing the complex, multiorgan signs and symptoms of CFIDS. In this model, the syndrome is initiated by immune activation and the stimulated release of interferons and cytokines that cause neurotoxicity and other systemic effects. We have proposed that GWI/CFIDS may be initiated by host responses to chronic infectious agents resulting in interferon and cytokine production.5 There were a number of potential sources of chronic infectious agents in the Persian Gulf Theater of Operations, including the vaccines that were used to immunize soldiers, endogenous infectious agents in the Persian Gulf, blow-back of Chemical/Biological Warfare (CBW) agents from bombing of CBW factories and supply depots and offensive Iraqi CBW weapons mounted on SCUD missiles and other projectiles. Of these, we consider the most likely sources of potential chronic infectious agents to be the vaccines and Iraqi offensive CBW weapons, some of which were originally purchased from U.S. companies and transferred to Iraq.9 We have concluded that in addition to the signs and symptoms, the causes of GWI/CFIDS are also complex, and as a disease it is probably caused by several types of agents or conditions.4

We have concentrated on chronic infectious agents as one possible cause for GWI/CFIDS. Irrespective of the source(s) of possible chronic infectious agents, it was necessary to develop diagnostic procedures to identify if these agents might be present in at least some of the Operation Desert Storm veterans and their family members who have GWI/CFIDS. From our own experience with GWI/CFIDS (our step-daughter returned with GWI/CFIDS 6 months after service in Operation Desert Storm in a U.S. Army Airborne Division; eventually our entire family presented with CFIDS signs and symptoms), we suggested that most of the GWI/CFIDS signs and symptoms could be explained by chronic pathogenic mycoplasma infections.11 Mycoplasma infections usually produce relatively benign diseases limited to particular tissue sites or organs, such as urinary tract or respiratory infections; however, the types of mycoplasmas that we have detected in Desert Storm veterans and their family members that may be causing the CFIDS and other symptoms are very pathogenic, colonize a variety of organs and tissues, and are difficult to treat.12

Mycoplasmal infections in GWI/CFIDS patients

Our personal experience with GWI/CFIDS suggested that this disorder might respond to particular antibiotics that are known to be effective against mycoplasmal infections, such as doxycycline.11 In fact, we found that of the 73 Desert Storm veterans who had most of the GWI/CFIDS symptoms listed in Figure 1, 55 had good responses with doxycycline, and after multiple 6-week courses (up to 6) of antibiotics eventually recovered.11 We then set out to test the hypothesis that chronic mycoplasmal infections were the underlying event that may have triggered the GWI/CFIDS syndrome by first analyzing for mycoplasmal infections. The types of mycoplasmas that we eventually found are not easily detected but can be identified in blood leukocytes (white blood cells) by a technique that we developed called Gene Tracking.13 This technique uses a very sensitive and specific DNA hybridization procedure to positively identify unique DNA sequences that are indicative of specific species of mycoplasmas and other organisms. In our preliminary study on 30 veterans with GWI/CFIDS and their families, we have found evidence of mycoplasmal infections in about one-half (14/30) of the GWI/CFIDS patients' blood leukocytes.12 Not every Desert Storm veteran had the same type of mycoplasma DNA sequences in their blood, but we found that the majority of the mycoplasmas identified in the nuclear fractions prepared from blood leukocytes were identified as Mycoplasma fermentans (incognitus strain).12 Even pathogenic mycoplasmas, such as M. fermentans (incognitus strain) or M. penetrans, should be treatable with multiple courses of antibiotics,11 such as doxycycline (200-300 milligrams/day).14 We found four antibiotics that were useful for treatment of GWI/CFIDS patients and have suggested that these be used in multiple 6 week courses: doxycycline (200-300 milligrams/day), azithromycin (Zithromax, 500 milligrams/day), minocycline (200-300 milligrams/day) and ciprofloxacin (Cipro, 1,000-1,500 milligrams/day). One cycle of antibiotic therapy was not sufficient to completely suppress the mycoplasmal infections. Most GWI/CFIDS patients required several (2-6) cycles of antibiotic therapy to completely recover, and even then some of these patients continued to relapse occasionally when they were physically stressed, although their symptoms were almost always less severe than their initial relapses after their first few cycles of antibiotic therapy.

Some GWI/CFIDS case reports

We consider it quite likely that a large fraction of the Desert Storm veterans suffering from GWI/CFIDS may have been infected with pathogenic mycoplasmas and other possible pathogens (invasive bacteria), and such infections can produce the signs and symptoms in Figure 1, sometimes long after exposure. This would also explain the apparent contagious nature of GWI/CFIDS seen in many veterans, and the appearance of similar GWI/CFIDS symptoms in their immediate family members.

One of our fist patients was a Special Forces officer (U.S. Navy SEAL) now in the Delta Force at Fort Bragg, NC. He was in charge of Special Forces units that were involved in sensitive covert missions during Operation Desert Storm. He presented several months after the Gulf War with a flu-like illness that progressed to chronic fatigue, fever, stomach cramps, joint pain, skin rashes, memory loss, dehydration, headaches, heart pain and other symptoms. His vision became so diminished that physicians at Womack Army Hospital at Fort Bragg were considering surgery. After several courses of doxycycline, he completely recovered and has recently been promoted to the Executive Officer of our DELTA FORCE.

Another subject was an U.S. Army officer who served in the 101st Airborne Division (Air Assault). He was deployed on the deep insertions into Iraq. His unit did not come under enemy fire, and he considered his service relatively uneventful, until months after he returned to the U.S. What started out as a relative benign series of flu-like illnesses became progressively worse with intermittent fever, coughing, nausea, gastrointestinal problems, skin rashes, joint pain, memory loss, vision problems and severe headaches. Then his wife began to have chronic fatigue and gynecological problems, aching joints, headaches, and her stomach began to swell, causing severe pain. His 7 year-old daughter also became ill with similar flu-like symptoms that did not go away and progressively became worse, resulting in chronic fatigue, skin lesions, vomiting, headaches, aching joints, and inability to gain weight. Several other families of Gulf War veterans at his base had similar health problems. These families were being told that their symptoms were the result of psychological problems, but their signs and symptoms were more consistent with GWI/CFIDS. This officer and his family tested positive for M. fermentans (incognitus strain) and were placed on several 6 week cycles of doxycycline (their child was placed on 50 milligrams/day doxycycline). They and others on their base have recovered and for the most part no longer have GWI/CFIDS, although some of their symptoms reappear occasionally.

Another patient was a U.S. Air Force intelligence officer attached to the 5th Special Forces Group based at King Fahd Airport west of Dhahran and at King Khalid Military City in Saudi Arabia. He was involved in the Special Forces operations in Iraq, but was not involved in combat. He was exposed repeatedly to attacks by SCUD B missiles. After his return to the U.S., he noticed that he had a constant sore throat, night sweats, and intermittent fevers that progressed to include shortness of breath, dizziness, joint pain, short term memory loss, vision problems, diarrhea and other bowel problems, skin rashes and severe to moderate fatigue. He eventually left the military and could not obtain treatment from VA hospitals for his GWI/CFIDS. He tested positive for M. fermentans (incognitus strain), received several courses of doxycycline, and he has completely recovered. Upon retesting his blood for the presence of M. fermentans (incognitus strain) after he recovered, this infection was no longer present.

One patient was a 48 year-old U.S. Marine Corps officer was attached to the Central Command Staff in Saudi Arabia at Operation Desert Storm Command Headquarters. His only noteworthy experience was that he examined SCUD B (SS-1) missile impact sites. Within 10 months after his return to the U.S. he presented with chronic fatigue, skin rashes, diarrhea, headaches, aching joints, muscle pain, fevers, sleep problems, nausea, vision problems, memory loss and dental problems. His wife also became ill with GWI/CFIDS and had similar symptoms. Using Gene Tracking both tested Positive for M. fermentans (incognitus strain) and were placed on doxycycline. After 2-3 cycles of therapy, he completely recovered and his wife is recovering, but still relapses occasionally with GWI/CFIDS signs and symptoms.



Conclusions

We have found that a sizable fraction of Desert Storm veterans and their immediate family members who have GWI/CFIDS also have chronic mycoplasmal infections.12 Almost all of these patients responded to antibiotics: doxycycline, azithromycin, minocycline or ciprofloxacin. Eventually these GWI/CFIDS patients recovered, but they still relapse occasionally with some of the GWI/CFIDS signs and symptoms.12 Not all Desert Storm veterans with GWI/CFIDS responded to antibiotic therapy, suggesting that some patients probably display GWI/CFIDS signs and symptoms because of some other type of chronic infection or other cause, such as chemical insults.15 In addition, some veterans have MCS and cannot take certain antibiotics, such as doxycycline; however, they seem to tolerate other antibiotics (azithromycin, ciprofloxacin), especially if given intravenously. Our results and those of others who are examining other possible causes for GWI/CFIDS indicate that there are multiple causes for these CFIDS illnesses, but a sizable fraction of veterans with GWI/CFIDS have identifiable chronic mycoplasmal infections that can be successfully treated with the appropriate antibiotics. We have begun to examine some civilians with CFIDS, and we have some preliminary evidence indicating that a subset of CFIDS patients may have chronic infections, such as caused by mycoplasmas, and these cases can be successfully treated with antibiotics similar to the GWI/CFIDS patients.

References

1. NIH Technology Assessment Workshop Panel: The Persian Gulf Experience and Health. JAMA. 1994;272:391-396.

2. Boaz Milner I, Axelrod BN, Pasquantonia J, Silanpaa M: Is there a Gulf War Syndrome? JAMA 1994; 271:661.

3. Schmidt P, Blanck RM: Gulf War Syndrome and CFS. CFIDS Chronicle 1995;8:25-27.

4. Nicolson GL, Hyman E, Korényi-Both A, Lopez DA, Nicolson NL, Rea W, Urnovitz H: Progress on Persian Gulf War Illnesses--reality and hypotheses. Int J Occup Med Tox 1995;4:365-370.

5. Nicolson GL, Nicolson NL: Chronic fatigue illnesses and Operation Desert Storm. J Occup Environ Med 1996;38:14-17.

6. Shafran S: The chronic fatigue syndrome. Amer J Med 1991;90:730-739.

7. Bell DS: Chronic fatigue syndrome update. Postgrad Med 1994;96:73-81.

8. Holmes GP, Kaplan JE, Gantz NM, Komaroff AL, et al.: Chronic Fatigue Syndrome: A working case definition. Ann Int Med 1988;108:387-389.

9. U.S. Senate Committee on Banking, Housing and Urban Affairs: U.S. chemical and biological warefare-related dual use exports to Iraq and their possible impact on the health consequences of the Persian Gulf War. U.S. Senate Report to the 103rd Congress, May 25, 1994.

10. Cheney PR: Proposed pathophysiologic model of CFIDS. CFIDS Chronicle 1994;7:1-3.

11. Nicolson GL, Nicolson NL: Doxycycline treatment and Desert Storm JAMA. 1995; 273:618-619.

12. Nicolson GL, Nicolson NL: Diagnosis and treatment of mycoplasmal infections in Persian Gulf War Illness-CFIDS patients. Int J Occup Med Immunol Tox 1996;5: 69-78.

13. Nicolson NL, Nicolson GL: The isolation, purification and analysis of specific gene-containing nucleoproteins and nucleoprotein complexes. Meth Mol Genet 1994;5:281-298.

14. Lo S-C, Buchholz CL, Wear DJ, Hohm RC, Marty AM: Histopathology and doxycycline treatment in a previously healthy non-AIDS patient systemically infected by Mycoplasma fermentans (incognitus strain). Mod Pathol 1991;6:750-754.

15. Vojdani A, Ghoneum M, Brautbar N: Immune alteration associated with exposure to toxic chemicals. Toxicol Ind Health 1992;8:239-254.

Figure Legend

Figure 1. Comparison of the most commonly found signs and symptoms in approximately 650 Desert Storm veterans suffering from GWI/CFIDS with CFIDS (from Nicolson and Nicolson12).


(Back)

8 posted on 10/08/2001 4:32:53 AM PDT by vannrox
[ Post Reply | Private Reply | To 3 | View Replies]

To: vannrox

Smallpox as a Biological Weapon

Medical and Public Health Management

Author Information  Donald A. Henderson, MD, MPH; Thomas V. Inglesby, MD; John G. Bartlett, MD; Michael S. Ascher, MD; Edward Eitzen, MD, MPH; Peter B. Jahrling, PhD; Jerome Hauer, MPH; Marcelle Layton, MD; Joseph McDade, PhD; Michael T. Osterholm, PhD, MPH; Tara O'Toole, MD, MPH; Gerald Parker, PhD, DVM; Trish Perl, MD, MSc; Philip K. Russell, MD; Kevin Tonat, PhD; for the Working Group on Civilian Biodefense

Objective  To develop consensus-based recommendations for measures to be taken by medical and public health professionals following the use of smallpox as a biological weapon against a civilian population.

Participants  The working group included 21 representatives from staff of major medical centers and research, government, military, public health, and emergency management institutions and agencies.

Evidence  The first author (D.A.H.) conducted a literature search in conjunction with the preparation of another publication on smallpox as well as this article. The literature identified was reviewed and opinions were sought from experts in the diagnosis and management of smallpox, including members of the working group.

Consensus Process  The first draft of the consensus statement was a synthesis of information obtained in the evidence-gathering process. Members of the working group provided formal written comments that were incorporated into the second draft of the statement. The working group reviewed the second draft on October 30, 1998. No significant disagreements existed and comments were incorporated into a third draft. The fourth and final statement incorporates all relevant evidence obtained by the literature search in conjunction with final consensus recommendations supported by all working group members.

Conclusions  Specific recommendations are made regarding smallpox vaccination, therapy, postexposure isolation and infection control, hospital epidemiology and infection control, home care, decontamination of the environment, and additional research needs. In the event of an actual release of smallpox and subsequent epidemic, early detection, isolation of infected individuals, surveillance of contacts, and a focused selective vaccination program will be the essential items of an effective control program.

JAMA. 1999;281:2127-2137JST90000

This is the second article in a series entitled Medical and Public Health Management Following the Use of a Biological Weapon: Consensus Statements of the Working Group on Civilian Biodefense.1 The working group has identified a limited number of widely known organisms that could cause disease and deaths in sufficient numbers to cripple a city or region. Smallpox is one of the most serious of these diseases.

If used as a biological weapon, smallpox represents a serious threat to civilian populations because of its case-fatality rate of 30% or more among unvaccinated persons and the absence of specific therapy. Although smallpox has long been feared as the most devastating of all infectious diseases,2 its potential for devastation today is far greater than at any previous time. Routine vaccination throughout the United States ceased more than 25 years ago. In a now highly susceptible, mobile population, smallpox would be able to spread widely and rapidly throughout this country and the world.

CONSENSUS METHODS


Members of the working group were selected by the chairman in consultation with principal agency heads in the Department of Health and Human Services (DHHS) and the US Army Medical Research Institute of Infectious Diseases (USAMRIID).

The first author (D.A.H.) conducted a literature search in conjunction with the preparation of another publication on smallpox2 as well as this article. The literature was reviewed and opinions were sought from experts in the diagnosis and management of smallpox, including members of the working group.

The first draft of the working group's consensus statement was the result of synthesis of information obtained in the evidence-gathering process. Members of the working group were asked to make written comments on the first draft of the document in September 1998. Suggested revisions were incorporated into the second draft of the statement. The working group was convened to review the second draft of the statement on October 30, 1998. Consensus recommendations were made and no significant disagreements existed at the conclusion of this meeting. The third draft incorporated changes suggested at the conference and working group members had an additional opportunity to suggest final revisions. The final statement incorporates all relevant evidence obtained by the literature search in conjunction with final consensus recommendations supported by all working group members.

