S.F. Dept. Public Health – Infectious Disease Emergencies July 2008 Page 1/1
Letter from Drs. Katz & Fernyak
How to Use This Guide
Contact Information
Roles and Responsibilities
Tab 1
What Happens When You Report a Disease
Clinician Roles in an Emergency
What to Report
Tab 2
Reportable Diseases and Conditions
Unusual Conditions to Report
Preparing for Emergencies
Tab 3
Preparing for Infectious Disease Emergencies
Clinic/Office Disaster and Emergency Planning
Personal Office Disaster Kit
High Priority Diseases
Tab 4
Anthrax
Introduction, Epidemiology, Clinical Features,
Differential Diagnosis, Laboratory Diagnosis,
Treatment and Prophylaxis, Complications and
Admission Criteria, Infection Control, References.
Avian Influenza
Agent, Epidemiology, Clinical Features, Surveillance
and Diagnosis, Treatment and Prophylaxis, Infection
Control, References.
Botulism
Introduction, Epidemiology, Clinical Features,
Differential Diagnosis, Laboratory Diagnosis,
Treatment and Prophylaxis, Complications and
Admission Criteria, Infection Control, References.
Brucellosis
Agent, Epidemiology, Clinical Features, Differential
Diagnosis, Laboratory Diagnosis, Treatment and
Prophylaxis, Infection Control, References.
Plague
Introduction, Epidemiology, Clinical Features,
Differential Diagnosis, Laboratory Diagnosis,
Treatment and Prophylaxis, Complications and
Admission Criteria, Infection Control, References.
Smallpox
Introduction, Epidemiology, Clinical Features,
Differential Diagnosis, Laboratory Diagnosis,
Treatment and Prophylaxis, Complications and
Admission Criteria, Infection Control, References.
Tularemia
Introduction, Epidemiology, Clinical Features,
Differential Diagnosis, Laboratory Diagnosis,
Treatment and Prophylaxis, Complications and
Admission Criteria, Infection Control, References.
Viral Hemorrhagic Fevers
Introduction, Epidemiology, Clinical Features,
Differential Diagnosis, Laboratory Diagnosis,
Treatment and Prophylaxis, Complications and
Admission Criteria, Infection Control, References.
Infection Control Precautions
Tab 5
Appendix
Tab 6
HIPAA Regulations
Title 17 Regulations
HAND Request Form
Laboratory Report Form
CMR Form
Posters
Bioterrorism Syndromes Chart
CDC Evaluating Patients for Smallpox
TABLE OF CONTENTS
City and County of San Francisco
San Francisco Department of Public Health
Communicable Disease Control & Prevention
101 Grove Street, Suite 204
San Francisco, CA 94102
Tel: (415) 554-2818
Fax: (415) 554-2854
www.sfcdcp.org
Gavin Newsom
Mayor
July 30, 2008
Dear San Francisco Clinician,
Monitoring and controlling infectious disease outbreaks is a priority for the San Francisco Department of
Public Health (SFDPH). Routine infectious disease emergencies vary in scope from a single case of
meningococcal meningitis, to a case of hepatitis A in a food handler, to an outbreak of influenza in a
nursing home. In San Francisco, prompt clinician reports of relevant clinical cases enable us to investigate
and begin disease control activities as soon as possible.
To improve control of relatively common outbreaks and to improve recognition and response to emerging
infectious diseases or diseases possibly related to bioterrorism, the Sentinel Event Enhanced Passive
Surveillance (SEEPS) Project developed the enclosed reference: Infectious Disease Emergencies: A
Preparedness and Response Guide for San Francisco Clinicians. This reference is provided in both hard
copy binder format and electronically on our website (www.sfcdcp.org
).
Throughout the US, clinicians have been the first to confront and alert public health officials to emerging
infections, bioterrorism attacks, and widespread outbreaks. Please familiarize yourself with the contents
of this guide to learn:
1. How SFDPH works to contain infectious diseases
2. Your role in reporting infectious diseases
3. Which diseases you should report to the health department
4. Your role in an infectious disease emergency
5. How to prepare for infectious disease emergencies
6. How to recognize and respond to potential bioterrorism agents and emerging
infectious diseases
Currently, SFDPH alerts and updates clinicians on important public health infectious disease threats via
fax using our Health Alert Notification Database (HAND) and by posting materials on our website.
Sign up for inclusion in our HAND if you are not yet registered or if you would like to
update your contact information. See instructions in the guide.
Visit and bookmark our
website (www.sfcdcp.org ).
Working together, clinicians and SFDPH can better protect the health of all San Franciscans.
Thank you for your efforts.
Sincerely,
Mitch Katz, MD Susan Fernyak, MD, MPH
Director Director
San Francisco Dept. of Public Health Communicable Disease Control & Prevention Section
S.F. Dept. Public Health – Infectious Disease Emergencies July 2008 Page 1/1
This reference guide seeks to provide a comprehensive resource to assist clinicians in preparing for,
recognizing, and responding to infectious disease emergencies. In the event of an infectious disease
emergency, the San Francisco Department of Public Health will supplement the information in this
guidebook with updates that will be faxed to clinicians and posted at: www.sfcdcp.org/healthalerts
.
Use of this guide by San Francisco clinicians will strengthen surveillance for, and reporting of, certain
critically important infectious diseases. Throughout the US, clinicians have been the first to confront and
alert public health officials to emerging infections, bioterrorism attacks, and widespread outbreaks. In
San Francisco the Department of Public Health relies daily on clinicians to recognize, respond to, and
report infectious diseases. In an emerging infectious disease outbreak or bioterrorist attack these duties
are especially critical.
This reference guide is divided into the following sections:
Tab 1 Roles & Responsibilities
Roles and responsibilities of the Department of Public Health and
San Francisco clinicians in an emergency
Tab 2 What to Report
List of legally reportable diseases and contact information
List of unusual conditions to report
Tab 3 Preparing for Infectious Disease Emergencies
Guidance for preparing family, home, and office for an infectious
disease emergency
Tab 4 High Priority Diseases
Detailed information on potential bioterrorism related diseases and
emerging infectious diseases
Tab 5 Infection Control Guidelines
Description of standard, contact, droplet, and airborne precautions
Tab 6 Appendix
Useful reporting forms and important regulations
An electronic copy of this guide is available at the SFDPH Communicable Disease Control & Prevention
website:
www.sfcdcp.org/publications
The guide is modular and updates will be made as new information becomes available. To access the
latest information visit the SFDPH Communicable Disease Control & Prevention website and review or
download the most up-to-date materials.
HOW TO USE THIS GUIDE
S.F. Dept. Public Health – Infectious Disease Emergencies July 2008 Page 1/ 1
Communicable Disease Control & Prevention
DISEASE REPORTING
Communicable Disease Reporting
(except TB, STDs, HIV/AIDS)
(415) 554-2830 24/7
(415) 554-2848 fax
Communicable Disease Control Unit
101 Grove Street, Room 408
San Francisco, CA 94102
cdcontrol@sfdph.org
HIV/AIDS Reporting
(415) 554-9050
(415) 431-0353 fax
STD reporting
(415) 487-5555
(415) 431-4628 fax
TB Reporting
(415) 206-8524
(415) 648-8369 fax
Foodborne Illness Reporting
Suspected food poisoning
(415) 554-2830
(415) 554-3875 fax
Outbreaks of foodborne illness
involving 4 or more persons
(415) 554-2830
(415) 554-3875 fax
Animal Bite Reporting (mammals only)
(415) 554-9422
(415) 554-9400 24/7
(415) 864-2866 fax
COMMUNICABLE DISEASE CONTROL & PREVENTION OFFICES
Bioterrorism & Infectious Disease
Emergencies Unit
(415) 554-2818
(415)554-2854 fax
101 Grove Street, Room 204
San Francisco, CA 94102
www.sfcdcp.org/idemergencies
Communicable Disease Control Unit
(415) 554-2830
(415) 554-2848 fax
101 Grove Street, Room 408
San Francisco, CA 94102
www.sfcdcp.org/diseasecontrol
Communicable Disease Prevention Unit
(415) 554-2830
(415) 554-2848 fax
101 Grove Street, Room 408
San Francisco, CA 94102
www.sfcdcp.org/izs
CONTACT INFORMATION
S.F. Dept. Public Health – Infectious Disease Emergencies July 2008 Page 1/ 1
Frequently Called Numbers
Animal Bite Reporting
Animal Care & Control
(415) 554-9422
(415) 554-9400 24-hour emergency dispatch
Bat Concerns
Animal Care & Control
(415)554-9400 24-hour emergency dispatch
Dead Bird Testing for West Nile Virus
California Department of Health Services
(877) WNV-BIRD (968-2473)
HIV Testing Services
Anonymous/confidential HIV counseling,
testing, linkages, referrals, and appointments
(800) 367-AIDS (2437)
9a-5p: M, W, Th, F; 9a-9p: T
Influenza Hotline
Communicable Disease Prevention Unit,
SFDPH
(415) 554-2681
Mosquito Control Problems
Environmental Health Services, SFDPH
(415) 252-3806
Needlestick Hotline
National Coalition Consultation Center
(888) 448-4911 (Consultation for clinicians,
post-exposure prophylaxis hotline)
Restaurant Sanitation Complaints
Environmental Health Services, SFDPH
(415) 252-3800
Rodents or Insect Infestations
Environmental Health Services, SFDPH
(415) 252-3800
Sewage Leak
Environmental Health Services, SFDPH
(415) 252-3800
Sexually Transmitted Diseases
STD Clinic, SFDPH
(415) 487-5500
Suspected Food Poisoning Reporting
(less than 4 people ill)
Environmental Health Section, SFDPH
(415) 252-3895
Travel Immunizations
Adult Immunization Clinic, SFDPH
(415) 554-2625
Tuberculosis (TB)
TB Clinic, SFDPH
(415) 206-8524
Unsanitary Living Conditions
Environmental Health Services, SFDPH
(415) 252-3805
CONTACT INFORMATION
S.F. Dept. Public Health – Infectious Disease Emergencies July 2008 Page 1/ 1
San Francisco Department of Public Health (SFDPH) Offices
SFDPH MAIN
San Francisco Department of Public Health
(415) 554-2500 main number
San Francisco Dept of Public Health
101 Grove Street, Room 100
San Francisco, CA 94102
www.sfdph.org
SFDPH OFFICES
AIDS Office
(415) 554-9000
25 Van Ness, 5
th
Floor
San Francisco, CA 94102
Communicable Disease Control &
Prevention Section
(415) 554-2830
(415) 554-2848 fax
101 Grove Street, Room 408
San Francisco, CA 94102
www.sfcdcp.org
Emergency Medical Services Section
(415) 355-2600
(415) 552-0194 fax
68 12
th
Street, Suite 220
San Francisco, CA 94103
www.sanfranciscoems.org
Environmental Health Section
(415) 252-3800
1390 Market Street, Suite 210
San Francisco, CA 94102
www.sfdph.org/eh
SF Public Health Laboratory
(415) 554-2800
(415) 431-0651 fax
101 Grove Street, Room 419
San Francisco, CA 94102
STD Prevention & Control
(415) 487-5500
(415) 437-9231 fax
356 7
th
Street
San Francisco, CA 94103
Tuberculosis Control Section
(415) 206-8524
(415) 648-8369 fax
SFGH Bldg 90, 4th floor (Ward 94)
2460 22nd Street
San Francisco, CA 94110
CONTACT INFORMATION
S.F. Dept. Public Health – Infectious Disease Emergencies July 2008 Page 1/1
COMMUNICABLE DISEASE REPORTING
Urgent Reports 24/7
(415) 554-2830
After hours, follow prompts to page
the on-call physician
Non-Urgent Reports
(415) 554-2830
(415) 554-2848
fax
cdcontrol@sfdph.org
101 Grove Street, Room 408
San Francisco, CA 94102
Business hours: Mon - Fri 8 am - 5 pm
The Communicable Disease Control Unit maintains a
reporting telephone line to respond to clinician infectious
disease reports 24 hours a day, 7 days a week. There are
over 80 legally reportable diseases and conditions in San
Francisco. Certain critical diseases must be reported
within one hour to the Department of Public Health while
others require same day notification or notification within
one week. See the list of legally reportable diseases in the
What to Report section.