This article is intended to provide the scientific foundation and initial framework for the detailed planning that would follow a bioterrorist attack with smallpox. This planning must encompass coordinated systems approaches to bioterrorism, including public policies and consequence management by local and regional public and private institutions. The assessment and recommendations provided herein represent the best professional judgment of the working group at this time based on data and expertise currently available. The conclusions and recommendations need to be regularly reassessed as new information becomes available.

HISTORY AND POTENTIAL AS A BIOWEAPON


Smallpox probably was first used as a biological weapon during the French and Indian Wars (1754-1767) by British forces in North America.3 Soldiers distributed blankets that had been used by smallpox patients with the intent of initiating outbreaks among American Indians. Epidemics occurred, killing more than 50% of many affected tribes. With Edward Jenner's demonstration in 1796 that an infection caused by cowpox protected against smallpox and the rapid diffusion worldwide of the practice of cowpox inoculation (ie, vaccination),4 the potential threat of smallpox as a bioweapon was greatly diminished.

A global campaign, begun in 1967 under the aegis of the World Health Organization (WHO), succeeded in eradicating smallpox in 1977.1 In 1980, the World Health Assembly recommended that all countries cease vaccination.5 A WHO expert committee recommended that all laboratories destroy their stocks of variola virus or transfer them to 1 of 2 WHO reference laboratoriesthe Institute of Virus Preparations in Moscow, Russia, or the Centers for Disease Control and Prevention (CDC) in Atlanta, Ga. All countries reported compliance. The WHO committee later recommended that all virus stocks be destroyed in June 1999, and the 1996 World Health Assembly concurred.6 In 1998, possible research uses for variola virus were reviewed by a committee of the Institute of Medicine (IOM).7 The IOM committee concluded, as did the preceding WHO committee, that there were research questions that might be addressed if the virus were to be retained. However, the IOM committee did not explore the costs or relative priority to be assigned to such an effort, and that committee was not asked to weigh the possible benefits resulting from such research activities contrasted with the possible benefits resulting from an international decision to destroy all virus stocks. These considerations will be weighed and decided by the 1999 World Health Assembly.

Recent allegations from Ken Alibek, a former deputy director of the Soviet Union's civilian bioweapons program, have heightened concern that smallpox might be used as a bioweapon. Alibek8 reported that beginning in 1980, the Soviet government embarked on a successful program to produce the smallpox virus in large quantities and adapt it for use in bombs and intercontinental ballistic missiles; the program had an industrial capacity capable of producing many tons of smallpox virus annually. Furthermore, Alibek reports that Russia even now has a research program that seeks to produce more virulent and contagious recombinant strains. Because financial support for laboratories in Russia has sharply declined in recent years, there are increasing concerns that existing expertise and equipment might fall into non-Russian hands.

The deliberate reintroduction of smallpox as an epidemic disease would be an international crime of unprecedented proportions, but it is now regarded as a possibility. An aerosol release of variola virus would disseminate widely, given the considerable stability of the orthopoxviruses in aerosol form9 and the likelihood that the infectious dose is very small.10 Moreover, during the 1960s and 1970s in Europe, when smallpox was imported during the December to April period of high transmission, as many as 10 to 20 second-generation cases were often infected from a single case. Widespread concern and, sometimes, panic occurred, even with outbreaks of fewer than 100 cases, resulting in extensive emergency control measures.2

EPIDEMIOLOGY


Smallpox was once worldwide in scope, and before vaccination was practiced, almost everyone eventually contracted the disease. There were 2 principal forms of the disease, variola major and a much milder form, variola minor (or alastrim). Before eradication took place, these forms could be differentiated clinically only when occurring in outbreaks; virological differentiation is now possible.11, 12 Through the end of the 19th century, variola major predominated throughout the world. However, at the turn of the century, variola minor was first detected in South Africa and later in Florida, from whence it spread across the United States and into Latin America and Europe.13 Typical variola major epidemics such as those that occurred in Asia resulted in case-fatality rates of 30% or higher among the unvaccinated, whereas variola minor case-fatality rates were customarily 1% or less.2

Smallpox spreads from person to person,10, 14 primarily by droplet nuclei or aerosols expelled from the oropharynx of infected persons and by direct contact. Contaminated clothing or bed linens can also spread the virus.15 There are no known animal or insect reservoirs or vectors.

Historically, the rapidity of smallpox transmission throughout the population was generally slower than for such diseases as measles or chickenpox. Patients spread smallpox primarily to household members and friends; large outbreaks in schools, for example, were uncommon. This finding was accounted for in part by the fact that transmission of smallpox virus did not occur until onset of rash. By then, many patients had been confined to bed because of the high fever and malaise of the prodromal illness. Secondary cases were thus usually restricted to those who came into contact with patients, usually in the household or hospital.

The seasonal occurrence of smallpox was similar to that of chickenpox and measlesits incidence was highest during winter and early spring.16 This pattern was consonant with the observation that the duration of survival of orthopoxviruses in the aerosolized form was inversely proportional to both temperature and humidity.9 Likewise, when imported cases occurred in Europe, large outbreaks sometimes developed during the winter months, rarely during the summer.17

The patient was most infectious from onset of rash through the first 7 to 10 days of rash (Figure 1).17, 18 As scabs formed, infectivity waned rapidly. Although the scabs contained large amounts of viable virus, epidemiological and laboratory studies indicate that they were not especially infectious, presumably because the virions were bound tightly in the fibrin matrix.19

The age distribution of cases depended primarily on the degree of smallpox susceptibility in the population. In most areas, cases predominated among children because adults were protected by immunity induced by vaccination or previous smallpox infection. In rural areas that had seen little vaccination or smallpox, the age distribution of cases was similar to the age distribution of the population. The age distribution pattern of cases in the United States presumably would be such if smallpox were to occur now because vaccination immunity in the population has waned so substantially.

MICROBIOLOGY


Smallpox, a DNA virus, is a member of the genus orthopoxvirus.20 The orthopoxviruses are among the largest and most complex of all viruses. The virion is characteristically a brick-shaped structure with a diameter of about 200 nm. Three other members of this genus (monkeypox, vaccinia, and cowpox) can also infect humans, causing cutaneous lesions, but only smallpox is readily transmitted from person to person.2 Monkeypox, a zoonotic disease, presently is found only in tropical rain forest areas of central and western Africa and is not readily transmitted among humans.21 Vaccinia and cowpox seldom spread from person to person.

PATHOGENESIS AND CLINICAL PRESENTATION


Natural infection occurs following implantation of the virus on the oropharyngeal or respiratory mucosa.2 The infectious dose is unknown but is believed to be only a few virions.10 After the migration of virus to and multiplication in regional lymph nodes, an asymptomatic viremia develops on about the third or fourth day, followed by multiplication of virus in the spleen, bone marrow, and lymph nodes. A secondary viremia begins on about the eighth day and is followed by fever and toxemia. The virus, contained in leukocytes, then localizes in small blood vessels of the dermis and beneath the oral and pharyngeal mucosa and subsequently infects adjacent cells.

At the end of the 12- to 14-day incubation period (range, 7-17 days), the patient typically experiences high fever, malaise, and prostration with headache and backache.2 Severe abdominal pain and delirium are sometimes present. A maculopapular rash then appears on the mucosa of the mouth and pharynx, face, and forearms, and spreads to the trunk and legs (Figure 2).2 Within 1 to 2 days, the rash becomes vesicular and, later, pustular. The pustules are characteristically round, tense, and deeply embedded in the dermis; crusts begin to form on about the eighth or ninth day of rash. As the patient recovers, the scabs separate and characteristic pitted scarring gradually develops. The scars are most evident on the face and result from the destruction of sebaceous glands followed by shrinking of granulation tissue and fibrosis.2

The lesions that first appear in the mouth and pharynx ulcerate quickly because of the absence of a stratum corneum, releasing large amounts of virus into the saliva.22 Virus titers in saliva are highest during the first week of illness, corresponding with the period during which patients are most infectious. Although the virus in some instances can be detected in swabs taken from the oropharynx as many as 5 to 6 days before the rash develops,22 transmission does not occur during this period.

Except for the lesions in the skin and mucous membranes and reticulum cell hyperplasia, other organs are seldom involved. Secondary bacterial infection is not common, and death, which usually occurs during the second week of illness, most likely results from the toxemia associated with circulating immune complexes and soluble variola antigens.2 Encephalitis sometimes ensues that is indistinguishable from the acute perivascular demyelination observed as a complication of infection due to vaccinia, measles, or varicella.23

Neutralizing antibodies can be detected by the sixth day of rash and remain at high titers for many years.24 Hemagglutinin-inhibiting antibodies can be detected on about the sixth day of rash, or about 21 days after infection, and complement-fixing antibodies appear approximately 2 days later. Within 5 years, hemagglutinin-inhibiting antibodies decline to low levels and complement-fixing antibodies rarely persist for longer than 6 months.2

Although at least 90% of smallpox cases are clinically characteristic and readily diagnosed in endemic areas, 2 other forms of smallpox are difficult to recognizehemorrhagic and malignant. Hemorrhagic cases are uniformly fatal and occur among all ages and in both sexes, but pregnant women appear to be unusually susceptible. Illness usually begins with a somewhat shorter incubation period and is characterized by a severely prostrating prodromal illness with high fever and head, back, and abdominal pain. Soon thereafter, a dusky erythema develops, followed by petechiae and frank hemorrhages into the skin and mucous membranes. Death usually occurs by the fifth or sixth day after onset of rash.23

In the frequently fatal malignant form, the abrupt onset and prostrating constitutional symptoms are similar. The confluent lesions develop slowly, never progressing to the pustular stage but remaining soft, flattened, and velvety to the touch. The skin has the appearance of a fine-grained, reddish-colored crepe rubber, sometimes with hemorrhages. If the patient survives, the lesions gradually disappear without forming scabs or, in severe cases, large amounts of epidermis might peel away.23

The illness associated with variola minor is generally less severe, with fewer constitutional symptoms and a more sparse rash.25 A milder form of disease is also seen among those who have residual immunity from previous vaccination. In partially immune persons, the rash tends to be atypical and more scant and the evolution of the lesions more rapid.15

There is little information about how individuals with different types of immune deficiency responded to natural smallpox infection. Smallpox was eradicated before human immunodeficiency virus (HIV) was identified and before suitable techniques became available for measuring cell-mediated immunity. However, it is probable that the underlying cause of some cases of malignant and hemorrhagic smallpox resulted from defective immune responses. Vaccination of immune-deficient persons sometimes resulted in a continually spreading primary lesion, persistent viremia, and secondary viral infection of many organs. One such case is documented to have occurred in a vaccinated soldier who had HIV infection.26

DIAGNOSIS


The discovery of a single suspected case of smallpox must be treated as an international health emergency and be brought immediately to the attention of national officials through local and state health authorities.

The majority of smallpox cases present with a characteristic rash that is centrifugal in distribution, ie, most dense on the face and extremities. The lesions appear during a 1- to 2-day period and evolve at the same rate. On any given part of the body, they are generally at the same stage of development. In varicella (chickenpox), the disease most frequently confused with smallpox, new lesions appear in crops every few days and lesions at very different stages of maturation (ie, vesicles, pustules, and scabs) are found in adjacent areas of skin. Varicella lesions are much more superficial and are almost never found on the palms and soles. The distribution of varicella lesions is centripetal, with a greater concentration of lesions on the trunk than on the face and extremities.

The signs and symptoms of both hemorrhagic and malignant smallpox were such that smallpox was seldom suspected until more typical cases were seen and it was recognized that a smallpox outbreak was in progress. Hemorrhagic cases were most often initially identified as meningococcemia or severe acute leukemia. Malignant cases likewise posed diagnostic problems, most often being mistaken for hemorrhagic chickenpox or prompting surgery because of severe abdominal pain.

Laboratory confirmation of the diagnosis in a smallpox outbreak is important. Specimens should be collected by someone who has recently been vaccinated (or is vaccinated that day) and who wears gloves and a mask. To obtain vesicular or pustular fluid, it is often necessary to open lesions with the blunt edge of a scalpel. The fluid can then be harvested on a cotton swab. Scabs can be picked off with forceps. Specimens should be deposited in a vacutainer tube that should be sealed with adhesive tape at the juncture of stopper and tube. This tube, in turn, should be enclosed in a second durable, watertight container. State or local health department laboratories should immediately be contacted regarding the shipping of specimens. Laboratory examination requires high-containment (BL-4) facilities and should be undertaken only in designated laboratories with the appropriate training and equipment. Once it is established that the epidemic is caused by smallpox virus, clinically typical cases would not require further laboratory confirmation.

Smallpox infection can be rapidly confirmed in the laboratory by electron microscopic examination of vesicular or pustular fluid or scabs. Although all orthopoxviruses exhibit identically appearing brick-shaped virions, history taking and clinical picture readily identify cowpox and vaccinia. Although smallpox and monkeypox virions may be indistinguishable, naturally occurring monkeypox is found only in tropical rain forest areas of Africa. Definitive laboratory identification and characterization of the virus involves growth of the virus in cell culture or on chorioallantoic egg membrane and characterization of strains by use of various biologic assays, including polymerase chain reaction techniques and restriction fragment-length polymorphisms.27-29 The latter studies can be completed within a few hours.

PREEXPOSURE PREVENTIVE VACCINATION


Before 1972, smallpox vaccination was recommended for all US children at age 1 year. Most states required that each child be vaccinated before school entry. The only other requirement for vaccination was for military recruits and tourists visiting foreign countries. Most countries required that the individual be successfully vaccinated within a 3-year period prior to entering the country. Routine vaccination in the United States stopped in 1972 and since then, few persons younger than 27 years have been vaccinated. The US Census Bureau reported that in 1998, approximately 114 million persons, or 42% of the US population, were aged 29 years or younger.30

In addition, the immune status of those who were vaccinated more than 27 years ago is not clear. The duration of immunity, based on the experience of naturally exposed susceptible persons, has never been satisfactorily measured. Neutralizing antibodies are reported to reflect levels of protection, although this has not been validated in the field. These antibodies have been shown to decline substantially during a 5- to 10-year period.24 Thus, even those who received the recommended single-dose vaccination as children do not have lifelong immunity. However, among a group who had been vaccinated at birth and at ages 8 and 18 years as part of a study, neutralizing antibody levels remained stable during a 30-year period.31 Because comparatively few persons today have been successfully vaccinated on more than 1 occasion, it must be assumed that the population at large is highly susceptible to infection.

In the United States, a limited reserve supply of vaccine that was produced by Wyeth Laboratories, Lancaster, Pa, in the 1970s is in storage. This supply is believed to be sufficient to vaccinate between 6 and 7 million persons. This vaccine, now under the control of the CDC, consists of vaccine virus (New York Board of Health strain) grown on scarified calves. After purification, it was freeze-dried in rubber-stoppered vials that contain sufficient vaccine for at least 50 doses when a bifurcated needle is used. It is stored at -20°C (James LeDuc, PhD, oral communication, 1998). Although quantities of vaccine have also been retained by a number of other countries, none have reserves large enough to meet more than their own potential emergency needs. WHO has 500,000 doses.32

There are no manufacturers now equipped to produce smallpox vaccine in large quantities. The development and licensure of a tissue cell culture vaccine and the establishment of a new vaccine production facility is estimated to require at least 36 months (Thomas Monath, MD, unpublished data, 1999).

Because of the small amounts of vaccine available, a preventive vaccination program to protect individuals such as emergency and health care personnel is not an option at this time. When additional supplies of vaccine are procured, a decision to undertake preventive vaccination of some portion of the population will have to weigh the relative risk of vaccination complications against the threat of contracting smallpox.

A further deterrent to extensive vaccination is the fact that presently available supplies of vaccinia immune globulin (VIG), also maintained by the CDC, are very limited in quantity. The working group recommends VIG for the treatment of severe cutaneous reactions occurring as a complication of vaccination.33, 34 Vaccinia immune globulin has also been given along with vaccination to protect those who needed vaccination but who were at risk of experiencing vaccine-related complications.33 It has been estimated that if 1 million persons were vaccinated, as many as 250 persons would experience adverse reactions of a type that would require administration of VIG (James LeDuc, PhD, oral communication, 1998). How much VIG would be needed to administer with vaccine to those at risk is unknown.

POSTEXPOSURE THERAPY


At this time, the best that can be offered to the patient infected with smallpox is supportive therapy plus antibiotics as indicated for treatment of occasional secondary bacterial infections. No antiviral substances have yet proved effective for the treatment of smallpox, and the working group is not aware of any reports that suggest any antiviral product is therapeutic. Encouraging initial reports in the 1960s describing the therapeutic benefits of the thiosemicarbazones, cytosine arabinoside, and adenine arabinoside proved questionable on further study.21, 35, 36

Recent studies on tissue culture, mice, and a small number of monkeys have suggested the possibility that cidofovir, a nucleoside analog DNA polymerase inhibitor, might prove useful in preventing smallpox infection if administered within 1 or 2 days after exposure (John Huggins, PhD, oral communication, 1998). At this time, there is no evidence that cidofovir is more effective than vaccination in this early period. Moreover, the potential utility of this drug is limited, given the fact that it must be administered intravenously and its use is often accompanied by serious renal toxicity.37

POSTEXPOSURE INFECTION CONTROL


A smallpox outbreak poses difficult public health problems because of the ability of the virus to continue to spread throughout the population unless checked by vaccination and/or isolation of patients and their close contacts.

A clandestine aerosol release of smallpox, even if it infected only 50 to 100 persons to produce the first generation of cases, would rapidly spread in a now highly susceptible population, expanding by a factor of 10 to 20 times or more with each generation of cases.2, 10, 38 Between the time of an aerosol release of smallpox virus and diagnosis of the first cases, an interval as long as 2 weeks or more is apt to occur because of the average incubation period of 12 to 14 days and the lapse of several additional days before a rash was sufficiently distinct to suggest the diagnosis of smallpox. By that time, there would be no risk of further environmental exposure from the original aerosol release because the virus is fully inactivated within 2 days.

As soon as the diagnosis of smallpox is made, all individuals in whom smallpox is suspected should be isolated immediately and all household and other face-to-face contacts should be vaccinated and placed under surveillance. Because the widespread dissemination of smallpox virus by aerosol poses a serious threat in hospitals, patients should be isolated in the home or other nonhospital facility whenever possible. Home care for most patients is a reasonable approach, given the fact that little can be done for a patient other than to offer supportive therapy.

In the event of an aerosol release of smallpox and a subsequent outbreak, the rationale for vaccinating patients suspected to have smallpox at this time is to ensure that some with a mistaken diagnosis are not placed at risk of acquiring smallpox. Vaccination administered within the first few days after exposure and perhaps as late as 4 days may prevent or significantly ameliorate subsequent illness.39 An emergency vaccination program is also indicated that would include all health care workers at clinics or hospitals that might receive patients; all other essential disaster response personnel, such as police, firefighters, transit workers, public health staff, and emergency management staff; and mortuary staff who might have to handle bodies. The working group recommends that all such personnel for whom vaccination is not contraindicated should be vaccinated immediately irrespective of prior vaccination status.

Vaccination administered within 4 days of first exposure has been shown to offer some protection against acquiring infection and significant protection against a fatal outcome.15 Those who have been vaccinated at some time in the past will normally exhibit an accelerated immune response. Thus, it would be prudent, when possible, to assign those who had been previously vaccinated to duties involving close patient contact.

It is important that discretion be used in identifying contacts of patients to ensure, to the extent that is possible, that vaccination and adequate surveillance measures are focused on those at greatest risk. Specifically, it is recommended that contacts be defined as persons who have been in the same household as the infected individual or who have been in face-to-face contact with the patient after the onset of fever. Experience during the smallpox global eradication program showed that patients did not transmit infection until after the prodromal fever had given way to the rash stage of illness.17, 18

Isolation of all contacts of exposed patients would be logistically difficult and, in practice, should not be necessary. Because contacts, even if infected, are not contagious until onset of rash, a practical strategy calls for all contacts to have temperatures checked at least once each day, preferably in the evening. Any increase in temperature higher than 38°C (101°F) during the 17-day period following last exposure to the case would suggest the possible development of smallpox2 and be cause for isolating the patient immediately, preferably at home, until it could be determined clinically and/or by laboratory examination whether the contact had smallpox. All close contacts of the patients should be promptly vaccinated.

Although cooperation by most patients and contacts in observing isolation could be ensured through counseling and persuasion, there may be some for whom forcible quarantine will be required. Some states and cities in the United States, but not all, confer broad discretionary powers on health authorities to ensure the safety of the public's health and, at one time, this included powers to quarantine. Under epidemic circumstances, this could be an important power to have. Thus, each state and city should review its statutes as part of its preparedness activities.

During the smallpox epidemics in the 1960s and 1970s in Europe, there was considerable public alarm whenever outbreaks occurred and, often, a demand for mass vaccination throughout a very widespread area, even when the vaccination coverage of the population was high.2 In the United States, where few people now have protective levels of immunity, such levels of concern must be anticipated. However, the US vaccine supply is limited at present; thus, vaccine would have to be carefully conserved and used in conjunction with measures to implement rapid isolation of smallpox patients.

HOSPITAL EPIDEMIOLOGY AND INFECTION CONTROL


Smallpox transmission within hospitals has long been recognized as a serious problem. For this reason, separate hospitals for smallpox patients were used for more than 200 years. Throughout the 1970s, both England and Germany had fully equipped standby hospitals in case smallpox should be imported.2 Infections acquired in hospitals may occur as the result of droplets spread from patients to staff and visitors in reasonably close contact or by a fine particle aerosol. In 1 such occurrence in Germany, a smallpox patient with a cough, although isolated in a single room, infected persons on 3 floors of a hospital.10 Persons with the usually fatal hemorrhagic or malignant forms of the disease pose a special problem because they often remain undiagnosed until they are near death and extremely contagious. A number of outbreaks have occurred in laundry workers who handled linens and blankets used by patients.15 The working group recommends that in an outbreak setting, all hospital employees as well as patients in the hospital be vaccinated. For individuals who are immunocompromised or for whom vaccination is otherwise contraindicated, VIG should be provided, if available. If it is not available, a judgment will have to be made regarding the relative risks of acquiring the disease in contrast with the risks associated with vaccination.

In the event of a limited outbreak with few cases, patients should be admitted to the hospital and confined to rooms that are under negative pressure and equipped with high-efficiency particulate air filtration. In larger outbreaks, home isolation and care should be the objective for most patients. However, not all will be able to be so accommodated and, to limit nosocomial infections, authorities should consider the possibility of designating a specific hospital or hospitals for smallpox care. All persons isolated as such and those caring for them should be immediately vaccinated. Employees for whom vaccination is contraindicated should be furloughed.

Standard precautions using gloves, gowns, and masks should be observed. All laundry and waste should be placed in biohazard bags and autoclaved before being laundered or incinerated. A special protocol should be developed for decontaminating rooms after they are vacated by patients (see "Decontamination" section).

Laboratory examination requires high-containment (BL-4) facilities and should be undertaken only in designated laboratories with the appropriate trained personnel and equipment. Specific recommendations for safe specimen transport are described in the section on "Differential Diagnosis and Diagnostic Tests."

Protecting against the explosive spread of virus from the hemorrhagic or malignant case is difficult. Such cases occurring during the course of an outbreak may be detected if staff is alert to the possibility that any severe, acute, prostrating illness must be considered smallpox until proven otherwise.

Patients who die of smallpox should be cremated whenever possible and mortuary workers should be vaccinated.

VACCINE ADMINISTRATION AND COMPLICATIONS


Smallpox vaccine is currently approved by the US Food and Drug Administration (FDA) for use only in persons in special-risk categories, including laboratory workers directly involved with smallpox or closely related orthopoxviruses. Under epidemic circumstances, widespread vaccination would be indicated, as recommended by the working group.

Vaccination has been successfully and safely administered to persons of all ages, from birth onward.40 However, there are certain groups for whom elective vaccination has not been recommended because of the risk of complications. Under epidemic circumstances, however, such contraindications will have to be weighed against the grave risks posed by smallpox. If available, VIG can be administered concomitantly with vaccination to minimize the risk of complications in these persons.

Vaccination is normally performed using the bifurcated needle (Figure 3). A sterile needle is inserted into an ampoule of reconstituted vaccine and, on withdrawal, a droplet of vaccine sufficient for vaccination is held by capillarity between the 2 tines. The needle is held at right angles to the skin; the wrist of the vaccinator rests against the arm. Fifteen perpendicular strokes of the needle are rapidly made in an area of about 5 mm in diameter.41, 42 The strokes should be sufficiently vigorous so that a trace of blood appears at the vaccination site after 15 to 30 seconds. After vaccination, excess vaccine should be wiped from the site with gauze that should be discarded in a hazardous waste receptacle. The site should be covered with a loose, nonocclusive bandage to deter the individual from touching the site and perhaps transferring virus to other parts of the body.

After about 3 days, a red papule appears at the vaccination site and becomes vesicular on about the fifth day (Figure 4). By the seventh day, it becomes the typical Jennerian pustulewhitish, umbilicated, multilocular, containing turbid lymph and surrounded by an erythematous areola that may continue to expand for 3 more days. Regional lymphadenopathy and fever is not uncommon. As many as 70% of children have 1 or more days of temperature higher than 39°C (100°F) between days 4 and 14.43 The pustule gradually dries, leaving a dark crust, which normally falls off after about 3 weeks.

A successful vaccination for those with partial immunity may manifest a gradient of responses. These range from what appears to be a primary take (as described herein) to an accelerated reaction in which there may be little more than a papule surrounded by erythema that reaches a peak between 3 and 7 days. A response that reaches a peak in erythema within 48 hours represents a hypersensitivity reaction and does not signify that growth of the vaccinia virus has occurred.2 Persons exhibiting such a reaction should be revaccinated.

Complications

The frequency of complications associated with use of the New York Board of Health strain (the strain used throughout the United States and Canada for vaccine) is the lowest for any established vaccinia virus strain, but the risks are not inconsequential.44, 45 Data on complications gathered by the CDC in 1968 are shown in Table 1. Complications occurred most frequently among primary vaccinees.

Postvaccinial Encephalitis.
Postvaccinial encephalitis occurred at a rate of 1 case per 300,000 vaccinations and was observed only in primary vaccinees; one fourth of these cases were fatal and several had permanent neurological residua. Between 8 and 15 days after vaccination, encephalitic symptoms developedfever, headache, vomiting, drowsiness, and, sometimes, spastic paralysis, meningitic signs, coma, and convulsions. Cerebrospinal fluid usually showed a pleocytosis. Recovery was either complete or associated with residual paralysis and other central nervous system symptoms and, sometimes, death. There was no treatment.

Progressive Vaccinia (Vaccinia Gangrenosa).
Cases of progressive vaccinia occurred both among primary vaccinees and revaccinees. It was a frequently fatal complication among those with immune deficiency disorders. The vaccinial lesion failed to heal and progressed to involve adjacent skin with necrosis of tissue, spreading to other parts of the skin, to bones, and to viscera. Vaccinia immune globulin was used for this problem.34, 46 One case in a soldier with acquired immunodeficiency syndrome was successfully treated with VIG and ribavirin. These treatment strategies were off-label and would be considered experimental.26

Eczema Vaccinatum.
A sometimes serious complication, eczema vaccinatum occurred in some vaccinees and contacts with either active or healed eczema. Vaccinial skin lesions extended to cover all or most of the area once or currently afflicted with eczema. Vaccinia immune globulin was therapeutic.46

Generalized Vaccinia.
A secondary eruption almost always following primary vaccination, generalized vaccinia resulted from blood-borne dissemination of virus. Lesions emerged between 6 and 9 days after vaccination and were either few in number or generalized. This complication was usually self-limited. In severe cases, VIG was indicated.46

Inadvertent Inoculation.
Transmission to close contacts or autoinoculation to sites such as face, eyelid, mouth, and genitalia sometimes occurred. Most lesions healed without incident, although VIG was useful in some cases of periocular implantation.

Miscellaneous.
Many different rashes have been associated with vaccination. Most common are erythema multiforme and variously distributed urticarial, maculopapular, and blotchy erythematous eruptions, which normally clear without therapy.

Groups at Special Risk for Complications

Consensus recommendations for special-risk groups as set forth herein reflect the best clinical and science-based judgment of the working group and do not necessarily correspond to FDA-approved uses.

Five groups of persons are ordinarily considered at special risk of smallpox vaccine complications: (1) persons with eczema or other significant exfoliative skin conditions; (2) patients with leukemia, lymphoma, or generalized malignancy who are receiving therapy with alkylating agents, antimetabolites, radiation, or large doses of corticosteroids; (3) patients with HIV infection; (4) persons with hereditary immune deficiency disorders; and (5) pregnant women. If persons with contraindications have been in close contact with a smallpox patient or the individual is at risk for occupational reasons, VIG, if available, may be given simultaneously with vaccination in a dose of 0.3 mL/kg of body weight to prevent complications. This does not alter vaccine efficacy. If VIG is not available, vaccine administration may still be warranted, given the far higher risk of an adverse outcome from smallpox infection than from vaccination.

VIG Therapy for Complications

Vaccinia immune globulin is valuable in treating patients with progressive vaccinia, eczema vaccinatum, severe generalized vaccinia, and periocular infections resulting from inadvertent inoculation. It is administered intramuscularly in a dose of 0.6 mL/kg of body weight. Because the dose is large (eg, 42 mL for a person weighing 70 kg), the product is given intramuscularly in divided doses over a 24- to 36-hour period and may be repeated, if necessary, after 2 to 3 days if improvement is not occurring.47 Because the availability of VIG is so limited, its use should be reserved for the most serious cases. Vaccinia immune globulin, as well as vaccinia vaccine, is made available by the CDC through state health departments. Consultative assistance in the diagnosis and management of patients with complications can be obtained through state health departments.

DECONTAMINATION


Vaccinia virus, if released as an aerosol and not exposed to UV light, may persist for as long as 24 hours or somewhat longer under favorable conditions.9 It is believed that variola virus would exhibit similar properties. However, by the time patients had become ill and it had been determined that an aerosol release of smallpox virus had occurred, there would be no viable smallpox virus in the environment. Vaccinia virus, if released as an aerosol, is almost completely destroyed within 6 hours in an atmosphere of high temperature (31°C-33°C) and humidity (80%) (Table 2).9 In cooler temperatures (10°C-11°C) and lower humidity (20%), nearly two thirds of a vaccinia aerosol survives for as long as 24 hours.9 It is believed that variola would behave similarly.

The occurrence of smallpox infection among personnel who handled laundry from infected patients is well documented15 and it is believed that virus in such material remains viable for extended periods. Thus, special precautions need to be taken to ensure that all bedding and clothing of smallpox patients is autoclaved or laundered in hot water to which bleach has been added. Disinfectants that are used for standard hospital infection control, such as hypochlorite and quaternary ammonia, are effective for cleaning surfaces possibly contaminated with virus.