After we receive an infectious disease report we
immediately take action to protect the health of San
Franciscans and our visitors.
H
OW WE RESPOND TO
INFECTIOUS DISEASE REPORTS….
INVESTIGATION
Case Investigation. Interview cases and
clinicians to identify risk factors and other potential
contacts. Evaluate patients/contacts in sensitive
occupations or settings that may pose a public health
concern (e.g. food handlers, daycare attendees,
health care workers or employees of group
residential facilities).
Source Investigation. Conduct an epidemiologic
investigation to identify the source of infection and
how it is being spread.
Lab Testing. Provide guidance on obtaining lab
tests to confirm diagnosis. Facilitate approvals for
obtaining specialized tests performed at city, state, or
federal public health labs.
INFECTION CONTROL
Recommendations. Work with infection control
practitioners to recommend measures to control and
prevent the spread of disease in healthcare settings.
Information & Education. Provide information
to cases, contacts, and the general public to prevent
and control the spread of disease in community
settings. In the event of an infectious disease
emergency, provide continued infection control
guidance and recommendations.
State & National Notification. Coordinate
notification of state and national health officials and
law enforcement, as necessary.
TREATMENT RECOMMENDATIONS
Postexposure & Preventive Treatment.
Assess the need for, and recommend preventive
treatments such as antibiotics and vaccines. In case
of mass exposure to a treatable infectious agent,
activate the local system for providing mass
treatment and/or prophylaxis.
COMMUNICATION WITH CLINICIANS
Health Alerts. Send Health Alerts, Advisories,
and Updates to clinicians regarding infectious
disease situations of public health concern.
Analysis of Surveillance Data. Analyze and
disseminate public health surveillance data to
clinicians and the general public.
WHAT HAPPENS WHEN YOU REPORT A DISEASE
S.F. Dept. Public Health – Infectious Disease Emergencies July 2008 Page 1/1
Clinicians perform many roles during infectious disease emergencies. Many actions assist the San
Francisco Department of Public Health (SFDPH) with timely investigations and effective public health
interventions. Other actions mitigate the need for patient treatment at acute care sites, address concerns of
the worried well, and maintain continuity of care for patients ill with diseases unrelated to the emergency.
KEY CLINICIAN ROLES
1. Recognize an infectious disease emergency.
See What to Report and Unusual Conditions
to Report to learn more about what we consider
infectious disease emergencies.
See the High Priority Disease chapters and the
“BT Syndrome Poster” to learn how to
recognize certain critical diseases.
2. Respond appropriately including
implementation of infection control measures,
initiation of diagnostic testing and therapy and
prophylaxis (if needed).
Familiarize yourself with initial patient
management protocols and infection control
measures. See the Infection Control and High
Priority Disease chapters.
Visit and bookmark the Communicable Disease
Control and Prevention website:
www.sfcdcp.org
.
Register to receive Health Alerts from SFDPH.
See instructions in the appendix.
3. Report the incident to response partners.
Keep SFDPH contact information and the
names and contact information of your hospital
infection control professionals handy.
CLINICIAN ROLES IN AN EMERGENCY
SFDPH Communicable Disease Reporting
(415) 554-2830 24/7
(415) 554-2848 fax
EMERGENCY TO DO LIST
Initial Steps
Implement infection control measures
o If patient is in the hospital, notify
Infection Control
Notify SFDPH
Notify your clinical lab and ensure
appropriate specimens are obtained for
routine and referral testing. Referral
testing may be coordinated through the
Public Health Lab system
Initiate patient management
If present, request that family and other
contacts remain for public health
interviews and prophylaxis if needed
Ensure that family and contacts are
educated about infection prevention
If family or other close contacts are not
present, obtain their contact
information to provide to SFDPH
Subsequent Steps
Follow incident progress and
recommendations via SFDPH Health
Alerts and/or our website:
www.sfcdcp.org
Make sure that your family, your staff,
and the families of your staff are safe
Keep office open unless advised
otherwise
Educate patients about measures to
prevent exposure and disease
Assess and care for the worried well
REPORTABLE DISEASES AND CONDITIONS
City and County of San Francisco San Francisco Department of Public Health
Title 17, California Code of Regulations (CCR) §2500, §2593, §2641-2643 and §2800-2812 §2500(b).
Every health care provider, knowing of or in attendance on a case or suspected case of any of the diseases or conditions listed below, must report to the local
health officer for the jurisdiction where the patient resides. Where no health care provider is in attendance, any individual having knowledge of a person who is suspected
to be suffering from one of the diseases or conditions listed below may make such a report to the local health officer for the jurisdiction where the patient resides.