Virus in scabs is more durable. At a temperature of 35°C and 65% relative humidity, the virus has persisted for 3 weeks.48 At cooler temperatures (26°C), the virus has survived for 8 weeks at high relative humidity and 12 weeks at a relative humidity less than 10%.48 Dutch investigators demonstrated that it was possible to isolate variola virus from scabs that had been sitting on a shelf for 13 years.49 It is unlikely, however, that the smallpox virus, bound in the fibrin matrix of a scab, is infectious in humans. This is borne out by studies conducted during the eradication program and by surveillance for cases in newly smallpox-free areas.2 It was reasoned that if the virus were able to persist in nature and infect humans, there would be cases occurring for which no source could be identified. Cases of this type were not observed. Rather, when cases were found, there were antecedent human cases with whom they had direct contact.

RESEARCH


Priority should be directed to 3 areas of smallpox research: vaccines, immunotherapy and drugs, and diagnostics.

The working group recommends that an emergency stockpile of at least 40 million doses of vaccine and a standby manufacturing capacity to produce more is a critical need. At a minimum, this quantity of vaccine would be needed in the control of an epidemic during the first 4 to 8 weeks after an attack. Smallpox vaccine, contained in glass-sealed ampoules and stored at -20°C, retains its potency almost indefinitely. However, several steps are necessary before manufacturing can begin. The traditional method for producing vaccine on the scarified flank of a calf is no longer acceptable because the product inevitably contains some microbial contaminants, however stringent the purification measures. Contemporary vaccines require the use of tissue cell cultures. Thus, as a first step, the traditional New York Board of Health strain needs to be grown in a suitable tissue cell culture and comparative studies performed of the reactogenicity and immunogenicity of calf-derived and tissue cell culture vaccines. This should be a comparatively straightforward exercise. The cost of such a stockpile should be comparatively modest because the vaccine would be packaged in 50-dose rather than costly single-dose containers. In the mid-1970s, 40 million doses would have cost less than $5 million (D.A.H., unpublished data, 1975).

The frequency of vaccine complications is sufficiently great to recommend development, if possible, of a more attenuated strain that, hopefully, would retain full efficacy. Development of an entirely new, genetically engineered strain would be both costly and time consuming. Moreover, it would be difficult at this time to justify its use in large numbers of human subjects to evaluate safety. There is, however, a candidate attenuated strain that was developed and field tested in Japan in the mid-1970s (a Lister strain–derived vaccine50 that has been produced in volume in rabbit kidney cell culture and has been given to more than 100,000 persons in Japan). Research showed no severe complications among the first 30,000 vaccinees.51 The cutaneous responses to vaccination were much less severe and far fewer vaccinees developed fever. More than 95% developed a Jennerian pustule; immunogenicity, as measured by neutralizing antibody, was slightly lower than for nonattenuated strains.

Vaccinia immune globulin has been used for the treatment of vaccine complications and for administration with vaccine to those for whom vaccine is otherwise contraindicated. Production of VIG should be a high priority for research. An alternative to VIG is also needed because VIG is difficult to produce and cumbersome to administer. Immunotherapy using humanized monoclonal antibodies is an alternative that should be explored. Studies of antiviral agents or drugs, already approved or near approval for marketing for use in other viral diseases, have suggested that 1 or more such products might prove useful.

Finally, a simple, rapid diagnostic test to identify variola virus in the oropharynx during the prodrome or early in the exanthematous phase of illness would be of considerable help in triage of suspected patients during the course of an outbreak.

SUMMARY


The specter of resurgent smallpox is ominous, especially given the enormous efforts that have been made to eradicate what has been characterized as the most devastating of all the pestilential diseases. Unfortunately, the threat of an aerosol release of smallpox is real and the potential for a catastrophic scenario is great unless effective control measures can quickly be brought to bear.

Early detection, isolation of infected individuals, surveillance of contacts, and a focused selective vaccination program are the essential items of a control program. Educating health care professionals about the diagnostic features of smallpox should permit early detection; advance regionwide planning for isolation and care of infected individuals in their homes as appropriate and in hospitals when home care is not an option will be critical to deter spread. Ultimately, success in controlling a burgeoning epidemic will depend on the availability of adequate supplies of vaccine and VIG. An adequate stockpile of those commodities would offer a relatively inexpensive safeguard against tragedy.


Author/Article Information


Author Affiliations: The Center for Civilian Biodefense Studies (Drs Henderson, Inglesby, Bartlett, O'Toole, Perl, and Russell), and the Schools of Public Health (Drs Henderson, O'Toole, and Russell) and Medicine (Drs Inglesby, Bartlett, and Perl), Johns Hopkins University, Baltimore, Md; Viral and Rickettsial Diseases, California Department of Health, Berkeley (Dr Ascher); US Army Medical Research Institute of Infectious Diseases, Frederick, Md (Drs Eitzen, Jahrling, and Parker); Office of Emergency Management (Mr Hauer) and Office of Communicable Disease, New York City Health Department (Dr Layton), New York, NY; Centers for Disease Control and Prevention, Atlanta, Ga (Dr McDade); Acute Disease Epidemiology, Minnesota Department of Health, Minneapolis (Dr Osterholm); and Office of Emergency Preparedness, Department of Health and Human Services, Rockville, Md (Dr Tonat).

Corresponding Author and Reprints: Donald A. Henderson, MD, MPH, Johns Hopkins Center for Civilian Biodefense Studies, Johns Hopkins University, Candler Bldg, Suite 850, 111 Market Pl, Baltimore, MD 21202 (e-mail: dahzero@aol.com).

Ex Officio Participants in the Working Group on Civilian Biodefense: George Curlin, MD, National Institutes of Health, Bethesda, Md; Margaret Hamburg, MD, and William Roub, PhD, Office of Assistant Secretary for Planning and Evaluation, DHHS, Washington, DC; Robert Knouss, MD, Office of Emergency Preparedness, DHHS, Rockville, Md; Marcelle Layton, MD, Office of Communicable Disease, New York City Health Department, New York, NY; and Brian Malkin and Stuart Nightingale, MD, FDA, Rockville, Md.

Funding/Support: Funding for this study primarily was provided by each participant's institution or agency. The Johns Hopkins Center for Civilian Biodefense Studies provided travel funds for 3 members of the group.

Disclaimers: In many cases, the indication and dosages and other information are not consistent with current FDA-approved labeling. The recommendations on the use of drugs and vaccine for uses not approved by the FDA do not represent the official views of the FDA or of any of the federal agencies whose scientists participated in these discussions. Unlabeled uses of the products recommended are noted in the sections of this article in which these products are discussed. Where unlabeled uses are indicated, information used as the basis for the recommendations is discussed.

The views, opinions, assertions, and findings contained herein are those of the authors and should not be construed as official US Department of Defense or US Department of Army positions, policies, or decisions unless so designated by other documentation.

Additional Articles: This article is second in a series entitled Medical and Public Health Management Following the Use of a Biological Weapon: Consensus Statements of the Working Group on Civilian Biodefense. See reference 1.

Acknowledgment: The working group wishes to thank Isao Arita, MD, Agency for Cooperation in International Health, Kumamoto, Japan; Joel Bremen, MD, DTPH, Fogarty International Center, Bethesda; Joseph Esposito, PhD, and Brian Mahy, PhD, ScD, CDC, Atlanta, Ga; Frank Fenner, MD, Australian National University, Canberra; and Ralph Henderson, MD, WHO, Geneva, Switzerland.


REFERENCES


1.
Inglesby TV, Henderson DA, Bartlett JG, et al.
Anthrax as a biological weapon: medical and public health management.
JAMA.
1999;281:1735-1745.
ABSTRACT  |  FULL TEXT  |  PDF  |  MEDLINE

2.
Fenner F, Henderson DA, Arita I, Jezek Z, Ladnyi ID.
Smallpox and Its Eradication.
Geneva, Switzerland: World Health Organization; 1988:1460.

3.
Stearn EW, Stearn AE.
The Effect of Smallpox on the Destiny of the Amerindian.
Boston, Mass: Bruce Humphries; 1945.

4.
Hopkins DR.
Princes and Peasants.
Chicago, Ill: University of Chicago Press; 1983.

5.
World Health Organization.
The Global Eradication of Smallpox: Final Report of the Global Commission for the Certification of Smallpox Eradication.
Geneva, Switzerland: World Health Organization; 1980.

6.
Breman JG, Henderson DA.
Poxvirus dilemmas: monkeypox, smallpox and biological terrorism.
N Engl J Med.
1998;339:556-559.
MEDLINE

7.
Institute of Medicine.
Assessment of Future Scientific Need for Live Variola Virus.
Washington, DC: National Academy Press; 1999.

8.
Alibek K.
Biohazard.
New York, NY: Random House Inc; 1999.

9.
Harper GJ.
Airborne micro-organisms: survival test with four viruses.
J Hyg.
1961;59:479-486.

10.
Wehrle PF, Posch J, Richter KH, Henderson DA.
An airborne outbreak of smallpox in a German hospital and its significance with respect to other recent outbreaks in Europe.
Bull World Health Organ.
1970;43:669-679.
MEDLINE

11.
Chapin CV, Smith J.
Permanency of the mild type of smallpox.
J Prev Med.
1932;1:1-29.

12.
Esposito JJ, Knight JC.
Orthopox DNA: a comparison of restriction profiles and maps.
Virology.
1985;143:230-251.
MEDLINE

13.
Chapin CV.
Variation in the type of infectious disease as shown by the history of smallpox in the United States, 1895-1912.
J Infect Dis.
1913;13:171-196.

14.
Anders W, Sosch J.
Die Pockenausbrucke 1961/61 in Nordrhein-Westfalen.
Bundesgesundheitsblatt.
1962;17:265-269.

15.
Dixon CW.
Smallpox.
London, England: J & A Churchill Ltd; 1962:1460.

16.
Joarder AK, Tarantola D, Tulloch J.
The Eradication of Smallpox From Bangladesh, New Delhi.
Geneva, Switzerland: WHO Regional Publications; 1980.

17.
Mack TM.
Smallpox in Europe, 1950-71.
J Infect Dis.
1972;125:161-169.
MEDLINE

18.
Mack TM, Thomas DB, Khan MM.
Epidemiology of smallpox in West Pakistan, II: determinants of intravillage spread other than acquired immunity.
Am J Epidemiol.
1972;95:157-168.
MEDLINE

19.
Rao AR.
Infected Inanimate Objects (Fomites) and Their Role in Transmission of Smallpox.
Geneva, Switzerland: World Health Organization; 1972. WHO/SE/72.40.

20.
Fenner F, Wittek R, Dumbell KR.
The Orthopoxviruses.
San Diego, Calif: Academic Press; 1988:432.

21.
Jezek Z, Fenner F.
Human Monkeypox.
Basel, Switzerland: S Karger; 1988.

22.
Sarkar JK, Mitra AC, Mukherjee MK, De SK.
Virus excretion in smallpox, 2: excretion in the throat of household contacts.
Bull World Health Organ.
1973;48:523-527.
MEDLINE

23.
Rao AR.
Smallpox.
Bombay, India: Kothari Book Depot; 1972.

24.
Downie AW, McCarthy K.
The antibody response in man following infection with viruses of the pox group, III: antibody response in smallpox.
J Hyg.
1958;56:479-487.

25.
Marsden JP.
Variola minor: a personal analysis of 13,686 cases.
Bull Hyg.
1948;23:735-746.

26.
Redfield RR, Wright CD, James WD, Jones ST, Brown C, Burke D.
Disseminated vaccinia in a military recruit with human immunodeficiency virus (HIV).
N Engl J Med.
1987;316:673-676.
MEDLINE

27.
Esposito JJ, Massung RF.
Poxvirus infections in humans.
In: Murray PR, Tenover F, Baron EJ, eds. Clinical Microbiology.Washington, DC: American Society of Microbiology; 1995:1131-1138.

28.
Knight JC, Massung RF, Esposito JJ.
Polymerase chain reaction identification of smallpox virus.
In: PCR: Protocols for Diagnosis of Human and Animal Viral DiseaseHeidelberg, Germany: Springer-Verlag; 1995:297-302.

29.
Ropp SL, Knight JC, Massung RF, Esposito JJ.
PCR strategy for identification and differentiation of smallpox and other orthopoxviruses.
J Clin Microbiol.
1995;33:2069-2076.
MEDLINE

30.
US Bureau of the Census.
Resident Population of the United States: Estimates, by Age and Sex.
Washington, DC: US Bureau of the Census; 1998.

31.
El-Ad R, Roth Y, Winder A.
The persistence of neutralizing antibodies after revaccination against smallpox.
J Infect Dis.
1990;161:446-448.
MEDLINE

32.
World Health Organization.
Smallpox vaccine and seed virus survey.
Working document for the meeting of the WHO Ad Hoc Expert Committee on Orthopoxvirus Infections; January 14-15, 1999; Geneva, Switzerland.

33.
Sharp JCM, Fletcher WB.
Experience of antivaccinia immunoglobulin in the United Kingdom.
Lancet.
1973;1:656-659.
MEDLINE

34.
Kempe CH.
Studies on smallpox and complications of smallpox vaccination.
Pediatrics.
1960;26:176-189.

35.
Koplan J, Monsur KA, Foster SO, et al.
Treatment of variola major with adenine arabinoside.
J Infect Dis.
1975;131:34-39.
MEDLINE

36.
Monsur KA, Hossain MS, Huq F, Rahaman MM, Haque MQ.
Treatment of variola major with cytosine arabinoside.
J Infect Dis.
1975;131:40-43.
MEDLINE

37.
Lalezari JP, Staagg RJ, Kuppermann BD, et al.
Intravenous cidofovir for peripheral cytomegalovirus retinitis in patients with AIDS: a randomized, controlled trial.
Ann Intern Med.
1997;126:257-263.
MEDLINE

38.
O'Toole T.
Smallpox: a case history.
Emerg Infect Dis.
In press.

39.
Dixon CW.
Smallpox in Tripolitania, 1946: an epidemiological and clinical study of 500 cases, including trials of penicillin treatment.
J Hyg.
1948;46:351-377.

40.
Centers for Disease Control and Prevention.
Vaccinia (smallpox) vaccine recommendations of the immunization practices advisory committee.
MMWR Morb Mortal Wkly Rep.
1990;40(RR-14):445-448.

41.
World Health Organization.
WHO Expert Committee on Smallpox Eradication.
Geneva, Switzerland: World Health Organization; 1972:493. WHO technical report series.

42.
Henderson DA, Arita I, Shafa E.
Studies of the bifurcated needle and recommendations for its use.
Geneva, Switzerland: World Health Organization; 1972. WHO Smallpox Eradications Paper SE/72.5.

43.
McIntosh K, Cherry JD, Benenson AS.
Standard percutaneous (smallpox) revaccination of children who received primary percutaneous vaccination.
J Infect Dis.
1990;161:445-448.

44.
Wyeth Smallpox Vaccine [package insert].
Lancaster, Pa: Wyeth Laboratories Inc; 1988.

45.
Lane JM, Ruben FL, Neff JM, Millar JD.
Complications of smallpox vaccination, 1968: national surveillance in the United States.
N Engl J Med.
1969;281:1201-1208.
MEDLINE

46.
Goldstein VA, Neff JM, Lande JM, Koplan J.
Smallpox vaccination reactions, prophylaxis and therapy of complications.
Pediatrics.
1975;55:342-347.
MEDLINE

47.
Centers for Disease Control and Prevention.
Vaccinia (smallpox) vaccine: recommendations of the Immunization Practices Advisory Committee.
MMWR Morb Mortal Wkly Rep.
1991;40:1-10.

48.
Huq F.
Effect of temperature and relative humidity on variola virus in crusts.
Bull World Health Organ.
1976;54:710-712.
MEDLINE

49.
Wolff HL, Croon JJ.
The survival of smallpox virus (variola minor) in natural circumstances.
Bull World Health Organ.
1968;38:492-493.
MEDLINE

50.
Hashizume S, Yoshizawa H, Morita M, Suzuki K.
Properties of attenuated mutant of vaccinia virus, LC16m8, derived from Lister strain.
In: Quinnan GV, ed. Vaccine Virus as Vectors for Vaccine Antigens.Amsterdam, the Netherlands: Elsevier Science Publishing; 1985:87-99.

51.
Hirayama M.
Smallpox vaccination in Japan.
In: Fukumi H, ed. The Vaccination: Theory and PracticeTokyo: International Medical Foundation of Japan; 1975:113-124.



9 posted on 10/08/2001 4:36:19 AM PDT by vannrox
[ Post Reply | Private Reply | To 8 | View Replies]

To: samantha06

document.write(logcode);Journals of AMA HomeFeedbackSite Map

SubscribeRegisterSearchDocument DeliveryE-Mail AlertClassified
How to use this site
Previous Vol. 285 No. 21,
June 6, 2001

Next
JAMACurrent IssueIndexesPast Issue Index
Consensus Statement




Printable version of this item

See Related:
Authors' Articles

Return to
Table of Contents

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom






























to TOP

ABSTRACT

INTRODUCTION

CONSENSUS METHODS

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON

EPIDEMIOLOGY

MICROBIOLOGY AND VIRULENCE FACTORS

PATHOGENESIS AND CLINICAL MANIFESTATIONS

DIAGNOSIS

VACCINATION

TREATMENT

POSTEXPOSURE ANTIBIOTIC RECOMMEND-
ATIONS

INFECTION CONTROL

ENVIRONMENTAL DECONTAMINATION AND PROTECTION

ADDITIONAL RESEARCH

AUTHOR/ARTICLE INFORMATION

REFERENCES

INDEX OF FIGURES AND TABLES
to bottom



Tularemia as a Biological Weapon

Medical and Public Health Management

Author Information  David T. Dennis, MD, MPH; Thomas V. Inglesby, MD; Donald A. Henderson, MD, MPH; John G. Bartlett, MD; Michael S. Ascher, MD; Edward Eitzen, MD, MPH; Anne D. Fine, MD; Arthur M. Friedlander, MD; Jerome Hauer, MHS; Marcelle Layton, MD; Scott R. Lillibridge, MD; Joseph E. McDade, PhD; Michael T. Osterholm, PhD, MPH; Tara O'Toole, MD, MPH; Gerald Parker, PhD, DVM; Trish M. Perl, MD, MSc; Philip K. Russell, MD; Kevin Tonat, DrPH, MPH; for the Working Group on Civilian Biodefense

Objective  The Working Group on Civilian Biodefense has developed consensus-based recommendations for measures to be taken by medical and public health professionals if tularemia is used as a biological weapon against a civilian population.

Participants  The working group included 25 representatives from academic medical centers, civilian and military governmental agencies, and other public health and emergency management institutions and agencies.

Evidence  MEDLINE databases were searched from January 1966 to October 2000, using the Medical Subject Headings Francisella tularensis, Pasteurella tularensis, biological weapon, biological terrorism, bioterrorism, biological warfare, and biowarfare. Review of these references led to identification of relevant materials published prior to 1966. In addition, participants identified other references and sources.

Consensus Process  Three formal drafts of the statement that synthesized information obtained in the formal evidence-gathering process were reviewed by members of the working group. Consensus was achieved on the final draft.

Conclusions  A weapon using airborne tularemia would likely result 3 to 5 days later in an outbreak of acute, undifferentiated febrile illness with incipient pneumonia, pleuritis, and hilar lymphadenopathy. Specific epidemiological, clinical, and microbiological findings should lead to early suspicion of intentional tularemia in an alert health system; laboratory confirmation of agent could be delayed. Without treatment, the clinical course could progress to respiratory failure, shock, and death. Prompt treatment with streptomycin, gentamicin, doxycycline, or ciprofloxacin is recommended. Prophylactic use of doxycycline or ciprofloxacin may be useful in the early postexposure period.

JAMA. 2001;285:2763-2773JST10001

I know of no other infection of animals communicable to man that can be acquired from sources so numerous and so diverse. In short, one can but feel that the status of tularemia, both as a disease in nature and of man, is one of potentiality.R. R. Parker1

Tularemia, a bacterial zoonosis, is the subject of this fifth article in a series providing recommendations for medical and public health management following use of various agents as biological weapons of terrorism.2-5 The causative agent of tularemia, Francisella tularensis, is one of the most infectious pathogenic bacteria known, requiring inoculation or inhalation of as few as 10 organisms to cause disease.6, 7 Humans become incidentally infected through diverse environmental exposures and can develop severe and sometimes fatal illness but do not transmit infection to others. The Working Group on Civilian Biodefense considers F tularensis to be a dangerous potential biological weapon because of its extreme infectivity, ease of dissemination, and substantial capacity to cause illness and death.8-11

CONSENSUS METHODS


The working group comprised 25 representatives from academic medical centers, civilian and military governmental agencies, and other public health and emergency management institutions. This group followed a specified process in developing a consensus statement. MEDLINE databases from January 1966 to October 2000 were searched using the Medical Subject Headings Francisella tularensis, Pasteurella tularensis, biological weapon, biological terrorism, bioterrorism, biological warfare, and biowarfare. Review of the bibliographies of these references led to identification of relevant materials published prior to 1966. In addition, participants identified other published and unpublished references and sources for review.

The first draft of the consensus statement was a synthesis of information obtained in the formal evidence-gathering process. Members of the working group were asked to make written comments on this first draft in May 1999. Subsequent revised drafts were reviewed and edited until full consensus of the working group was achieved.

HISTORY AND POTENTIAL AS A BIOLOGICAL WEAPON


Tularemia was first described as a plaguelike disease of rodents in 1911 and, shortly thereafter, was recognized as a potentially severe and fatal illness in humans.12 Tularemia's epidemic potential became apparent in the 1930s and 1940s, when large waterborne outbreaks occurred in Europe and the Soviet Union13-15 and epizootic-associated cases occurred in the United States.16, 17 As well, F tularensis quickly gained notoriety as a virulent laboratory hazard.18, 19 Public health concerns impelled substantial early investigations into tularemia's ecology, microbiology, pathogenicity, and prevention.19-22

Francisella tularensis has long been considered a potential biological weapon. It was one of a number of agents studied at Japanese germ warfare research units operating in Manchuria between 1932 and 194523; it was also examined for military purposes in the West. A former Soviet Union biological weapons scientist, Ken Alibeck, has suggested that tularemia outbreaks affecting tens of thousands of Soviet and German soldiers on the eastern European front during World War II may have been the result of intentional use.24 Following the war, there were continuing military studies of tularemia. In the 1950s and 1960s, the US military developed weapons that would disseminate F tularensis aerosols10; concurrently, it conducted research to better understand the pathophysiology of tularemia and to develop vaccines and antibiotic prophylaxis and treatment regimens. In some studies, volunteers were infected with F tularensis by direct aerosol delivery systems and by exposures in an aerosol chamber.10 A live attenuated vaccine was developed that partially protected against respiratory and intracutaneous challenges with the virulent SCHU S-4 strain of F tularensis,6, 7 and various regimens of streptomycin, tetracyclines, and chloramphenicol were found to be effective in prophylaxis and treatment.25-27 By the late 1960s, F tularensis was one of several biological weapons stockpiled by the US military.10 According to Alibeck, a large parallel effort by the Soviet Union continued into the early 1990s and resulted in weapons production of F tularensis strains engineered to be resistant to antibiotics and vaccines.24

In 1969, a World Health Organization expert committee estimated that an aerosol dispersal of 50 kg of virulent F tularensis over a metropolitan area with 5 million inhabitants would result in 250 000 incapacitating casualties, including 19 000 deaths.28 Illness would be expected to persist for several weeks and disease relapses to occur during the ensuing weeks or months. It was assumed that vaccinated individuals would be only partially protected against an aerosol exposure. Referring to this model, the Centers for Disease Control and Prevention (CDC) recently examined the expected economic impact of bioterrorist attacks and estimated the total base costs to society of an F tularensis aerosol attack to be $5.4 billion for every 100 000 persons exposed.9

The United States terminated its biological weapons development program by executive order in 1970 and, by 1973, had destroyed its entire biological arsenal.10 Since then, the US Army Medical Research Institute of Infectious Diseases has been responsible for defensive medical research on F tularensis and other potential biological warfare agents to better protect the US military, including protocols on decontamination, prophylaxis, clinical recognition, laboratory diagnosis, and medical management.29 The CDC operates a national program for bioterrorism preparedness and response that incorporates a broad range of public health partnerships.30, 31

EPIDEMIOLOGY


Geographic Distribution and Human Exposures

Tularemia occurs throughout much of North America and Eurasia.15, 21, 22, 32 In the United States, human cases have been reported from every state except Hawaii; however, most cases occur in south-central and western states (especially Missouri, Arkansas, Oklahoma, South Dakota, and Montana).33-35 In Eurasia, the disease is also widely endemic, although the greatest numbers of human cases are reported from northern and central Europe, especially Scandinavian countries and those of the former Soviet Union.36, 37 Tularemia is almost entirely a rural disease, although urban and suburban exposures occasionally do occur.38-41

Throughout its range, F tularensis is found in widely diverse animal hosts and habitats and can be recovered from contaminated water, soil, and vegetation.15, 20-22, 32 A variety of small mammals, including voles, mice, water rats, squirrels, rabbits, and hares, are natural reservoirs of infection. They acquire infection through bites by ticks, flies, and mosquitoes, and by contact with contaminated environments. Although enzootic cycles of F tularensis typically occur without notice, epizootics with sometimes extensive die-offs of animal hosts may herald outbreaks of tularemia in humans.16, 22, 42, 43 Humans become infected with F tularensis by various modes, including bites by infective arthropods,42, 44-47 handling infectious animal tissues or fluids,17, 48, 49 direct contact with or ingestion of contaminated water, food, or soil,13, 20, 40, 50, 51 and inhalation of infective aerosols.43, 52-56 Persons of all ages and both sexes appear to be equally susceptible to tularemia. Certain activities, such as hunting, trapping, butchering, and farming, are most likely to expose adult men. Laboratory workers are especially vulnerable to infection, either by accidentally inoculating themselves or by inhaling aerosolized organisms.18, 22, 56-58 Ordinary exposures during examination of an open culture plate can cause infection. Although F tularensis is highly infectious and pathogenic, its transmission from person to person has not been documented.

Incidence

The worldwide incidence of tularemia is not known, and the disease is probably greatly underrecognized and underreported. In the United States, reported cases have dropped sharply from several thousand per year prior to 1950 to less than 200 per year in the 1990s.33-35 Between 1985 and 1992, 1409 cases and 20 deaths were reported in the United States, for a mean of 171 cases per year and a case-fatality rate of 1.4%.34 Persons in all age groups were affected, but most were children younger than 10 years and adults aged 50 years or older. Of 1298 cases for which information on sex was available, 942 (72.6%) occurred in males, and males outnumbered females in all age groups. Most cases occur in June through September, when arthropod-borne transmission is most common.17, 35, 59 Cases in winter usually occur among hunters and trappers who handle infected animal carcasses.17, 35, 48 In the United States, cases are mostly sporadic or occur in small clusters34, 35, 49; in Eurasia, waterborne, arthropod-borne, and airborne outbreaks involving hundreds of persons have been reported.40, 43, 44, 51, 53-55

Natural Occurrences of Inhalational Tularemia

The largest recorded airborne tularemia outbreak occurred in 1966-1967 in an extensive farming area of Sweden.43 This outbreak involved more than 600 patients infected with strains of the milder European biovar of F tularensis (F tularensis biovar palaearctica) [type B]), most of whom acquired infection while doing farm work that created contaminated aerosols. Case exposures and disease onsets occurred during a period of months but peaked during the winter, when rodent-infested hay was being sorted and moved from field storage sites to barns. Among 140 serologically confirmed cases thought to have been infected by inhalation, most had typical acute symptoms of fever, fatigue, chills, headache, and malaise; only 14 (10%) of confirmed patients had symptoms of pneumonia, such as dyspnea and chest pains. Patients generally responded well to tetracycline, and no deaths were reported. Inhalational tularemia in the United States has involved only single cases or small clusters of cases, variously resulting from laboratory exposures,18, 56, 57 disturbance of contaminated animal carcasses,38, 39, 41 and suspected infective environmental aerosols.41, 52 Cases of inhalational tularemia in the United States are thought to be due mostly to the more virulent F tularensis biovar tularensis (type A) and usually follow an acute and severe course, with prominent pneumonitis. Some cases, however, have radiographic evidence of pleuropneumonia with minimal or absent respiratory signs on physical examination.39, 41, 52

Although airborne F tularensis would be expected to principally cause primary pleuropneumonic infection, some exposures might contaminate the eye, resulting in ocular tularemia; penetrate broken skin, resulting in ulceroglandular or glandular disease; or cause oropharyngeal disease with cervical lymphadenitis. In the aforementioned Swedish outbreak, conjunctivitis was reported in 26% of 140 confirmed cases and an infected ulcer of the skin was reported in nearly 12%; pharyngitis was reported in 31% and oral ulcers in about 9% of the cases; and 32% of these patients had various exanthemas, such as erythema multiforme and erythema nodosum.43 Tularemia outbreaks arising from similar agricultural exposures have been reported from Finland,53 mostly presenting with general constitutional symptoms rather than specific manifestations of pneumonia; enlargement of hilar nodes was the principal radiographic finding in these cases.54

Inhalational Tularemia Following Use as a Biological Weapon

Although F tularensis could be used as a weapon in a number of ways, the working group believes that an aerosol release would have the greatest adverse medical and public health consequences. Release in a densely populated area would be expected to result in an abrupt onset of large numbers of cases of acute, nonspecific febrile illness beginning 3 to 5 days later (incubation range, 1-14 days), with pleuropneumonitis developing in a significant proportion of cases during the ensuing days and weeks. Public health authorities would most likely become aware of an outbreak of unusual respiratory disease in its early stages, but this could be difficult to distinguish from a natural outbreak of community-acquired infection, especially influenza or various atypical pneumonias. The abrupt onset of large numbers of acutely ill persons, the rapid progression in a relatively high proportion of cases from upper respiratory symptoms and bronchitis to life-threatening pleuropneumonitis and systemic infection affecting, among others, young, previously healthy adults and children should, however, quickly alert medical professionals and public health authorities to a critical and unexpected public health event and to bioterrorism as a possible cause (Table 1). Until the etiology became clear, clinicians would need to work closely with epidemiologists and diagnostic laboratories to differentiate the illness from various community-acquired pneumonias and to determine if it could have resulted from use of one of several potential bioterrorism weapons agents, such as those causing tularemia, plague, anthrax, or Q fever.2, 4, 29

In general, tularemia would be expected to have a slower progression of illness and a lower case-fatality rate than either inhalational plague or anthrax. Plague would most likely progress very rapidly to severe pneumonia, with copious watery or purulent sputum production, hemoptysis, respiratory insufficiency, sepsis, and shock.4 Inhalational anthrax would be differentiated by its characteristic radiological findings of prominent symmetric mediastinal widening and absence of bronchopneumonia.2 As well, anthrax patients would be expected to develop fulminating, toxic, and fatal illness despite antibiotic treatment.29 Milder forms of inhalational tularemia could be clinically indistinguishable from Q fever; establishing a diagnosis of either would be problematic without reference laboratory testing. Presumptive laboratory diagnoses of plague or anthrax would be expected to be made relatively quickly, although microbiological confirmation could take days. Isolation and identification of F tularensis using routine laboratory procedures could take several weeks.