WHO TO REPORT TO
DISEASE OR CONDITION / URGENCY REPORTING REQUIREMENTS
Acquired Immune Deficiency Syndrome (AIDS)
to AIDS Office
Alzheimer’s Diseases and Related Conditions
Amebiasis
Animal bites (mammals only)
to Animal Care and Control
Anthrax*
Avian Influenza (human)
Babesiosis
Botulism* (Infant, Foodborne, Wound)
Brucellosis*
Campylobacteriosis
Cancer, including benign and borderline brain tumors
(except (1) basal and squamous skin cancer unless
occurring on genitalia, and (2) carcinoma in-situ and CIN III
of the cervix)
Chancroid to STD Clinic
Chickenpox (only hospitalizations and deaths)
Chlamydial infections to STD Clinic
Cholera
Ciguatera Fish Poisoning
Coccidioidomycosis
Colorado Tick Fever
Conjunctivitis, Acute Infectious of the Newborn
(specify etiology)
Creutzfeldt-Jakob Disease (CJD)
Cryptosporidiosis
Cysticercosis
Dengue
Diarrhea of the Newborn, outbreaks
Diphtheria
Disorders Characterized by Lapses
of Consciousness
Domoic Acid Poisoning (Amnesic
Shellfish Poisoning)
Ehrlichiosis
Encephalitis, infectious (specify etiology)
Escherichia coli shiga toxin producing (STEC)
including E. coli O157
Foodborne illness
(2 or more cases from different households)
Giardiasis
Gonococcal infections to STD Clinic
Haemophilus influenzae invasive disease
(less than 15 years of age)
Hantavirus infections
Hemolytic Uremic Syndrome
Hepatitis, viral
Hepatitis A
Hepatitis B (specify acute case
or chronic)
Hepatitis C (specify acute case
or chronic)
Hepatitis D
(Delta)
Hepatitis, other acute
Human Immunodeficiency Virus (HIV) to
AIDS Office
Influenza deaths (less than 18 years of
age)
Kawasaki Syndrome (Mucocutaneous
Lymph Node Syndrome)
Legionellosis
Leprosy (Hansen Disease)
Leptospirosis
Listeriosis
Lyme Disease
Lymphogranuloma Venereum (LGV) to
STD Clinic
Malaria
Measles (Rubeola)
Meningitis (specify etiology)
Meningococcal infections
Mumps
Paralytic Shellfish Poisoning
Pelvic Inflammatory Disease (PID)
to STD Clinic
Pertussis (Whooping Cough)
Pesticide-related illness or injury (known or
suspected cases) to Environmental Health
Services
Plague (human or animal)*
Poliomyelitis, Paralytic
Psittacosis
Q Fever
Rabies (human or animal)
Relapsing Fever
Rheumatic Fever, Acute
Rocky Mountain Spotted Fever
Rubella (German Measles)
Rubella Congenital Syndrome
Salmonellosis (other than Typhoid Fever)
Scombroid Fish Poisoning
Severe Acute Respiratory Syndrome
(SARS)
Shiga toxin (detected in feces)
Shigellosis
Smallpox (Variola)*
Staphylococcus aureus infections, severe
(ICU/death) in a previously healthy person
Streptococcal infections, outbreaks
of any type and individual cases in food
handlers and dairy workers only
Syphilis to STD Clinic
Taeniasis
Tetanus
Toxic Shock Syndrome
Toxoplasmosis
Transmissable Spongiform
Encephalopathies (TSE)
Trichinosis
Tuberculosis to Tuberculosis Clinic
Tularemia*
Typhoid Fever (cases and carriers)
Typhus Fever
Vibrio infections
Viral Hemorrhagic Fevers* (e.g. Crimean-
Congo, Ebola, Lassa and Marburg viruses)
Water-associated disease (e.g. Swimmer’s
Itch and Hot Tub Rash)
West Nile Virus
Yellow Fever
Yersiniosis
ANY UNUSUAL DISEASES
NEW DISEASE OR SYNDROME
NOT PREVIOUSLY RECOGNIZED
OUTBREAKS OF ANY DISEASE
REPORT OUTBREAKS, DISEASES, AND CONDITIONS TO COMMUNICABLE DISEASE CONTROL UNIT UNLESS OTHERWISE INDICATED
*Potential Bioterrorism Agents
Effective February 2008 / Revised 12/08
URGENCY REPORTING KEY
Report immediately by telephone Report within one working day of identification Report within seven calendar days by FAX, phone or mail
COMMUNICABLE DISEASE CONTROL UNIT
PHONE:
(415) 554-2830 FAX: (415) 554-2848
M-F 8AM to 5PM
For urgent reports after hours, follow the prompts
to page the on-call MD
AIDS OFFICE
PHONE:
(415) 554-9050
STD CLINIC
PHONE:
(415) 487-5555 FAX: (415) 431-4628
1
7
7
7
7
7
7
7
7
7
7
7
7
7
7
7
7
7
7
7
7
7
7
7
7
7
7
7
7
7
7
7
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
7
7
7
7
7
7
7
TUBERCULOSIS CLINIC
PHONE:
(415) 206-8524 FAX: (415) 648-8369
ANIMAL BITES (mammals only)
PHONE:
(415) 554-9422 FAX: (415) 864-2866
ENVIRONMENTAL HEALTH SERVICES
PHONE:
(415) 252-3862 FAX: (415) 252-3818
7
7
S.F. Dept. Public Health – Infectious Disease Emergencies July 2008 Page 1/1
C
OMMUNICABLE DISEASE REPORTING
Urgent Reports 24/7
(415) 554-2830
After hours, follow prompts to page the on-
call physician
Non-urgent Reports
(415) 554-2830
(415) 554-2848
fax
cdcontrol@sfdph.org
101 Grove Street, Room 408
San Francisco, CA 94102
Business Hours: Mon - Fri 8 am - 5 pm
The San Francisco Department of Public Health (SFDPH)
depends on clinicians to identify and report cases of
communicable diseases. Clinicians may be the first to see
a potential outbreak in the making and their prompt
notification to SFDPH enables us to investigate and begin
disease control activities as soon as possible. For some
diseases every hour makes a difference in preventing
illness and death.
To improve our ability to control outbreaks the Sentinel
Event Enhanced Passive Surveillance (SEEPS) Project
works to strengthen clinicians’ ability to identify emerging
infectious diseases and those that may result from
biological terrorism. Due to their deadliness it is
particularly important that cases are recognized,
diagnosed, and reported quickly. Knowing the clinical
features and maintaining an index of suspicion for unusual
cases and reporting them to SFDPH could save lives.
Potentially unusual patterns of disease include:
1. Multiple similarly presenting cases, especially if these are geographically
associated or closely clustered in time
Example: persons who attended the same event or who work in the same building
2. An increase in a common syndrome occurring out of season
Example: many cases of influenza-like illness in summer
3. An unusual age distribution for common diseases
Example: many cases of chickenpox-like illness in adult patients expected to be immune
4. Serious, unexpected, unexplained acute illness with atypical host characteristics
Examples: severe illness in a young patient without immunologic defects, underlying
illness, recent travel or other exposure to a potential source of infection
Due to their rarity, some of the following diseases and conditions may not be immediately recognizable.
However, maintaining a reasonable index of suspicion and reporting unusual conditions could assist in
treating patients and safeguarding the public.
IMMEDIATE NOTIFICATION REQUIRED TO SFDPH (within one hour)
Anthrax*
Botulism*
Brucellosis*
Plague*
Smallpox*
Tularemia*
Viral hemorrhagic
fevers*
Meningococcal infections
Measles
Avian Influenza
SARS
Diphtheria
Hantavirus infections
Yellow fever
Shiga toxin producing E. coli
(STEC) including E. coli 0157
Shiga toxin (in feces)
Hemolytic Uremic Syndrome
Scombroid fish poisoning
Ciguatera fish poisoning
Paralytic shellfish poisoning
Domoic acid poisoning
Rabies
Cholera
Diarrhea of the
Newborn (Outbreak)
Any unusual diseases
Outbreaks -any disease
* Potential bioterrorism
agents
MMWR Morb Mortal Wkly Rep. 2001 Oct 19;50(41):893-7.
UNUSUAL CONDITIONS TO REPORT
S.F. Dept. Public Health – Infectious Disease Emergencies July 2008 Page 1/2
Community practitioners should be personally and professionally prepared to respond to a variety of
infectious disease emergencies. The following are suggestions to help with this process.
FAMILY PLAN
Ensure that your family is well.
Create and practice a family disaster plan. For more information see:
o SF OES, Household Family Plan: www.72hours.org
o Red Cross, Your Family Disaster Plan:
www.prepare.org/basic/DisasterPlan.pdf
Place fully stocked disaster kits in your home and car with a three-day supply of food and water.
o Red Cross, Emergency supply Kit Guide: www.prepare.org/basic/SuppliesKit.pdf
Encourage staff to develop and practice family disaster plans.
CLINIC & OFFICE PLAN
Take steps to ensure the safety and well being of your staff. For suggestions and resources, see the Clinic
and Office Disaster & Emergency Planning section in this Guide.
Provide personal emergency kits and emergency contact numbers to staff.
Make a telephone tree to notify staff in an emergency
Develop and practice your clinic or office disaster and evacuation plan.
Know the expected clinic/office emergency roles and responsibilities (including who assists patients
and who will account for them when leaving the building).
Know clinician roles and responsibilities in a community disaster. See the Roles and
Responsibilities section.
Identify items that should be taken in an evacuation (medicine, backup data, etc.)
EMERGENCY INFORMATION
Know where to obtain reliable San Francisco specific information.
Visit and bookmark the Communicable Disease Control and Prevention website:
www.sfcdcp.org
.
Register for Health Alerts. See instructions in the appendix.
Note the radio stations that will provide emergency information:
KCBS 740 AM, KGO 810 AM, KNBR 680 AM, KQED 88.5 FM, KSJO 92.3 FM
PREPARING FOR INFECTIOUS DISEASE EMERGENCIES
S.F. Dept. Public Health – Infectious Disease Emergencies July 2008 Page 2/2
LEARN ABOUT INFECTIOUS DISEASE EMERGENCY
Know the details of infectious disease emergencies.
Know what to report. See the What to Report section containing:
o List of diseases clinicians are legally required to report
o List of unusual conditions for which we request reports
Review the potential bioterrorism related syndromes and the biological threat
diseases (e.g., anthrax, avian influenza, botulism, brucellosis, plague,
smallpox, tularemia, viral hemorrhagic fevers):
o See the Bioterrorism Syndromes poster
o See information in the High Priority Diseases section
Maintain a reasonable index of suspicion
Become knowledgeable and train staff on infection control measures. See
Infection Control Guidelines.
REPORT TO SFDPH ON A ROUTINE BASIS
Routinely use components of your response plan. Informing SFDPH about diagnosed or suspected cases
of reportable communicable diseases assists SFDPH disease control interventions and improves the
ability to communicate with SFDPH in emergencies.
Review and post:
o List of diseases clinicians are legally required to report
o List of unusual conditions for which we request reports
Place SFDPH contact information in rolodex files
Place SFDPH stickers on or near primary phones
S.F. Dept. Public Health – Infectious Disease Emergencies July 2008 Page 1/3
SUGGESTED ITEMS TO INCLUDE IN
A CLINIC OR OFFICE DISASTER PLAN
Purpose of the disaster plan
Scope of the disaster plan
Plan activation
Who can active the plan
Circumstances when the plan
should be activated.
Disaster plan mission statement
Leadership and succession
leadership
Delegation of authority
Supporting plans and resources
Legal authorities, codes, and policies
Plan administration
(e.g., distribution, updates)
Staff activation and call down
procedures
Mutual aid agreements
Communication procedures
Organization chart
Job action sheets
Specific plans
Evacuation plan
Transportation plan
Medical
m
ana
g
ement
Most health facilities in California (e.g., hospitals, long term
care facilities, primary care clinics, adult day care centers)
are required by state law to have a plan or program for
addressing disasters. Medical offices are required to comply
with local business ordinances including building and fire
codes. It is also prudent for medical offices to develop
disaster plans.
Several organizations have created documents to assist in the
development of disaster plans. The California Office of
Emergency Services (CA OES) and the California Primary
Care Association have developed guidance and templates for
clinic disaster plans.* The CDC and Red Cross provide
guides for developing business disaster plans.*
Development and implementation of a disaster plan is
divided into four phases:
1. Hazard Mitigation
2. Preparedness
3. Response
4. Recovery
You will find issues under each stage that need to be
addressed in the development of your facility’s disaster plan.