Once a substantial cluster of cases of inhalational tularemia had been identified, epidemiological findings should suggest a bioterrorist event. The abrupt onset and single peak of cases would implicate a point-source exposure without secondary transmission. Among exposed persons, attack rates would likely be similar across sex and age groups, and risk would be related to degree of exposure to the point source (Table 1). An outbreak of inhalational tularemia in an urban setting should trigger a high level of suspicion of an intentional event, since all reported inhalational tularemia outbreaks have occurred in rural areas.

MICROBIOLOGY AND VIRULENCE FACTORS


Francisella tularensis is a small, nonmotile, aerobic, gram-negative coccobacillus. It has a thin lipopolysaccharide-containing envelope and is a hardy non–spore-forming organism that survives for weeks at low temperatures in water, moist soil, hay, straw, and decaying animal carcasses.21, 22, 60, 61 Francisella tularensis has been divided into 2 major subspecies (biovars) by virulence testing, biochemical reactions, and epidemiological features.62 Francisella tularensis biovar tularensis (type A) may be highly virulent in humans and animals, produces acid from glycerol, demonstrates citrulline ureidase activity, and is the most common biovar isolated in North America.22, 60 Francisella tularensis biovar palaearctica (type B) is relatively avirulent, does not produce acid from glycerol, and does not demonstrate citrulline ureidase activity. In Europe and Asia, all human tularemia is thought to be caused by the milder type B strains, although recent studies there have identified naturally occurring F tularensis related to F tularensis biovar tularensis.63, 64 A few rapidly growing strains of F tularensis have been recovered from the blood of immunocompromised patients not showing seroreactivity to F tularensis.65

Transformed plasmids have been engineered to express chloramphenicol and tetracycline resistance in F tularensis.66 Virulent, streptomycin-resistant F tularensis strains have been examined in biowarfare agent studies both in the United States and the Soviet Union.24, 27, 56 Although F tularensis virulence factors are poorly understood and characterized,67, 68 it is possible that strain virulence could be enhanced through laboratory manipulation.

PATHOGENESIS AND CLINICAL MANIFESTATIONS


Pathogenesis

Francisella tularensis can infect humans through the skin, mucous membranes, gastrointestinal tract, and lungs. It is a facultative intracellular bacterium that multiplies within macrophages.68, 69 The major target organs are the lymph nodes, lungs and pleura, spleen, liver, and kidney.19, 20, 49, 70-72 Untreated, bacilli inoculated into skin or mucous membranes multiply, spread to the regional lymph nodes and further multiply, and may then disseminate to organs throughout the body. Bacteremia may be common in the early phase of infection. The initial tissue reaction to infection is a focal, intensely suppurative necrosis consisting largely of accumulations of polymorphonuclear leukocytes, followed by invasion of macrophages, epithelioid cells, and lymphocytes. Suppurative lesions become granulomatous, and histopathological examination of the granulomas shows a central necrotic, sometimes caseating zone surrounded by a layer of epithelioid cells, multinucleated giant cells, and fibroblasts in a radial arrangement, typical of other granulomatous conditions, such as tuberculosis and sarcoidosis.20, 70, 71

Monkeys that inhaled the virulent SCHU S-4 strain of F tularensis (type A) developed acute bronchiolitis within 24 hours of exposure to 1-µm particles and within 48 hours of exposure to 8-µm particles.73 By 72 hours following challenge, inflammation was present in peribronchial tissues and alveolar septa. Bronchopneumonia was most pronounced in animals exposed to the smaller particles and was characterized by tracheobronchial lymph node enlargement and reddish, firm, 0.2- to 0.5-cm-diameter discrete inflammatory lesions scattered throughout the lungs. In the absence of treatment, the disease progressed to pneumonic consolidation and organization, granuloma formation, and eventual chronic interstitial fibrosis.

Humans with inhalational exposures also develop hemorrhagic inflammation of the airways early in the course of illness, which may progress to bronchopneumonia.54 Histopathological examination of affected lungs shows alveolar spaces filled with an exudate of mononuclear cells. Pleuritis with adhesions and effusion and hilar lymphadenopathy are common radiological and pathological findings.70, 72

Clinical Manifestations

The primary clinical forms of tularemia vary in severity and presentation according to virulence of the infecting organism, dose, and site of inoculum. Primary disease presentations include ulceroglandular, glandular, oculoglandular, oropharyngeal, pneumonic, typhoidal, and septic forms.19, 20, 49, 70, 72, 74, 75 The term typhoidal tularemia has been used to describe illness in tularemia patients with systemic infections manifesting as fever and other constitutional signs without cutaneous or mucosal membrane lesions or regional lymphadenitis. Sometimes, these patients present with prominent gastrointestinal manifestations, such as diarrhea and pain. Confusion is created when typhoidal tularemia is used to describe the illness in patients infected by inhalation, especially when there are signs of pleuropneumonic disease; this usage can be misleading and has been discouraged.54, 75

The onset of tularemia is usually abrupt, with fever (38°C-40°C), headache, chills and rigors, generalized body aches (often prominent in the low back), coryza, and sore throat. A pulse-temperature dissociation has been noted in as many as 42% of patients.49 A dry or slightly productive cough and substernal pain or tightness frequently occur with or without objective signs of pneumonia, such as purulent sputum, dyspnea, tachypnea, pleuritic pain, or hemoptysis.7, 19, 26, 70, 74 Nausea, vomiting, and diarrhea sometimes occur. Sweats, fever and chills, progressive weakness, malaise, anorexia, and weight loss characterize the continuing illness. Studies of volunteers have shown that F tularensis aerosol exposures can incapacitate some persons in the first 1 or 2 days of illness, and significant impairment in performing tasks can continue for days after antibiotic treatment is begun.76 In untreated tularemia, symptoms often persist for several weeks and, sometimes, for months, usually with progressive debility. Any form of tularemia may be complicated by hematogenous spread, resulting in secondary pleuropneumonia, sepsis, and, rarely, meningitis.74, 77

Prior to the advent of antibiotics, the overall mortality from infections with the more severe type A strains was in the range of 5% to 15%, and fatality rates as high as 30% to 60% were reported for untreated pneumonic and severe systemic forms of disease.72, 78 Currently, the overall case-fatality rate of reported cases in the United States is less than 2%.34, 49 Type B infections are rarely fatal.

In ulceroglandular tularemia, the form that typically arises from handling a contaminated carcass or following an infective arthropod bite, a local cutaneous papule appears at the inoculation site at about the time of onset of generalized symptoms, becomes pustular, and ulcerates within a few days of its first appearance. The ulcer is tender, generally has an indolent character, and may be covered by an eschar. Typically, one or more regional afferent lymph nodes may become enlarged and tender within several days of the appearance of the papule. Even with antibiotic treatment, the affected nodes may become fluctuant and rupture. In oculoglandular tularemia, which follows direct contamination of the eye, ulceration occurs on the conjunctiva, accompanied by pronounced chemosis, vasculitis, and regional lymphadenitis. Glandular tularemia is characterized by lymphadenopathy without an ulcer.

Oropharyngeal tularemia is acquired by drinking contaminated water, ingesting contaminated food, and, sometimes, by inhaling contaminated droplets or aerosols.14, 20, 36, 43, 50, 51, 79 Affected persons may develop stomatitis but more commonly develop exudative pharyngitis or tonsillitis, sometimes with ulceration. Pronounced cervical or retropharyngeal lymphadenopathy may occur (Figure 1).74, 79

Tularemia pneumonia can be the direct result of inhaling contaminated aerosols or be secondary to hematogenous spread from a distal site. An aerosol release of F tularensis would be expected to result in acute illness with signs and symptoms of 1 or more of pharyngitis, bronchiolitis, pleuropneumonitis, and hilar lymphadenitis, accompanied by various manifestations of systemic illness. Inhalational exposures, however, commonly result in an initial clinical picture of systemic illness without prominent signs of respiratory disease.7, 43, 53, 56 The earliest pulmonary radiographic findings of inhalational tularemia may be peribronchial infiltrates, typically advancing to bronchopneumonia in 1 or more lobes, and often accompanied by pleural effusions and hilar lymphadenopathy (Figure 2).72, 75 Signs may, however, be minimal or absent, and some patients will show only 1 or several small, discrete pulmonary infiltrates or scattered granulomatous lesions of lung parenchyma or pleura. Although volunteers challenged with aerosols of virulent F tularensis (type A) regularly developed systemic symptoms of acute illness 3 to 5 days following exposure, only 25% to 50% of participants had radiological evidence of pneumonia in the early stages of infection.7, 26 On the other hand, pulmonary infection can sometimes rapidly progress to severe pneumonia, respiratory failure, and death.72, 80 Lung abscesses occur infrequently.75

Typhoidal tularemia is used to describe systemic illness in the absence of signs indicating either site of inoculation or anatomic localization of infection. This should be differentiated from inhalational tularemia with pleuropneumonic disease.54, 75

Tularemia sepsis is potentially severe and fatal. As in typhoidal tularemia, nonspecific findings of fever, abdominal pain, diarrhea, and vomiting may be prominent early in the course of illness. The patient typically appears toxic and may develop confusion and coma. Unless treated promptly, septic shock and other complications of systemic inflammatory response syndrome may ensue, including disseminated intravascular coagulation and bleeding, acute respiratory distress syndrome, and organ failure.80

DIAGNOSIS


Tularemia in humans occurs infrequently, resulting in a low index of diagnostic suspicion among clinicians and laboratorians. Since rapid diagnostic testing for tularemia is not widely available, the first indication of intentional tularemia might follow recognition by public health authorities of a clustering of acute, severe respiratory illness with unusual epidemiological features (Table 1). Suspicion of tularemia might be triggered in alert clinicians encountering patients with findings of atypical pneumonia, pleuritis, and hilar lymphadenopathy. Identification of F tularensis in clinical specimens may be missed or delayed for days or weeks when procedures for routine microbiological screening of bacterial pathogens are followed, and it is unlikely that a serendipitous laboratory identification would be the sentinel event that alerted authorities to a major bioterrorism action.

Physicians who suspect inhalational tularemia should promptly collect specimens of respiratory secretions and blood and alert the laboratory to the need for special diagnostic and safety procedures. Francisella tularensis may be identified by direct examination of secretions, exudates, or biopsy specimens using direct fluorescent antibody or immunohistochemical stains.81-83 By light microscopy, the organism is characterized by its small size (0.2 µm 0.2-0.7 µm), pleomorphism, and faint staining. It does not show the bipolar staining characteristics of Yersinia pestis,4 the agent of plague, and is easily distinguished from the large gram-positive rods characteristic of vegetative forms of Bacillus anthracis (Figure 3).2 Microscopic demonstration of F tularensis using fluorescent-labeled antibodies is a rapid diagnostic procedure performed in designated reference laboratories in the National Public Health Laboratory Network; test results can be made available within several hours of receiving the appropriate specimens if the laboratory is alerted and prepared. Suspicion of inhalational tularemia must be promptly reported to local or state public health authorities so timely epidemiological and environmental investigations can be made (BOX).

Growth of F tularensis in culture is the definitive means of confirming the diagnosis of tularemia.60, 81 Francisella tularensis can be grown from pharyngeal washings, sputum specimens, and even fasting gastric aspirates in a high proportion of patients with inhalational tularemia.56 It is only occasionally isolated from the blood. Francisella tularensis grows best in cysteine-enriched broth and thioglycollate broth and on cysteine heart blood agar, buffered charcoal-yeast agar, and chocolate agar. Selective agar (such as chocolate agar selective for Neisseria gonorrhea isolation) may be useful when culturing materials from nonsterile sites, such as sputum. Inoculated media should be incubated at 37°C. Although growth may be visible as early as 24 to 48 hours after inoculation, growth may be delayed and cultures should be held for at least 10 days before discarding. Under ideal conditions, bacterial colonies on cysteine-enriched agar are typically 1 mm in diameter after 24 to 48 hours of incubation and 3 to 5 mm in diameter by 96 hours.60, 81 On cysteine heart agar, F tularensis colonies are characteristically opalescent and do not discolor the medium (Figure 4).

Antigen detection assays, polymerase chain reaction, enzyme-linked immunoassays, immunoblotting, pulsed-field gel electrophoresis, and other specialized techniques may be used to identify F tularensis and to characterize strains.84-87 These procedures are usually performed only in research and reference laboratories, however. In laboratories where advanced methods are established, results of antigen detection and polymerase chain reaction analyses can be obtained within several hours of receipt of isolates. Typically, serum antibody titers do not attain diagnostic levels until 10 or more days after onset of illness, and serology would provide minimal useful information for managing an outbreak. Serological confirmation of cases, however, may be of value for forensic or epidemiological purposes. Most laboratories use tube agglutination or microagglutination tests that detect combined immunoglobulin M and immunoglobulin G.84, 85 A 4-fold change in titer between acute and convalescent serum specimens, a single titer of at least 1:160 for tube agglutination or 1:128 for microagglutination is diagnostic for F tularensis infection. Information on reference diagnostic testing and shipping/handling of specimens can be obtained from state public health laboratories and from the Division of Vector-Borne Infectious Diseases, CDC, Fort Collins, Colo (telephone: [970] 221-6400; e-mail: dvbid@cdc.gov).

VACCINATION


Beginning in the 1930s, the Soviet Union used a live attenuated vaccine to immunize tens of millions of persons living in tularemia-endemic areas.88 In the United States, a live attenuated vaccine derived from the avirulent live vaccine strain has been used to protect laboratorians routinely working with F tularensis; until recently, this vaccine was available as an investigational new drug.89 It is currently under review by the US Food and Drug Administration (FDA), and its future availability is undetermined.

In a retrospective study of civilians working with F tularensis at a US Army research facility, the incidence of accidental acute inhalational tularemia among laboratorians declined from 5.70 cases per 1000 person-years of risk at a time when a killed vaccine was in use to 0.27 cases per 1000 person-years of risk after introduction of the live vaccine.58 Although the incidence of ulceroglandular disease remained unchanged in the 2 periods, signs and symptoms were considered milder among those who received the live vaccine. In volunteer studies, the live attenuated vaccine did not protect all recipients against aerosol challenges with virulent F tularensis.7, 26

Correlates of protective immunity appear about 2 weeks following natural infection or vaccination. Given the short incubation period of tularemia and incomplete protection of current vaccines against inhalational tularemia, vaccination is not recommended for postexposure prophylaxis. The working group recommends use of the live vaccine strain only for laboratory personnel routinely working with F tularensis.

TREATMENT


Contained Casualty Situation

Adults
In a contained casualty situation, in which logistics permit individual medical management, the working group recommends parenteral antimicrobial therapy for tularemia (Table 2). Streptomycin is the drug of choice.49, 74, 90, 91 Gentamicin, which is more widely available and may be used intravenously, is an acceptable alternative.49, 74, 90-93 Treatment with aminoglycosides should be continued for 10 days. Tetracyclines and chloramphenicol are also used to treat tularemia49, 74, 90; however, relapses and primary treatment failures occur at a higher rate with these bacteriostatic agents than with aminoglycosides, and they should be given for at least 14 days to reduce chance of relapse.27, 74, 90 Fluoroquinolones, which have intracellular activity, are promising candidates for treating tularemia. Ciprofloxacin, which is not labeled for use in tularemia, has been shown to be active against F tularensis in vitro94 and in animals95 and has been used to successfully treat tularemia in both adults and children.90, 94, 96, 97 Treatment with ciprofloxacin should be continued for 10 days. In persons beginning treatment with parenteral doxycycline, ciprofloxacin, or chloramphenicol, therapy can be switched to oral antibiotic administration when clinically indicated. Very limited experiences in treating tularemia patients with beta-lactam and macrolide antibiotics have been reported, and treatment failures have occurred.98 Use of beta-lactam and macrolide antibiotics in treating tularemia is neither FDA-approved nor recommended by the working group.

Children
In children, streptomycin or gentamicin is recommended by the working group as first-line treatment in a contained casualty situation (Table 2). Doxycycline, ciprofloxacin (1 g/d), and chloramphenicol can be used as alternatives to aminoglycosides. Fluoroquinolones have been reported to cause cartilage damage in immature animals and are not FDA-approved for use in children. However, short courses of these agents have not been associated with arthropathy in pediatric patients, and the potential risks of their use must be weighed against their benefits in treating serious infections.96, 99, 100

Mass Casualty Situation

Doxycycline and ciprofloxacin, administered orally, are the preferred choices for treatment in the mass casualty setting, for both adults and children (Table 3). The ciprofloxacin dosage for children should not exceed 1 g/d. In a mass casualty situation, the working group believes the benefits to children from short courses of doxycycline or fluoroquinolones (Table 3) outweigh the risks of their use.

Since it is unknown whether drug-resistant organisms might be used in a bioterrorist event, antimicrobial susceptibility testing of isolates should be conducted quickly and treatments altered according to test results and clinical responses.

Antibiotics for treating patients infected with tularemia in a bioterrorism scenario are included in a national pharmaceutical stockpile maintained by the CDC, as are ventilators and other emergency equipment needed to respond to situations of large numbers of critically ill persons that strip local and state resources.30

Management of Special Groups

Pregnant Women
In a contained casualty situation, short courses of gentamicin are likely to pose a low risk to fetuses when used to treat tularemia in pregnant women (Table 2). Rare cases of fetal nerve deafness and renal damage have been reported with other aminoglycosides but have not been reported with gentamicin. The benefits of gentamicin in treating pregnant women with tularemia are expected to outweigh any potential risk to fetuses. In a mass casualty situation, oral ciprofloxacin is considered the best alternative to gentamicin for pregnant women (Table 3).

Immunosuppressed Persons
There is scant experience in treating tularemia in immunocompromised patients. However, considering the greater occurrence in immunocompetent patients of tularemia relapses and treatment failures following use of bacteriostatic antimicrobial agents compared with aminoglycosides, streptomycin or gentamicin should be used when possible to treat patients with known immune dysfunction in either contained casualty or mass casualty situations (Table 2).

POSTEXPOSURE ANTIBIOTIC RECOMMENDATIONS


Persons beginning treatment with streptomycin, gentamicin, doxycycline, or ciprofloxacin in the incubation period of tularemia and continuing treatment daily for 14 days might be protected against symptomatic infection. In studies of aerosol challenge with infective doses of the virulent SCHU S-4 strain of F tularensis, each of 8 volunteers given oral dosages of tetracycline, 1 g/d for 28 days, and each of 8 volunteers given tetracycline, 2 g/d for 14 days, were fully protected when treatment was begun 24 hours following challenge.27 Two of 10 volunteers given tetracycline, 1 g/d for only 5 days, developed symptomatic tularemia after antibiotic treatment was stopped.

In the unlikely event that authorities quickly become aware that an F tularensis biological weapon has been used and are able to identify and reach exposed persons during the early incubation period, the working group recommends that exposed persons be prophylactically treated with 14 days of oral doxycycline or ciprofloxacin (Table 3). In a circumstance in which the weapon attack has been covert and the event is discovered only after persons start to become ill, persons potentially exposed should be instructed to begin a fever watch. Persons who develop an otherwise unexplained fever or flulike illness within 14 days of presumed exposure should begin treatment as outlined in Table 2 and Table 3.

In the laboratory, persons who have had potentially infective exposures to F tularensis should be administered oral postexposure antibiotic prophylaxis if the risk of infection is high (eg, spill, centrifuge accident, or needlestick). If the risk is low, exposed persons can be placed on a fever watch and treated if they develop symptoms.

Postexposure prophylactic antibiotic treatment of close contacts of tularemia patients is not recommended since human-to-human transmission of F tularensis is not known to occur.

INFECTION CONTROL


Isolation is not recommended for tularemia patients, given the lack of human-to-human transmission. In hospitals, standard precautions101 are recommended by the working group for treatment of patients with tularemia.

Microbiology laboratory personnel should be alerted when tularemia is clinically suspected. Routine diagnostic procedures can be performed in biological safety level 2 (BSL-2) conditions. Examination of cultures in which F tularensis is suspected should be carried out in a biological safety cabinet. Manipulation of cultures and other activities involving infectious materials with a potential for aerosol or droplet production (centrifuging, grinding, vigorous shaking, growing cultures in volume, animal studies) require BSL-3 conditions.102 When F tularensis is presumptively identified in a routine BSL-2 clinical laboratory (level A), specimens should be forwarded to a BSL-3 laboratory (level B) (eg, a state public health laboratory) for confirmation of agent and other studies, such as antimicrobial susceptibility testing.11 Bodies of patients who die of tularemia should be handled using standard precautions. Autopsy procedures likely to cause aerosols, such as bone sawing, should be avoided. Clothing or linens contaminated with body fluids of patients infected with F tularensis should be disinfected per standard precautions protocols.101

ENVIRONMENTAL DECONTAMINATION AND PROTECTION


Under natural conditions, F tularensis may survive for extended periods in a cold, moist environment. The working group lacks information on survival of intentionally dispersed particles but would expect a short half-life due to desiccation, solar radiation, oxidation and other environmental factors, and a very limited risk from secondary dispersal. In circumstances of a laboratory spill or intentional use in which authorities are concerned about an environmental risk (eg, inanimate surfaces wet with material thought to contain F tularensis), decontamination can be achieved by spraying the suspected contaminant with a 10% bleach solution (1 part household bleach and 9 parts water). After 10 minutes, a 70% solution of alcohol can be used to further clean the area and reduce the corrosive action of the bleach. Soap water can be used to flush away less hazardous contaminations. Persons with direct exposure to powder or liquid aerosols containing F tularensis should wash body surfaces and clothing with soap water. Standard levels of chlorine in municipal water sources should protect against waterborne infection.60 Following an urban release, the risk to humans of acquiring tularemia from infected animals or arthropod bites is considered minimal and could be reduced by educating the public on simple avoidance of sick or dead animals and on personal protective measures against biting arthropods.

ADDITIONAL RESEARCH


Simple, rapid, and reliable diagnostic tests that could be used to identify persons infected with F tularensis in the mass exposure setting need to be developed. Further methods should be designed to rapidly define the molecular genetic characteristics of organisms, especially as they may relate to engineered attributes, such as enhanced virulence and resistance to antimicrobial agents or normally lethal environmental conditions. Complete sequencing and analysis of the genome of natural strains of F tularensis would provide an archival base for understanding genetic variants, functions of genes, and mechanisms of action useful in developing means to protect against F tularensis. Research is also needed to develop accurate and reliable procedures to rapidly detect F tularensis in environmental samples.

New technologies should be explored for developing active (eg, DNA-based) or passive (eg, monoclonal antibody–based) vaccines for rapid preexposure or postexposure protection.


Author/Article Information


Author Affiliations: National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Ga (Drs Dennis, Lillibridge, and McDade); Center for Civilian Biodefense Studies, Johns Hopkins University Schools of Medicine (Drs Inglesby, Bartlett, and Perl) and Public Health (Drs Henderson, O'Toole, and Russell), Baltimore, Md; Viral and Rickettsial Diseases Laboratory, California Department of Health Services, Berkeley (Dr Ascher); US Army Medical Research Institute of Infectious Diseases, Ft Detrick, Md (Drs Eitzen, Friedlander, and Parker); Bureau of Communicable Disease, New York City Health Department (Drs Fine and Layton), and Kroll Associates (Mr Hauer), New York, NY; ican Inc, Eden Prairie, Minn (Dr Osterholm); and Office of Emergency Preparedness, Department of Health and Human Services, Rockville, Md (Dr Tonat).

Corresponding Author and Reprints: David T. Dennis, MD, MPH, Division of Vector-Borne Infectious Diseases, National Center for Infectious Diseases, Centers for Disease Control and Prevention, PO Box 2087, Fort Collins, CO 80522 (e-mail: dtd1@cdc.gov).

Box. Clinicians Caring for Patients With Suspected Tularemia Should Immediately Contact Their:

(1) Hospital epidemiologist or infection control practitioner and

(2) Local or state health departments

Consult your local telephone operator, the telephone directory under "governmental listings," or the Internet at http://www.cdc.gov/other.htm#states or http://www.astho.org/state.html

If the local and state health departments are unavailable, contact the Centers for Disease Control and Prevention at (970) 221-6400 or http://www.cdc.gov/ncidod/dvbid/dvbid.htm

Return to text.



Ex Officio Participants in the Working Group on Civilian Biodefense: George Counts, MD, CDC; Margaret Hamburg, MD, former assistant secretary for planning and evaluation, Department of Health and Human Services (DHHS); Robert Knouss, MD, Office of Emergency Preparedness, DHHS; Brian Malkin, Esq, formerly with the FDA; and Stuart Nightingale, MD, Office of the Assistant Secretary for Planning and Evaluation, DHHS.

Funding/Support: Funding for this study primarily was provided by each participant's institution or agency. The Johns Hopkins Center for Civilian Biodefense Studies provided travel funds for 5 of the group.

Disclaimers: In some instances, the indications, dosages, and other information in this article are not consistent with current approved labeling by the US Food and Drug Administration (FDA). The recommendations on use of drugs and vaccine for uses not approved by the FDA do not represent the official views of the FDA nor of any of the federal agencies whose scientists participated in these discussions. Unlabeled uses of the products recommended are noted in the sections of this article in which these products are discussed. Where unlabeled uses are indicated, information used as the basis for the recommendation is discussed.

The views, opinions, assertions, and findings contained herein are those of the authors and should not be construed as official US Department of Defense or US Department of Army positions, policies, or decisions unless so designated by other documentation.

Additional Articles: This article is the fifth in a series entitled Medical and Public Health Management Following the Use of a Biological Weapon: Consensus Statements of the Working Group on Civilian Biodefense. See references 2 through 5.

Acknowledgment: We thank May C. Chu, PhD, CDC, for assistance with laboratory diagnostic aspects of tularemia, and Edward B. Hayes, MD, CDC, for assistance with clinical and epidemiological aspects of tularemia.


REFERENCES


1.
Parker RR.
Recent studies of tick-borne diseases made at the United States Public Health Service Laboratory at Hamilton, Montana.
In: Proceedings of the Fifth Pacific Congress; 1934:3367-3374.

2.
Inglesby TV, Henderson DA, Bartlett JG, et al, for the Working Group on Civilian Biodefense.
Anthrax as a biological weapon: medical and public health management.
JAMA.
1999;281:1735-1745.
ABSTRACT  |  FULL TEXT  |  PDF  |  MEDLINE

3.
Henderson DA, Inglesby TV, Bartlett JG, et al, for the Working Group on Civilain Biodefense.
Smallpox as a biological weapon: medical and public health management.
JAMA.
1999;281:2127-2137.
ABSTRACT  |  FULL TEXT  |  PDF  |  MEDLINE

4.
Inglesby TV, Dennis DT, Henderson DA, et al, for the Working Group on Civilian Biodefense.
Plague as a biological weapon: medical and public health management.
JAMA.
2000;283:2281-2290.
ABSTRACT  |  FULL TEXT  |  PDF  |  MEDLINE

5.
Arnon SA, Schecter R, Inglesby TV, et al, for the Working Group on Civilain Biodefense.
Botulinum toxin as a biological weapon: medical and public health management.
JAMA.
2001;285:1059-1070.
ABSTRACT  |  FULL TEXT  |  PDF  |  MEDLINE

6.
Saslaw S, Eigelsbach HT, Wilson HE, Prior JA, Carhart S.
Tularemia vaccine study, I: intracutaneous challenge.
Arch Intern Med.
1961;107:121-133.

7.
Saslaw S, Eigelsbach HT, Prior JA, Wilson HE, Carhart S.
Tularemia vaccine study, II: respiratory challenge.
Arch Intern Med.
1961;107:134-146.

8.
World Health Organization.
Health Aspects of Chemical and Biological Weapons.
Geneva, Switzerland: World Health Organization; 1970:75-76.

9.
Kaufmann AF, Meltzer MI, Schmid GP.
The economic impact of a bioterrorist attack: are prevention and post-attack intervention programs justifiable?
Emerg Infect Dis.
1997;3:83-94.
MEDLINE

10.
Christopher GW, Cieslak TJ, Pavlin JA, Eitzen EM.
Biological warfare: a historical perspective.
JAMA.
1997;278:412-417.
MEDLINE

11.
Centers for Disease Control and Prevention.
Biological and chemical terrorism: strategic plan for preparedness and response: recommendations of the CDC Strategic Planning Workgroup.
MMWR Morb Mortal Wkly Rep.
2000;49(RR-4):1-14.

12.
Francis E.
Tularemia.
JAMA.
1925;84:1243-1250.

13.
Karpoff SP, Antononoff NI.
The spread of tularemia through water as a new factor in its epidemiology.
J Bacteriol.
1936;32:243-258.

14.
Silchenko VS.
Epidemiological and clinical features of tularemia caused by waterborne infection.
Zh Mikrobiol Epidemiol Immunobiol.
1957;28:788-795.

15.
Gelman AC.
The ecology of tularemia.
In: May JM, ed. Studies in Disease Ecology. New York, NY: Hafner Publishing Co; 1961:89-108.

16.
Jellison WL, Kohls GM.
Tularemia in Sheep and Sheep Industry Workers in Western United States.
Washington, DC: US Public Health Service; 1955:1-17. Public health monograph 28.

17.
Francis E.
Sources of infection and seasonal incidence of tularemia in man.
Public Health Rep.
1937;52:103-113.

18.
Lake GC, Francis E.
Six cases of tularemia occurring in laboratory workers.
Public Health Rep.
1922;37:392-413.

19.
Simpson WM.
Tularemia (Francis' disease).
Ann Intern Med.
1928;1:1007-1059.

20.
Francis E.
A summary of present knowledge of tularemia.
Medicine.
1928;7:411-432.

21.
Hopla CE.
The ecology of tularemia.
Adv Vet Sci Comp Med.
1974;18:25-53.
MEDLINE

22.
Jellison WL.
Tularemia in North America.
Missoula: University of Montana; 1974:1-276.

23.
Harris S.
Japanese biological warfare research on humans: a case study of microbiology and ethics.
Ann N Y Acad Sci.
1992;666:21-52.
MEDLINE

24.
Alibek K.
Biohazard.
New York, NY: Random House; 1999:29-38.

25.
McCrumb FR Jr, Snyder MJ, Woodward TE.
Studies on human infection with Pasteurella tularensis: comparison of streptomycin and chloramphenicol in the prophylaxis of clinical disease.
Trans Assoc Am Physicians.
1957;70:74-80.