Ideally plans should coordinate with neighborhood, local
hospital, county and state partners.
* EMERGENCY PLAN RESOURCES
CLINIC/OFFICE DISASTER & EMERGENCY PLANNING
San Francisco Department of Emergency Management
Homepage: www.sfgov.org/site/oes_index.asp
How to prepare an emergency (family) plan: www.72hours.org
Red Cross, Family Disaster Plan Guidance, Emergency Supply Kit Guide, Workplace Preparedness:
www.prepare.org
California Primary Care Association
Clinic Disaster Plan Template: www.cpca.org/resources/cepp
California Office of Emergency Services, Clinic Disaster Plan Guide & Templates:
www.oes.ca.gov/Operational/OESHome.nsf/ PDF/ClinicDisasGuide/$file/ClinicDisasGuide.pdf
CDC Emergency Preparedness For Business
www.cdc.gov/niosh/topics/prepared
S.F. Dept. Public Health – Infectious Disease Emergencies July 2008 Page 2/3
DISASTER PLAN DEVELOPMENT PHASES
The information described below on the phases of disaster plan development and implementation is adapted
from the California Office of Emergency Service Clinic Disaster Plan Guidance from June 2002. (See url
listed above for access to the complete document.)
Hazard Mitigation
Hazard mitigation will identify ways of minimizing future losses.
a. Hazard Vulnerability Analysis is the identification of hazards and the direct and indirect effect
these hazards may have on the facility.
b. Structural Mitigation is reinforcing, bracing, anchoring, bolting, strengthening, or replacing any
portion of the building that may become damaged and cause injury.
c. Nonstructural Mitigation reduces the threat to safety posed by the effects of earthquakes on such
nonstructural elements as building contents, internal utility systems, interior glass and decorative
architectural walls and ceilings. These actions involve identifying nonstructural fixtures and
equipment, which are vulnerable to an earthquake and which are either essential to continued
operations or a threat to public safety.
Preparedness
a. Disaster Plan. A well-written plan that has been tested will provide for an efficient systematic
response to any type of a disaster or emergency.
b. Hazardous Materials Management - Internal and External. Clinics and/or offices may store
and/or handle hazardous materials and the potential for these materials to be released is significant.
Each clinic should identify these materials and develop procedures for safely handling, containing
and neutralizing them. Staff training should include, but not be limited to, location of hazardous
materials, safe handling, proper notification procedures, proper evacuation procedures, potential risks,
storage, containment, neutralization, decontamination techniques and medical management of
victims. Many Federal, State and local statutes, regulation and ordinances govern the handling and
storage of hazardous materials. To determine the level of and need for compliance it is important that
clinics or offices contact SFDPH Hazardous Materials Unified Program Agency at (415) 252-3900.
c. Weapons of Mass Destruction (WMD). Preparations for an event involving weapons of mass
destruction – chemical, biological, nuclear, radiological or explosives (CBRNE) – begins with
understanding the threat agents and the consequences of their use. This reference guide describes
diseases potentially related to bioterrorism, appropriate initial responses and the roles and
responsibilities of responders including community health care providers. Share the Infectious
Disease Emergencies Guide with staff and place in a prominent and easily accessible location.
d. Managing Volunteers. Volunteers have a role in a disaster response but management of this resource
is crucial.
S.F. Dept. Public Health – Infectious Disease Emergencies July 2008 Page 3/3
e. Donations Management. Donations can quickly overwhelm a clinic especially when they are
unsolicited. Coordination is accomplished by developing a plan prior to the emergency to handle
receiving and distribution of the goods.
f. Training and Exercises. Training is achieved through exercising the clinic disaster plan without the
stress of an actual disaster/emergency. This provides staff with the opportunity to become familiar
with the plan and procedures, their roles and responsibilities, and the information and skills required
to perform their duties during an emergency.
Response
a. Implementation of the Disaster Plan.
b. Organizational Chart. The organizational chart provides structure to a disaster or emergency
response and features command, operations, planning and intelligence, logistics, and
finance/administration positions. With small offices or clinics some functions may not be activated
and/or some people may be responsible for more than one function.
c. Emergency Operation Center (EOC). The EOC is a key to successful response and recovery
operations. It is the central location where all activities are coordinated. Coordination of activities will
ensure that all tasks are accomplished with little or no duplication of effort.
Recovery
Clinics and offices should try to remain operational following an emergency. Planning can enable a more
rapid and successful recovery or return to normal activities and minimize financial losses. Recovery
issues to prepare for include: coping with structural and nonstructural damage to facilities, maintaining an
inventory of damage and/or loss, accounting for lost revenue through disruption of services, personnel
policies during and after an emergency, and meeting the psychological needs of staff and patients.
a. Financial Recovery Sources. In order to recover costs related to the disaster, complete
documentation including photographs of damage is essential. Resources available to your facility
during and after a major disaster could include:
Public Assistance (FEMA/OES)
Small Business Administration (SBA)
Federal Grant
Insurance Carriers
S.F. Dept. Public Health – Infectious Disease Emergencies July 2008 Page 1/1
For the workplace, where you might be confined for several hours, or perhaps overnight, the following
supplies are recommended. More information is at: www.redcross.org/services/disaster/beprepared.
Flashlight with extra batteries
Use a flashlight if the power is out. Do not use
candles or open flames.
Battery-powered radio
News about the emergency may change rapidly
as events unfold. Radio reports will give
information about the areas most affected.
Food
Enough non-perishable food to sustain you for at
least one day, is suggested. Select foods that
require no refrigeration, preparation or cooking,
and little or no water. The following items are
suggested:
• Ready-to-eat canned meals, meats, fruits, and
vegetables.
• Canned juices.
• High-energy foods (granola/energy bars, etc.)
Water
Keep at least one gallon of water available, or
more if you are on medications that require
water or that increase thirst.
Medications
Include usual non-prescription medications that
you take, including pain relievers, stomach
remedies, etc.
If you use prescription medications, keep at least
a three-day’s supply of these medications at your
workplace. Consult with your physician or
pharmacist how these medications should be
stored, and your employer about storage
concerns.
First Aid Supplies
Have the following essentials:
(20) Adhesive bandages, various sizes.
• (1) 5” x 9” sterile dressing.
• (1) Conforming roller gauze bandage.
• (2) Triangular bandages.
• (2) 3 x 3 Sterile gauze pads.
• (2) 4 x 4 Sterile gauze pads.
• (1) Roll 3” cohesive bandage.
• (2) Germicidal hand wipes or waterless
alcohol-based hand sanitizer.
• (6) Antiseptic wipes.
• (2) Pair large medical grade non-latex gloves
• Adhesive tape, 2” width.
• Anti-bacterial ointment.
• Cold pack.
• Scissors (small, personal).
• Tweezers.
• CPR breathing barrier, such as a face shield
Tools and Supplies
• Emergency “space” blanket (mylar).
• Paper plates and cups, plastic utensils.
• Non-electric can opener.
• Personal hygiene items, including a toothbrush,
toothpaste, comb, brush, soap, contact lens
supplies, and feminine supplies.
• Plastic garbage bags, ties (for personal
sanitation uses).
• At least one complete change of clothing and
footwear, including a long sleeved shirt and
long pants, as well as closed-toed shoes or
boots.
• If you wear glasses, keep an extra pair with
your workplace disaster supplies.
General Information
• Your kit should be adjusted based on your own
personal needs.
• Do not include candles, weapons, toxic
chemicals, or controlled drugs unless
prescribed by a physician.
PERSONAL OFFICE DISASTER KIT
Excerpted from the American Red Cross Personal Workplace Disaster Supplies Kit
S. F Dept. Public Health – Infectious Disease Emergencies ANTHRAX, July 2008 Page 1/14
Outline
Introduction
Epidemiology
Clinical Features
Differential Diagnosis
Laboratory Diagnosis
Treatment and Prophylaxis
Complications
Infection Control
Pearls and Pitfalls
References
Immediately report any suspected or
confirmed cases of anthrax to:
SFDPH Communicable Disease Control
(24/7 Tel: 415-554-2830)
- By law, health care providers must report suspected
or confirmed cases of anthrax to their local health
department immediately [within 1 hr].
- SFDPH Communicable Disease Control can facilitate
specialized testing and will initiate the public health
response as needed.
Also notify your:
Infection Control Professional
Clinical Laboratory
INTRODUCTION
Anthrax is an acute infection caused by Bacillus anthracis, a large, gram-positive, spore-forming,
aerobic, encapsulated, rod-shaped bacterium. Spores germinate and form bacteria in nutrient-rich
environments, whereas bacteria form spores in nutrient-poor environments. The anthrax bacillus
produces high levels of two toxins: Edema toxin causes massive edema at the site of germination,
and lethal toxin leads to sepsis. Severity of anthrax disease depends on the route of infection and
the presence of complications, with case-fatality ranging from 5% to 95% if untreated.
1-3
The Working Group for Civilian Biodefense considers B. anthracis to be one of the most serious
biological threats. Anthrax has been weaponized and used. It can be fairly easily disseminated and
causes illness and death. Of the potential ways that B. anthracis could be used as a biological
weapon, an aerosol release is expected to have the most severe medical and public health
outcomes.
1
EPIDEMIOLOGY
Anthrax as a Biological Weapon
Anthrax was successfully used as a biological weapon in the United States in October 2001. Cases
resulted from direct or indirect exposure to mail that was deliberately contaminated with anthrax
spores. In total, 22 cases were identified, 11 with inhalational (five fatal) and 11 with cutaneous
anthrax (seven confirmed, four suspected).
Several countries have had anthrax weaponization programs in the past, including the United
States. In 1979 an outbreak of anthrax in the Soviet Union resulted from accidental release of
ANTHRAX JULY 2008
S. F Dept. Public Health – Infectious Disease Emergencies ANTHRAX, July 2008 Page 2/14
anthrax spores from a facility producing weaponized anthrax. Of 77 reported human cases, all but
two were inhalational, and there was an 86% fatality rate.
4
Experts believe that an aerosol release of weapons-grade spores is the most likely mechanism for
use of anthrax as a biological weapon in the future. Anthrax spores could also be used to
deliberately contaminate food and water. Spores remain stable in water for several days and are
not destroyed by pasteurization.
1
An intentional release of anthrax may have the following characteristics:
1-3
Multiple similarly presenting cases clustered in time:
o Severe acute febrile illness or febrile death
o Severe sepsis not due to predisposing illness
o Respiratory failure with a widened mediastinum on CXR
Atypical host characteristics: unexpected, unexplained cases of acute illness in previously
healthy persons who rapidly develop a progressive respiratory illness
Multiple similarly presenting cases clustered geographically:
o Acute febrile illness in persons who were in close proximity to a deliberate release
of anthrax
Absence of risk factors: patients lack anthrax exposure risk factors (e.g., veterinary or
other animal handling work, meat processing, work that involves animal hides, hair, or
bones, or agricultural work in areas with endemic anthrax)
Intentionally released anthrax spores may be altered for more efficient aerosolization and lethality
(e.g., highly concentrated, treated to reduce clumping and reduce particle size, genetically modified
to increase virulence, resist antimicrobials and reduce vaccine efficacy).
Naturally Occurring Anthrax
Reservoir
The natural reservoir for B. anthracis is soil, and the predominant hosts are herbivores (cattle,
sheep, goats, horses, pigs, and others) that acquire infection from consuming contaminated soil or
feed. Anthrax spores can persist in soil for years and are resistant to drying, heat, ultraviolet light,
gamma radiation, and some disinfectants.
5
Anthrax in animals is endemic in many areas of the
world and anthrax outbreaks in animals occur sporadically in the United States.
Mode of transmission
Anthrax is generally a zoonotic disease. Humans become infected through contact with infected
animals and animal products through several mechanisms:
1, 5, 6
contact with infected animal tissues (e.g., veterinarians, animal handlers, meat processors,
and other processes that involve animal hides, hair, and bones) or contaminated soil
ingestion of contaminated, undercooked meat from infected animals
inhalation of infectious aerosols (e.g., those generated during processing of animal
products, such as tanning hides, processing wool or bone)
Person-to-person transmission of B. anthracis does not occur with gastrointestinal (GI) or
inhalational anthrax, but has been reported rarely with cutaneous anthrax.
1
S. F Dept. Public Health – Infectious Disease Emergencies ANTHRAX, July 2008 Page 3/14
Worldwide Occurrence
Worldwide, approximately 2000 cases are reported annually. Anthrax is more common in
developing countries with less rigorous animal disease control programs. Cases of human anthrax
are most often reported in South and Central America, Southern and Eastern Europe, Asia, Africa,
the Caribbean, and the Middle East.
1
The largest reported outbreak of human anthrax occurred in
Zimbabwe (1979-1985), which involved more than 10,000 individuals and was associated with
anthrax disease in cattle.
2
United States Occurrence
Naturally occurring anthrax is rare in the United States, with approximately 1-2 cases reported each
year. The majority of anthrax cases in the United States are cutaneous and acquired occupationally
in workers who come in contact with animals or animal products. Only 19 cases of naturally
occurring inhalational anthrax have been reported since 1900, and there have been no confirmed
gastrointestinal cases.
2, 7
Recent cases of naturally occurring anthrax include:
In 2006, a New York City resident contracted inhalational anthrax while making drums from
goat hides imported from Africa. The untanned hides were contaminated with anthrax
spores, which may have been aerosolized during removal of hair.
7
The CDC believes that
this was an isolated case and considers handling animal skins or making drums to be a low
risk for cutaneous anthrax and extremely low risk for inhalational anthrax.
8
In 2002, two cases of cutaneous anthrax were reported. A laboratory worker from a Texas
lab that processed environmental B. anthracis specimens contracted cutaneous anthrax
through direct contact with a contaminated surface.
9
The second case occurred in a
veterinarian who contracted the infection from a cow during necropsy.
10
Two cases of human cutaneous anthrax were reported following epizootics in North Dakota
(2000) and southwest Texas (2001). Both cases resulted from exposure during disposal of
infected animal carcasses.
11
Occurrence in California and San Francisco
From 1994 to 2007, no cases of anthrax were reported in California.
12-15
However, in 2001 an
outbreak of bovine anthrax caused the death of 21 beef cattle in a rural section of Santa Clara
County.
CLINICAL FEATURES
There are three primary clinical types of anthrax disease, inhalational, cutaneous and
gastrointestinal, which result from the way infection is acquired. Anthrax meningitis, which
generally occurs as a complication of these primary forms of disease, is most likely to be seen with
inhalational anthrax.
Anthrax infection is a severe clinical illness and can be life-threatening. Case fatality varies by the
clinical type of disease. Overall case-fatality rates have declined because of more prompt
administration of antibiotics and improved supportive care. Compared to historical rates, mortality
has decreased from 86-95% to 45% for inhalational anthrax, 5-20% to less than 1% for cutaneous
anthrax, and 25-60% to 12% for gastrointestinal anthrax. Anthrax meningitis case-fatality rates
approach 95% even with antibiotic treatment.
2, 3, 16
S. F Dept. Public Health – Infectious Disease Emergencies ANTHRAX, July 2008 Page 4/14
In the event of bioterrorism, the method of dissemination would influence the type of clinical
disease that would be expected. Following an aerosol release, the majority of cases would be
inhalational with some cutaneous cases whereas use of a small volume powder could result in both
inhalational and cutaneous anthrax cases (as seen in the 2001 attacks). Gastrointestinal cases
might occur following contamination of food or water.
Inhalational Anthrax
Inhalational anthrax is caused by inhalation of spores that reach the alveoli, undergo phagocytosis
and travel to regional lymph nodes. The spores then germinate to become bacterial cells, which
multiply in the lymphatic system and cause lymphadenitis of the mediastinal and peribronchial
lymph nodes. The bacteria release toxins that cause hemorrhage, edema, and necrosis. Bacteria
entering the bloodstream lead to septicemia, septic shock, and death. Systemic infection following
inhalational anthrax is almost always fatal.
1
One of the key clinical features of inhalational anthrax is evidence of pleural effusion and
mediastinal widening on CXR or chest computed tomographic (CT) scan. Based on experience from
the 2001 attacks, chest CT (without contrast) was found to be more sensitive than CXR for
identification of mediastinal widening typical of inhalational anthrax.
3
CLINICAL FEATURES: INHALATIONAL ANTHRAX
1, 2, 6, 17
Incubation
Period
1-6 days (range <1 day to 8 weeks)
Transmission Inhalation of aerosolized spores
Signs and
Symptoms
Initial presentation: Non-specific symptoms (low-grade fever, chills, nonproductive
cough, malaise, fatigue, myalgias, profound sweats, chest discomfort)
Intermediate presentation: Abrupt onset of high fever, dyspnea, progressive respiratory
distress, confusion, nausea or vomiting
Fulminant disease progression, if untreated
Progression
and
Complications
Severe respiratory distress (dyspnea, stridor, cyanosis), which may be preceded by 1-3
days of improvement
Pleural effusions
Meningitis
Shock
Laboratory and
Radiographic
Findings
Chest CT or radiograph: mediastinal widening (often), pleural effusions that are
commonly hemorrhagic (often), infiltrates (rare)
Gram-positive bacilli on unspun peripheral blood smear or CSF
Elevated transaminases
Hypoxemia
Metabolic acidosis
Total WBC count normal or slightly elevated with elevated percentage of neutrophils or
band forms
CSF, cerebrospinal fluid; WBC, white blood (cell) count.
S. F Dept. Public Health – Infectious Disease Emergencies ANTHRAX, July 2008 Page 5/14
Cutaneous Anthrax
In cutaneous anthrax, spores or bacilli are introduced through cuts or breaks in the skin. Spores
germinate at the site of contact and release toxins, causing development of a lesion and edema.
Organisms may be carried to regional lymph nodes and cause painful lymphadenopathy and
lymphangitis. Septicemic complications of cutaneous anthrax occur in 10-20% of untreated cases.
1-
3
CLINICAL FEATURES: CUTANEOUS ANTHRAX
1, 2, 6, 17
Incubation
Period
3-4 days (range 1–12 days)
Transmission
Direct skin contact with spores; in nature, contact with infected animals or animal
products (usually related to occupational exposure)
Bite of infective arthropod (rare)
Signs and
Symptoms
Local skin involvement after direct contact with spores or bacilli (commonly seen on
hands, forearms, head, and neck)
Skin lesion with the following progression: 1) Development of a papular lesion and
localized itching, 2) papule turns into vesicular or bulbous lesion accompanied by
painless edema, 3) lesion becomes necrotic and vesicles may surround the ulcer, and 4)
lesion develops painless black eschar within 7–14 days of initial lesion
Lymphadenopathy and lymphangitis
Fever and malaise (common)
Progression
and
Complications
Bacteremia
Meningitis
Extensive edema causing airway compression
Sepsis
Laboratory
Findings
Bacilli may be seen on Gram stain of subcutaneous tissue
Pediatric considerations: A case of cutaneous anthrax occurred in a 7-month old during the anthrax
attack of 2001. This case was difficult to recognize and rapidly progressed to severe systemic
illness despite timely antibiotic treatment. Clinical features included a painless draining lesion with
edema that developed into an eschar, fever, leukocytosis, severe microangiopathic hemolytic
anemia, renal failure, and coagulopathy.
18
Gastrointestinal Anthrax
Gastrointestinal (GI) anthrax results from ingestion of B. anthracis bacteria, such as may be found
in poorly cooked meat from infected animals. The incubation period for GI anthrax is 1-7 days.
Two clinical presentations have been described: intestinal and oropharyngeal.