26.
McCrumb FR Jr.
Aerosol infection in man with Pasteurella tularensis.
Bacteriol Rev.
1961;25:262-267.

27.
Sawyer WD, Dangerfield HG, Hogge AL, Crozier D.
Antibiotic prophylaxis and therapy of airborne tularemia.
Bacteriol Rev.
1966;30:542-548.
MEDLINE

28.
Health Aspects of Chemical and Biological Weapons.
Geneva, Switzerland: World Health Organization; 1970:105-107.

29.
Franz DR, Jahrling PB, Friedlander AM, et al.
Clinical recognition and management of patients exposed to biological warfare agents.
JAMA.
1997;278:399-411.
MEDLINE

30.
Khan AS, Morse S, Lillibridge S.
Public health preparedness for biological terrorism in the USA.
Lancet.
2000;356:1179-1182.
MEDLINE

31.
Tucker JB.
National health and medical services response to incidents of chemical and biological terrorism.
JAMA.
1997;278:362-368.
MEDLINE

32.
Hopla CE, Hopla AK.
Tularemia.
In: Beran GW, Steele JH, eds. Handbook of Zoonoses. 2nd ed. Boca Raton, Fla: CRC Press; 1994:113-126.

33.
Centers for Disease Control and Prevention.
Summary of notifiable diseases, United States, 1997.
MMWR Morb Mortal Wkly Rep.
1998;46:ii-vii, 3-87.

34.
Dennis DT.
Tularemia.
In: Wallace RB, ed. Maxcy-Rosenau-Last Public Health and Preventive Medicine. 14th ed. Stamford, Conn: Appleton & Lange; 1998:354-357.

35.
Boyce JM.
Recent trends in the epidemiology of tularemia in the United States.
J Infect Dis.
1975;131:197-199.
MEDLINE

36.
Tärnvik A, Sandström G, Sjöstedt A.
Epidemiological analysis of tularemia in Sweden 1931-1993.
FEMS Immunol Med Microbiol.
1996;13:201-204.
MEDLINE

37.
Pollitzer R.
History and Incidence of Tularemia in the Soviet Union: A Review.
Bronx, NY: Institute for Contemporary Russian Studies, Fordham University; 1967:1-103.

38.
Halsted CC, Klasinghe HP.
Tularemia pneumonia in urban children.
Pediatrics.
1978;61:660-662.
MEDLINE

39.
Martone WJ, Marshall LW, Kaufmann AF, Hobbs JH, Levy ME.
Tularemia pneumonia in Washington, DC. A report of three cases with possible common-source exposures.
JAMA.
1979;242:2315-2317.
MEDLINE

40.
Rogutsky SV, Khramtsov MM, Avchinikov AV, et al.
Epidemiological investigation of an outbreak of tularemia in the Smolensk region.
Zh Mikrobiol (Moscow).
1997;2:33-37.

41.
McCarthy VP, Murphy MD.
Lawnmower tularemia.
Pediatr Infect Dis J.
1990;9:298-299.
MEDLINE

42.
Klock LE, Olsen PF, Fukushima T.
Tularemia epidemic associated with the deerfly.
JAMA.
1973;226:149-152.
MEDLINE

43.
Dahlstrand S, Ringertz O, Zetterberg.
Airborne tularemia in Sweden.
Scand J Infect Dis.
1971;3:7-16.
MEDLINE

44.
Christenson B.
An outbreak of tularemia in the northern part of central Sweden.
Scand J Infect Dis.
1984;16:285-290.
MEDLINE

45.
Warring WB, Ruffin JS.
A tick-borne epidemic of tularemia.
N Engl J Med.
1946;234:137-140.

46.
Ohara Y, Sato T, Homma M.
Arthropod-borne tularemia in Japan: clinical analysis of 1,374 cases observed between 1924 and 1996.
J Med Entomol.
1998;35:471-473.
MEDLINE

47.
Markowitz LE, Hynes NA, de la Cruz P, et al.
Tick-borne tularemia: an outbreak of lymphadenopathy in children.
JAMA.
1985;254:2922-2925.
MEDLINE

48.
Young LS, Bicknell DS, Archer BG, et al.
Tularemia epidemic, Vermont, 1968: forty-seven cases linked to contact with muskrats.
N Engl J Med.
1969;280:1253-1260.
MEDLINE

49.
Evans ME, Gregory DW, Schaffner W, McGee ZA.
Tularemia: a 30-year experience with 88 cases.
Medicine (Baltimore).
1985;64:251-269.
MEDLINE

50.
Jellison WL, Epler DC, Kuhns E, Kohls GM.
Tularemia in man from a domestic rural water supply.
Public Health Rep.
1950;65:1219-1226.

51.
Mignani E, Palmieri F, Fontana M, Marigo S.
Italian epidemic of waterborne tularaemia.
Lancet.
1988;2:1423.
MEDLINE

52.
Teutsch SM, Martone WJ, Brink EW, et al.
Pneumonic tularemia on Martha's Vineyard.
N Engl J Med.
1979;301:826-828.
MEDLINE

53.
Syrjälä H, Kujala P, Myllylä V, Salminen A.
Airborne transmission of tularemia in farmers.
Scand J Infect Dis.
1985;17:371-375.
MEDLINE

54.
Syrjälä H, Sutinen S, Jokinen K, Nieminen P, Tuuponen T, Salminen A.
Bronchial changes in airborne tularemia.
J Laryngol Otol.
1986;100:1169-1176.
MEDLINE

55.
Puntigam F.
Erkrakungen an torakalen formen der tularämia bei arbeitnehmern in Zuckerfabriken.
Z Hyg.
1960;147:162-168.

56.
Overholt EL, Tigertt WD, Kadull PJ, et al.
An analysis of forty-two cases of laboratory-acquired tularemia.
Am J Med.
1961;30:785-806.

57.
Pike RM.
Laboratory-associated infections: summary and analysis of 3921 cases.
Health Lab Sci.
1976;13:105-114.
MEDLINE

58.
Burke DS.
Immunization against tularemia: analysis of the effectiveness of live Francisella tularensis vaccine in prevention of laboratory-acquired tularemia.
J Infect Dis.
1977;135:55-60.
MEDLINE

59.
Centers for Disease Control and Prevention.
Summary of notifiable diseases, United States, 1994.
MMWR Morb Mortal Wkly Rep.
1994;43:1-80.

60.
Bell JF.
Tularemia.
In: Steele JH, ed. CRC Handbook Series in Zoonoses. Vol 2. Boca Raton, Fla: CRC Press; 1980:161-193.

61.
Pomanskaia LA.
The survival times of the organisms of tularaemia on grain and straw.
J Microbiol Epidemiol Immunobiol.
1957;28:597-603.

62.
Wong JD, Shapiro DS.
Francisella.
In : Murray PR, ed. Manual of Clinical Microbiology. 7th ed. Washington, DC: ASM Press; 1999:647-651.

63.
Johansson A, Ibrahim A, Goransson I, et al.
Evaluation of PCR-based methods for discrimination of Francisella species and subspecies and development of a specific PCR that distinguishes the two major subspecies of Francisella tularensis.
J Clin Microbiol.
2000;38:4180-4185.
MEDLINE

64.
Gurycova D.
First isolation of Francisella tularensis subspecies tularensis in Europe.
Eur J Epidemiol.
1998;14:797-802.
MEDLINE

65.
Clarridge JE III, Raich TJ, Sjösted A, et al.
Characterization of two unusual clinically significant Francisella strains.
J Clin Microbiol.
1996;34:1995-2000.
MEDLINE

66.
Pavlov VM, Mokrievich, Volkovoy K.
Cryptic plasmid pFNL10 from Francisella novicida-like F6168: the base of plasmid vectors for Francisella tularensis.
FEMS Immunol Med Microbiol.
1996;13:253-256.
MEDLINE

67.
Sandström G, Sjöstedt A, Johansson T, Kuoppa K, Williams JC.
Immunogenicity and toxicity of lipopolysaccharide from Francisella tularensis LVS.
FEMS Microbiol Immunol.
1992;5:201-210.
MEDLINE

68.
Tärnvik A.
Nature of protective immunity to Francisella tularensis.
Rev Infect Dis.
1989;11:440-451.
MEDLINE

69.
Fortier AH, Green SJ, Polsinelli T, et al.
Life and death of an intracellular pathogen: Francisella tularensis and the macrophage.
Immunol Ser.
1994;60:349-361.
MEDLINE

70.
Pullen RL, Stuart BM.
Tularemia: analysis of 225 cases.
JAMA.
1945;129:495-500.

71.
Lillie RD, Francis EI.
The pathology of tularaemia in man (Homo sapiens).
In: The Pathology of Tularaemia. Washington, DC: US Government Printing Office; 1937:1-81. National Institute of Health Bulletin No. 167.

72.
Stuart BM, Pullen RL.
Tularemic pneumonia: Review of American literature and report of 15 additional cases.
Am J Med Sci.
1945;210:223-236.

73.
White JD, Rooney JR, Prickett PA, Derrenbacher EH, Beard CW, Griffith WR.
Pathogenesis of experimental respiratory tularemia in monkeys.
J Infect Dis.
1964;114:277-283.

74.
Cross JT, Penn RL.
Francisella tularensis (tularemia).
In: Mandell GL, et al. eds. Principles and Practice of Infectious Diseases. Philadelphia, Pa: Churchill Livingstone; 2000:2393-2402.

75.
Avery FW, Barnett TB.
Pulmonary tularemia: a report of five cases and consideration of pathogenesis and terminology.
Am Rev Respir Dis.
1967;95:584-591.
MEDLINE

76.
Alluisi EA, Beisel WR, Bartonelli PJ, Coates GD.
Behavioral effects of tularaemia and sandfly fever in man.
J Infect Dis.
1973;128:710-717.
MEDLINE

77.
Stuart BM, Pullen RL.
Tularemic meningitis: review of the literature and report of a case with postmortem observations.
Arch Intern Med.
1945;76:163-166.

78.
American Public Health Association.
Tularemia.
In: Chin J, ed. Control of Communicable Diseases Manual. Washington, DC: American Public Health Association; 2000:532-535.

79.
Amoss HL, Sprunt DH.
Tularemia: review of literature of cases contracted by ingestion of rabbit and the report of additional cases with a necropsy.
JAMA.
1936;106:1078-1080.

80.
Sunderrajan EV, Hutton J, Marienfeld D.
Adult respiratory distress syndrome secondary to tularemia pneumonia.
Arch Intern Med.
1985;145:1435-1437.
MEDLINE

81.
Centers for Disease Control and Prevention.
Basic laboratory protocols for the presumptive identification of Francisella tularensis.
Available at: http://www.bt.cdc.gov/Agent/Tularemia/tularemia20010417.pdf. Accessed April 20, 2001.

82.
White JD, McGavran MH.
Identification of Pasteurella tularensis by immunofluorescence.
JAMA.
1965;194:294-296.
MEDLINE

83.
Guarner J, Greer PW, Bartlett J, Chu MC, Shieh WJ, Zaki SR.
Immunohistochemical detection of Francisella tularensis in formalin-fixed paraffin-embedded tissue.
Appl Immunohistochem Mol Morphol.
1999;7:122-126.

84.
Syrjälä H, Koskela P, Ripatti T, Salminen A, Herva E.
Agglutination and ELISA methods in the diagnosis of tularemia in different clinical forms and severities of the disease.
J Infect Dis.
1986;153:142-145.
MEDLINE

85.
Bevanger L, Macland JA, Naess AI.
Agglutinins and antibodies to Francisella tularensis outer membrane antigens in the early diagnosis of disease during an outbreak of tularemia.
J Clin Microbiol.
1988;26:433-437.
MEDLINE

86.
Grunow R, Splettstoesser W, McDonald S, et al.
Detection of Francisella tularensis in biological specimens using a capture enzyme-linked immunosorbent assay, an immunochromatographic handheld assay, and a PCR.
Clin Diagn Lab Immunol.
2000;7:86-90.
MEDLINE

87.
Higgins JA, Hubalek Z, Halouzka J, et al.
Detection of Francisella tularensis in infected mammals and vectors using a proble-based polymerase chain reaction.
Am J Trop Med Hyg.
2000;62:310-318.
MEDLINE

88.
Sjöstedt A, Tärnvik A, Sandström G.
Francisella tularensis: host-parasite interaction.
FEMS Immunol Med Microbiol.
1996;13:181-184.
MEDLINE

89.
French GR, Plotkin SA.
Miscellaneous limited-use vaccines.
In: Plotkin S, Mortimer EA, eds. Vaccine. Philadelphia, Pa: WB Saunders; 1999:728-733.

90.
Enderlin G, Morales L, Jacobs RF, Cross TJ.
Streptomycin and alternative agents for the treatment of tularemia: review of the literature.
Clin Infect Dis.
1994;19:42-47.
MEDLINE

91.
Jacobs RF, Narain JP.
Tularemia in children.
Pediatr Infect Dis.
1983;2:487-491.
MEDLINE

92.
Mason WL, Eigelsbach HT, Little SF, et al.
Treatment of tularemia, including pulmonary tularemia, with gentamicin.
Am Rev Respir Dis.
1980;121:39-45.
MEDLINE

93.
Cross JT, Schutze GE, Jacobs RF.
Treatment of tularemia with gentamicin in pediatric patients.
Pediatr Infect Dis J.
1995;14:151-152.
MEDLINE

94.
Syrjälä H, Schildt R, Räisäinen S.
In vitro susceptibility of Francisella tularensis to fluoroquinolones and treatment of tularemia with norfloxacin and ciprofloxacin.
Eur J Clin Microbiol Infect Dis.
1991;10:68-70.
MEDLINE

95.
Russell P, Eley SM, Fulop MJ, Bell DL, Titball RW.
The efficacy of ciprofloxacin and doxycycline against tularemia.
J Antimicrob Chemother.
1998;41:461-465.
MEDLINE

96.
Limaye AP, Hooper CJ.
Treatment of tularemia with fluoroquinolones: two cases and review.
Clin Infect Dis.
1999;29:922-924.
MEDLINE

97.
Johansson A, Berglund L, Gothefors L, et al.
Ciprofloxacin for treatment of tularemia in children.
Pediatr Infect Dis J.
2000;19:449-453.
MEDLINE

98.
Cross JT, Jacobs RF.
Tularemia: treatment failures with outpatient use of ceftriaxone.
Clin Infect Dis.
1993;17:976-980.
MEDLINE

99.
Quinolones.
In: AHFS Drug Information 1999. Bethesda, Md: American Society of Health-System Pharmacists; 1999:670-684.

100.
American Academy of Pediatrics.
Antimicrobials and related therapy.
In: Peter G, ed. Red Book 2000: Report of the Committee on Infectious Diseases. 25th ed. Elk Grove Village, Ill: American Academy of Pediatrics; 2000:645-646.

101.
Garner JS.
Guideline for isolation precautions in hospitals.
Infect Control Hosp Epidemiol.
1996;17:53-80.
MEDLINE

102.
US Department of Health and Human Services.
Laboratory biosafety level criteria.
In: Richmond JY, McKinney RW, eds. Biosafety in Microbiological and Biomedical Laboratories. 4th ed. Washington, DC: Dept of Health and Human Services; 1999:17-52.






to Top





© 2001 American Medical Association. All rights reserved.

AMA HomeJAMA Info Centers Home Short Cut

10 posted on 10/08/2001 4:39:41 AM PDT by vannrox
[ Post Reply | Private Reply | To 1 | View Replies]

To: vannrox
Thanks to Pure Life Institute for this information. www.purelife.cc
11 posted on 10/08/2001 8:51:41 AM PDT by samantha06
[ Post Reply | Private Reply | To 10 | View Replies]

Disclaimer: Opinions posted on Free Republic are those of the individual posters and do not necessarily represent the opinion of Free Republic or its management. All materials posted herein are protected by copyright law and the exemption for fair use of copyrighted works.

Free Republic
Browse · Search
News/Activism
Topics · Post Article

FreeRepublic, LLC, PO BOX 9771, FRESNO, CA 93794
FreeRepublic.com is powered by software copyright 2000-2008 John Robinson