With intestinal anthrax, intestinal lesions occur in the ileum or cecum and are followed by regional
lymphadenopathy. Symptoms of intestinal anthrax are initially nonspecific and include low-grade
fever, malaise, nausea, vomiting, anorexia and fever. As disease progresses, abdominal pain,
hematemesis, and bloody diarrhea develop. The patient may present with findings of an acute
abdomen. After 2-4 days, ascites develop and abdominal pain lessens. Hematogenous spread with
S. F Dept. Public Health – Infectious Disease Emergencies ANTHRAX, July 2008 Page 6/14
resultant septicemia can occur. Mesenteric adenopathy on CT scan is likely, and mediastinal
widening on CXR is possible.
1, 2, 5, 6
In oropharyngeal anthrax, a mucosal ulcer occurs initially in the mouth or throat, associated with
fever, throat pain, and dysphasia. This is followed by cervical edema and regional
lymphadenopathy. Ulcers may become necrotic with development of a white patch covering the
ulcer. Swelling can become severe enough to affect breathing. Hematogenous spread, septicemia,
and meningitis can occur.
1, 2, 5, 6
Gram stain of ascitic fluid, oropharyngeal ulcers, or unspun peripheral blood may show Gram-
positive rods. Leukocytosis with left shift may be present. B. anthracis can be cultured from
oropharyngeal swabs and stool specimens.
6
Anthrax Meningitis
Anthrax meningitis can occur as a complication of cutaneous, inhalational, or GI anthrax, but is
most commonly seen with inhalational anthrax (up to 50%). Patients may or may not present with
symptoms of the primary site of infection. In addition to typical symptoms of bacterial meningitis,
anthrax meningitis may involve hemorrhage or meningoencephalitis. Case fatality with anthrax
meningitis is greater than 90%. Even one case of anthrax meningitis should alert public health
authorities to identify the source of exposure and investigate the possibility of bioterrorism.
3
Anthrax and Pregnant Women
Maternal and perinatel complications are not completely understood, because anthrax infection
during pregnancy is rare. Preterm delivery may be one of the major complications.
19
DIFFERENTIAL DIAGNOSIS
Because of its mild, nonspecific nature in the early states, a high index of suspicion is necessary to
make a timely diagnosis of anthrax. Screening protocols and clinical prediction tools have been
proposed and partially evaluated.
20
Prompt administration of antibiotics can be critical to patient
survival; therefore, clinicians should administer appropriate antibiotics when the diagnosis is
suspected.
1
Differential: Inhalational Anthrax
1, 2, 5, 6, 21
Early disease mimics influenza and other respiratory infections. However nasal symptoms are
typically not present and rapid diagnostic tests, such as nasopharyngeal swabs for detection of
respiratory virus antigens, would typically be negative.
Key features that distinguish inhalational anthrax from other conditions are:
CXR is abnormal even during early stages of influenza like illness
CXR or chest CT show widened mediastinum and pleural effusion but minimal or no
pneumonitis
S. F Dept. Public Health – Infectious Disease Emergencies ANTHRAX, July 2008 Page 7/14
Features that distinguish inhalational anthrax from influenza:
Neurological symptoms without headache (e.g., confusion, syncope) and nausea/vomiting
are more common in inhalational anthrax
Rhinorrhea and pharyngitis were uncommon in inhalational anthrax cases from 2001 U.S.
attack
Other conditions to consider are:
bacterial pneumonia (Mycoplasma,
Staphylococcus, Streptococcus, Haemophilus,
Klebsiella, Moraxella, Legionella)
Chlamydia infection
influenza
other viral pneumonia (respiratory syncytial
virus [RSV], cytomegalovirus [CMV],
hantavirus)
Q fever
pneumonic plague
tularemia
primary mediastinitis
ruptured aortic aneurysm
histoplasmosis
coccidiodomycosis
silicosis
sarcoidosis
Differential: Cutaneous Anthrax
3, 5, 6
Key features that distinguish cutaneous anthrax are:
painlessness of the lesion itself
large extent of local edema
Other conditions to consider:
ecthyma gangrenosum
ulceroglandular tularemia
bubonic plague
cellulitis (staphylococcal or streptococcal)
brown recluse spider bite
necrotizing soft tissue infections,
(e.g., Streptococcus, Clostridium)
Coumadin or heparin necrosis
rickettsial infection
necrotic herpes simplex infection
orf virus infection
glanders
cutaneous leishmaniasis
cat scratch fever
melioidosis
Differential: Gastrointestinal Anthrax
2, 5
The differential diagnosis for the intestinal form of the disease includes:
typhoid fever
intestinal tularemia
acute bacterial gastroenteritis (e.g., Campylobacter, Shigella, toxicogenic Escherichia coli,
Yersinia)
bacterial peritonitis
peptic or duodenal ulcer
any other causes of acute abdomen
S. F Dept. Public Health – Infectious Disease Emergencies ANTHRAX, July 2008 Page 8/14
If you are testing or considering testing for
anthrax, you should:
IMMEDIATELY notify
SFDPH Communicable Disease Control
(24/7 Tel: 415-554-2830).
SFDPH can authorize and facilitate testing, and
will initiate the public health response as needed.
Inform your lab that anthrax is under
suspicion. Labs may view Gram-positive
bacilli as contaminants and may not pursue
further identification unless notified.
The differential diagnosis for the oropharyngeal form of the disease includes:
streptococcal pharyngitis
infectious mononucleosis
diphtheria
pharyngeal tularemia
other causes of pharyngitis (e.g., enteroviral vesicular, herpetic, anaerobic or Vincent’s
angina, Yersinia enterocolitica)
Differential: Anthrax Meningitis
3, 5
A key feature that distinguishes anthrax meningitis is bloody cerebrospinal fluid (CSF) containing
gram-positive bacilli.
Other conditions to consider are:
subarachnoid hemorrhage
bacterial meningitis
aseptic meningitis
LABORATORY AND RADIOGRAPHIC FINDINGS
The diagnosis of anthrax requires a high index
of suspicion because the disease often presents
with nonspecific symptoms. Routine laboratory
and radiographic findings for specific clinical
presentations of anthrax are listed in the
clinical features tables.
Initial identification and diagnosis of the
organism relies on evaluation of infected tissue
(blood, sputum, CSF, fluid collected from an
unroofed vesicle, ulcer, eschar, or skin lesion
scraping, or stool). The gold standard for
anthrax diagnosis is direct culture of clinical
specimens onto blood agar with demonstration of typical Gram stain, motility, and biochemical
features. Blood cultures, which are positive nearly 100% of the time in inhalational anthrax, should
be obtained prior to antibiotic administration because there is rapid sterilization of blood after a
single dose of antibiotics. Because laboratories may view gram-positive bacilli as contaminants and
because B. anthracis may be a risk to laboratory personnel, clinicians should notify the laboratory
when anthrax is suspected.
1-3, 5
Although rapid diagnostic tests are not widely available, the public health laboratory system may be
able to provide this testing on clinical specimens. Other tests available through the public health
S. F Dept. Public Health – Infectious Disease Emergencies ANTHRAX, July 2008 Page 9/14
These recommendations are current as of this document date. SFDPH will provide periodic updates as needed
and situational guidance in response to events (www.sfcdcp.org).
laboratory system include polymerase chain reaction (PCR), serologic tests, and
immunohistochemistry.
22
Testing for Exposure to aerosolized Anthrax
Nasal swab cultures have been used to study environmental exposure to aerosolized anthrax, they
are not recommended for use in the clinical setting. The sensitivity, specificity, and predictive value
of nasal swab cultures is not known.
6
TREATMENT AND PROPHYLAXIS
Treatment of Confirmed or Suspected Anthrax
This section refers to individuals with suspected or confirmed anthrax disease.
The basic components of treatment for anthrax consist of hospitalization with intensive supportive
care and IV antibiotics. After obtaining appropriate cultures, antimicrobials should be started
immediately on suspicion and prior to confirmation of the diagnosis.
1
Patients with inhalational
anthrax who received antibiotics within 4.7 days of exposure had a 40% case fatality, compared to
a 75% case-fatality in those with treatment initiated after 4.7 days.
21
Because susceptibility data will be delayed, initial antibiotics must be chosen empirically.
Recommendations for initial empiric therapy of suspected or confirmed anthrax disease are
described below. Empiric therapy with at least two agents is recommended because of the
potential for infection with strains of B. anthracis engineered to be penicillin- and/or tetracycline-
resistant.
1
Antibiotic resistance to amoxicillin is of greater concern than resistance to doxycycline
or ciprofloxacin; therefore, amoxicillin is not recommended as a first-line agent unless the strain
has been proven susceptible. Therapy may be switched to oral antimicrobials when clinically
indicated. Therapy should be continued for a total duration of 60 days because spores can persist
and then germinate for prolonged periods. There is a possibility that spores could germinate and
cause illness up to 100 days after exposure.
1
Contained casualty setting: Parenteral antimicrobial therapy with at least two agents is
recommended for inhalational and GI anthrax when individual medical management is available.
After clinical improvement is noted, treatment can be switched to oral therapy with ciprofloxacin or
doxycycline, based on susceptibilities and clinical considerations. Draining of pleural effusions has
also been associated with reduced mortality.
21
Cutaneous anthrax can be treated with oral antibiotics. If in addition to cutaneous lesions there are
signs of systemic disease or extensive edema, or if lesions are present on the head or the neck,
then the multidrug IV regimen is recommended.
S. F Dept. Public Health – Infectious Disease Emergencies ANTHRAX, July 2008 Page 10/14
Anthrax meningitis can be treated using the inhalational anthrax guidelines; however, IV treatment
with a fluoroquinolone plus 1-2 antimicrobials with good central nervous system (CNS) penetration
ANTHRAX: TREATMENT AND POST-EXPOSURE PROPHYLAXIS RECOMMENDATIONS
A
I
NITIAL IV THERAPY
B,C
FOR
INHALATIONAL, GI ANTHRAX, OR
CUTANEOUS ANTHRAX WITH
COMPLICATIONS
D
I
NITIAL THERAPY FOR CUTANEOUS
ANTHRAX
B,D
T
HERAPY FOR ANTHRAX IN THE MASS
CASUALTY SETTING, OR POSTEXPOSURE
PROPHYLAXIS, OR AFTER CLINICAL
IMPROVEMENT ON IV THERAPY
E,F
Adult
Ciprofloxacin, 400 mg IV q12 hr or
Doxycycline
G
, 100 mg IV q12 hr
AND
One or two additional antimicrobials
(agents with in vitro activity include
rifampin, vancomycin, penicillin, ampicillin,
chloramphenicol, imipenem, clindamycin,
and clarithromycin)
H
Ciprofloxacin, 500 mg orally q12 hrs
for 60 days or
Doxycycline, 100 mg orally q12 hrs for
60 days
Ciprofloxacin
I
, 500 mg orally twice
daily for 60 days or
Doxycycline
J
, 100 mg orally twice daily
for 60 days
Children
Ciprofloxacin
K,L
, 10 mg/kg IV q12 hrs
(max 400 g/dose) or
Doxycycline:
G,L,M
>
45 kg, 100 mg IV q12 hr
<45 kg, give 2.2 mg/kg IV q12 hrs
(max 200 mg/day)
AND
One or two additional antimicrobials
(agents with in vitro activity include
rifampin, vancomycin, penicillin, ampicillin,
chloramphenicol, imipenem, clindamycin,
and clarithromycin)
H
Ciprofloxacin, 15 mg/kg orally q12 hrs
(max 500 mg/dose) for 60 days or
Doxycycline:
G,L,M
>45 kg, give 100 mg orally q12 hrs
for 60 days
<45 kg, give 2.2 mg/kg orally q12 hrs
(max 200 mg/day) for 60 days
Ciprofloxacin
I
, 15 mg/kg orally twice
daily (max 500 mg/dose) for 60 days or
Doxycycline
J
:
>
45 kg, give 100 mg orally twice daily
for 60 days
<45 kg, give 2.2 mg/kg orally twice
daily (max 200 mg/day) for 60 days
or
Amoxicillin
N
>20 kg: 500 mg orally three times
daily for 60 days
<20 kg: 80 mg/kg/day orally in
three divided doses every 8 hrs for
60 days
Pregnant
women
Same as for non-pregnant adults
O
Same as for non-pregnant adults
O
Same as for non-pregnant adults or
Amoxicillin
N
500 mg orally three times
daily for 60 days
Immuno-
compromised
Persons
Same as for non-immunocompromised
persons and children
Same as for non-immunocompromised
persons and children
Same as for non-immunocompromised
persons and children
A
The treatment recommendations included in this table are adapted from guidance developed during the 2001 anthrax outbreaks. Therapy
recommendations in other situations should be guided by antimicrobial susceptibility results.
1, 2, 6, 17
B
Ciprofloxacin or doxycycline should be considered an essential part of first-line therapy for inhalational anthrax.
C
Steroids may be considered an adjunct therapy for patients with severe edema and for meningitis based on experience with bacterial meningitis of
other etiologies.
D
Cutaneous anthrax cases with signs of systemic involvement, extensive edema, or lesions on the head or neck require intravenous therapy, and a
multidrug approach is recommended.
E
Initial therapy may be altered based on clinical course of patient; one or two antimicrobial agents (eg, ciprofloxacin or doxycycline) may be adequate as
patient improves.
F
If pharmaceutical resources permit in a mass casualty setting, therapy with at least two agents is recommended over monotherapy.
G
If meningitis is suspected, doxycycline may be less optimal because of poor central nervous system penetration.
H
Because of concerns of constitutive and inducible beta-lactamases in Bacillus anthracis isolates, penicillin and ampicillin should not be used alone.
Consultation with an infectious disease specialist is advised.
I
In vitro studies suggest that ofloxacin (400 mg orally every 12 hours) or levafloxacin, (500 mg orally every 24 hours) could be used in place of
ciprofloxacin – if supplies were limited in a mass casualty or post-exposure prophylaxis situation. FDA has approved levafloxacin for PEP in adults and
children.
J
In vitro studies suggest that 500 mg of tetracycline orally every 6 hours could be used in place of doxycycline – if supplies were limited in a mass
casualty or post-exposure prophylaxis situation.
K
If intravenous ciprofloxacin is not available, oral ciprofloxacin may be acceptable because it is rapidly and well absorbed from the gastrointestinal tract
with no substantial loss by first-pass metabolism. Maximum serum concentrations are attained 1-2 hours after oral dosing but may not be achieved if
vomiting or ileus is present.
L
Tetracycline and quinolone antibiotics are generally not recommended during pregnancy or childhood; however their use may be indicated for life-
threatening illness. Ciprofloxacin may be preferred in pregnant women and children up to 8 years of age because of the known adverse event profile of
doxycycline (e.g., tooth discoloration). Doxycycline may be preferred in children 8 years and older because of the adverse event profile of ciprofloxacin
(e.g., arthropathies).
M
American Academy of Pediatrics recommends treatment of young children with tetracyclines for serious infections (eg, Rocky Mountain spotted fever).
N
Amoxicillin is not approved by the FDA for post-exposure prophylaxis or treatment of anthrax. However, CDC has indicated that if the isolate is
determined to be susceptible to amoxicillin, it could be used for pregnant women and children for post-exposure prophylaxis or for completion of 60 days
antibiotic therapy after initial treatment with ciprofloxacin or doxycycline. Amoxicillin resistance to anthrax is of greater concern than that of doxycycline
or ciprofloxacin, and amoxicillin is not recommended as a first-line agent unless the isolate is proven to be susceptible.
O
Although tetracyclines are not recommended for pregnant women, their use may be indicated for life-threatening illness. Adverse effects on developing
teeth and bones are dose-related; therefore, doxycycline might be used for a short time (7-14 days) before 6 months of gestation.
S. F Dept. Public Health – Infectious Disease Emergencies ANTHRAX, July 2008 Page 11/14
and activity against B. anthracis (i.e., rifampin, vancomycin, penicillin, ampicillin, meropenem) is
recommended. The addition of corticosteroids may help manage cerebral edema.
23
Mass casualty setting: Use of oral antibiotics may be necessary if the number of patients
exceeds the medical care capacity for individual medical management. If pharmaceutical resources
permit, therapy with at least two agents is recommended over monotherapy.
Prophylaxis of Persons Exposed but Without Symptoms
Postexposure prophylaxis (PEP) is the administration of antibiotics, with or without vaccine, after
suspected exposure to anthrax has occurred but before symptoms are present. (If symptoms are
present, see section on treatment, above). In general, PEP is recommended for persons exposed to
an air space or package contaminated with B. anthracis. Unvaccinated laboratory workers exposed
to B. anthracis cultures should also receive PEP.
1
As there is no known person to person
transmission of inhalational anthrax, prophylaxis should not be offered to contacts of cases, unless
also exposed to the original source.
1
Postexposure prophylaxis of potential inhalational anthrax consists of oral administration of either
ciprofloxacin or doxycycline. Therapy should be continued for 60 days. Patients treated for
exposure should be informed of the importance of completing the full course of antibiotic
prophylaxis regardless of the absence of symptoms.
1, 22, 24
The Food and Drug Administration (FDA)
has also approved levafloxacin and penicillin G procaine for PEP of inhalational anthrax. .
25
And
levafloxacin was approved recently for children older than 6 months.
26
Because of concerns about
use of doxycycline or ciprofloxacin in children and about doxycycline use in pregnant women, the
CDC has indicated that for prophylaxis, therapy can be switched to amoxicillin in these groups if the
isolate is determined to be susceptible. Amoxicillin may also be considered for patients allergic to
both ciprofloxacin and doxycycline.
1, 22, 24
The Advisory Committee on Immunization Practices recommends the use of combined antimicrobial
prophylaxis and vaccine [Biothrax (formerly Anthrax vaccine absorbed, AVA)]. Biothrax is not
licensed for this use by the FDA, and would need to be given under an Investigational New Drug
(IND) application. The recommended regimen is three vaccine doses (given at 0, 2, and 4 weeks
after exposure) and at least a 30-day course of antimicrobial therapy. The CDC does not
recommend vaccination in pregnant women given lack of data.
27
Following the 2001 attacks, exposed persons were given the option of (1) 60 days of antibiotic
prophylaxis; (2) 100 days of antibiotic prophylaxis, and (3) 100 days of antibiotic prophylaxis, plus
anthrax vaccine (under IND protocol).
28
Anthrax Vaccine
The anthrax vaccine Biothrax (formerly Anthrax vaccine absorbed, AVA) is available but only in
limited supply that is controlled by federal authorities. It is an inactivated cell-free filtrate of an
avirulent strain of B. anthracis. Local reactions and mild systemic reactions are common. Severe
allergic reactions are rare (<1 per 100,000).
1, 27
S. F Dept. Public Health – Infectious Disease Emergencies ANTHRAX, July 2008 Page 12/14
These recommendations are current as of this document date. SFDPH will provide periodic updates as needed
and situational guidance in response to events (www.sfcdcp.org).
The anthrax vaccine is licensed for pre-exposure use to prevent cutaneous anthrax in healthy,
nonpregnant adults 18-65 years of age who have a high likelihood of coming into contact with
anthrax, including certain laboratory workers and animal processing workers. AVA is not currently
licensed for postexposure use and must be given in this context under an FDA investigational drug
protocol. The CDC may recommend its use for PEP under some circumstances. Research is
underway on new anthrax vaccines.
1, 27
Developmental Anthrax Therapeutics
Additional therapeutic candidates for treatment and prophylaxis of anthrax are currently under
development. The Department of Health and Human Services announced plans to purchase the
antibody-based therapeutic candidates immune globulin (AIG) and ABthrax™ (raxibacumab) for the
strategic national stockpile. These therapeutic approaches use antibodies to neutralize anthrax
toxin. Neither AIG nor ABthrax™ is FDA approved, but either may be authorized for use as an
investigational new drug in an emergency. Studies are still in progress to determine efficacy of
these therapeutics in anthrax treatment and prophylaxis.
6
COMPLICATIONS AND ADMISSION CRITERIA
Without early antibiotic treatment, inhalational anthrax progresses to pneumonitis marked by
severe respiratory distress and cyanosis, and is often accompanied by pleural effusion. Patients
with anthrax pneumonitis are particularly likely to develop septicemia and septic shock due to
hematogenous dissemination of the bacteria. Sepsis may also develop as a complication of
cutaneous anthrax or gastrointestinal anthrax. Anthrax meningitis may occur as a consequence of
hematogenous dissemination.
Patients with suspected or confirmed inhalational, gastrointestinal, or meningeal anthrax, as well as
those with cutaneous anthrax who exhibit head or neck lesions, extensive edema, or systemic signs
of illness, require admission for intravenous antibiotic therapy and supportive care.
INFECTION CONTROL
Clinicians should notify local public health authorities, their institution’s infection control
professional, and their laboratory of any suspected anthrax cases. Public health authorities may
conduct epidemiologic investigations and implement disease control interventions to protect the
public. Both HICPAC (Hospital Infection Control Practices Advisory Committee) of the CDC and the
Working Group for Civilian Biodefense recommend Standard Precautions for anthrax patients in a
hospital setting without the need for isolation. Person to person transmission has only rarely been
reported for patients with cutaneous anthrax and Standard Precautions are considered adequate.
Routine laboratory procedures should be carried out under Biosafety Level 2 (BSL-2) conditions.
1, 29
S. F Dept. Public Health – Infectious Disease Emergencies ANTHRAX, July 2008 Page 13/14
Decontamination
Contaminated surfaces can be disinfected with commercially available bleach or a 1:10 dilution of
household bleach and water. All persons exposed to an aerosol containing B. anthracis should be
instructed to wash body surfaces and clothing with soap and water.
1
PEARLS AND PITFALLS
The initial (prodromal) phase of inhalational anthrax resembles an influenza-like syndrome and can
be difficult to distinguish from seasonal respiratory illnesses. Nasal congestion and rhinorrhea,
however, are common features of seasonal influenza-like syndromes and are unusual with
pulmonary anthrax.
The classic radiographic findings of inhalational anthrax – CXR showing a widened mediastinum
(due to hilar lymphadenopathy) and pulmonary effusion – although not unique to anthrax, should
nonetheless prompt a high level of clinical suspicion.
The necrotic, edematous, eschar-covered skin lesion of cutaneous anthrax is usually painless,
which is an important differentiating feature from a brown recluse spider bite.
REFERENCES
1. Inglesby TV, O'Toole T, Henderson DA, et al. Anthrax as a biological weapon, 2002: updated
recommendations for management. Jama. May 1 2002;287(17):2236-2252.
2. Lucey D. Chapter 205 - Bacillus anthracis (Anthrax). In: Mandell GL, Bennett JE, Dolin R, eds.
Principles and practice of infectious diseases, Ed 6. New York, NY: Churchill Livingstone; 2005.
3. Lucey D. Chapter 324 - Anthrax. In: Mandell GL, Bennett JE, Dolin R, eds. Principles and
practice of infectious diseases, Ed 6. New York, NY: Churchill Livingstone; 2005.
4. Abramova FA, Grinberg LM, Yampolskaya OV, Walker DH. Pathology of inhalational anthrax in
42 cases from the Sverdlovsk outbreak of 1979. Proc Natl Acad Sci U S A. Mar 15
1993;90(6):2291-2294.
5. Dixon TC, Meselson M, Guillemin J, Hanna PC. Anthrax. N Engl J Med. Sep 9
1999;341(11):815-826.
6. CIDRAP. Anthrax: Current, comprehensive information on pathogenesis, microbiology,
epidemiology, diagnosis, treatment, and prophylaxis. Center for Infectious Disease Resarch
and Policy, University of Minnesota. Available at:
http://www.cidrap.umn.edu/cidrap/content/bt/anthrax/biofacts/anthraxfactsheet.html
.
7. CDC. Inhalation Anthrax Associated with Dried Animal Hides --- Pennsylvania and New York
City, 2006. MMWR. 2006;55(10):280-282.
8. CDC. Anthrax Q & A: Anthrax and animal hides. Available at:
http://www.bt.cdc.gov/agent/anthrax/faq/pelt.asp
.
9. CDC. Suspected cutaneous anthrax in a laboratory worker—Texas 2002. MMWR.
2002;51(13):279-281.
10. CDC. Summary of notifiable diseases—United States,
2002. MMWR. 2004;51(53):37.
S. F Dept. Public Health – Infectious Disease Emergencies ANTHRAX, July 2008 Page 14/14
11. Bales ME, Dannenberg AL, Brachman PS, Kaufmann AF, Klatsky PC, Ashford DA. Epidemiologic
response to anthrax outbreaks: field investigations, 1950-2001. Emerg Infect Dis. Oct
2002;8(10):1163-1174.
12. CDPH. California Monthly Summary Report Selected Reportable Diseases. Division of
Communicable Disease Control, California Department of Publid Health. Available at:
http://www.cdph.ca.gov/data/statistics/Pages/CD_Tables.aspx
.
13. SFDPH. San Francisco Communicable Disease Report, 1986-2003. San Francisco Department
of Public Health. May, 2005. Available at: http://www.sfcdcp.org/publications
.
14. SFDPH. Annual Report of Communicable Diseases in San Francisco, 2004-2005. San Francisco
Department of Public Health. August, 2006. Available at: http://www.sfcdcp.org/publications
.
15. SFDPH. Annual Report of Communicable Diseases in San Francisco, 2006. San Francisco
Department of Public Health. January 2008. Available at: http://www.sfcdcp.org/publications
.
16. American Public Health Association. Anthrax. In: Chin J, ed. Control of Communicable Diseases
Manual. 17 ed. Washington, DC: American Public Health Association; 2000:20-25.
17. Jernigan JA, Stephens DS, Ashford DA, et al. Bioterrorism-related inhalational anthrax: the first
10 cases reported in the United States. Emerg Infect Dis. Nov-Dec 2001;7(6):933-944.
18. Freedman A, Afonja O, Chang MW, et al. Cutaneous anthrax associated with microangiopathic
hemolytic anemia and coagulopathy in a 7-month-old infant. Jama. Feb 20 2002;287(7):869-
874.
19. Kadanali A, Tasyaran MA, Kadanali S. Anthrax during pregnancy: case reports and review. Clin
Infect Dis. May 15 2003;36(10):1343-1346.
20. Howell JM, Mayer TA, Hanfling D, et al. Screening for inhalational anthrax due to bioterrorism:
evaluating proposed screening protocols. Clin Infect Dis. Dec 15 2004;39(12):1842-1847.
21. Holty JE, Bravata DM, Liu H, Olshen RA, McDonald KM, Owens DK. Systematic review: a
century of inhalational anthrax cases from 1900 to 2005. Ann Intern Med. Feb 21
2006;144(4):270-280.
22. Bell DM, Kozarsky PE, Stephens DS. Clinical issues in the prophylaxis, diagnosis, and treatment
of anthrax. Emerg Infect Dis. Feb 2002;8(2):222-225.
23. Sejvar JJ, Tenover FC, Stephens DS. Management of anthrax meningitis. Lancet Infect Dis.
May 2005;5(5):287-295.
24. Swartz MN. Recognition and management of anthrax--an update. N Engl J Med. Nov 29
2001;345(22):1621-1626.
25. Meyerhoff A, Murphy D. Guidelines for treatment of anthrax. Jama. Oct 16
2002;288(15):1848-1849; author reply 1848-1849.
26. FDA. Levaquin (levofloxacin) supplemental NDA approval. May 5, 2008. Available at:
http://www.fda.gov/cder/foi/appletter/2008/020634se5-047020635se5-051021721se5-
015ltr.pdf.
27. CDC. Use of anthrax vaccine in response to terrorism: supplemental recommendations of the
Advisory Committee on Immunization Practices. MMWR. 2002;51(45):1024-1026.
28. CDC. Additional options for preventive treatment for persons exposed to inhalational anthrax.
MMWR. 2001;50(50):1142.
29. Siegel JD, Rhinehart E, Jackson M, Chiarello L, HICPAC. Guideline for Isolation Precautions:
Preventing Transmission of Infectious Agents in Healthcare Settings, 2007. Centers for Disease
Control and Prevention. Available at: http://www.cdc.gov/ncidod/dhqp/gl_isolation.html
.
S.F.Dept. Public Health – Infectious Disease Emergencies AVIAN INFLUENZA, August 2005 Page 1/9
Outline
Agent
Epidemiology
Clinical Features
Surveillance and Diagnosis
Treatment and Prophylaxis
Infection Control
References
Influenza A is not a reportable condition under
Calif. law. However, health care providers are
required to report any UNUSUAL disease to the
local health department within one hour.
In the event of Avian Influenza outbreak, SFDPH
will issue guidelines for case identification,
infection control, and disease reporting, at
www.sfdph.org/cdcp.
SFDPH communicable disease control may be
contacted by phone at 415-554-2830.
AGENT
Influenza virus belongs to the Orthomyxovirus family and contains 8 different segments of
negative-stranded RNA. There are 3 types: A, B, and C, distinguishable by internal virus proteins.
Influenza A is responsible for most human influenza disease, causes avian influenza, and is the
source of all past influenza pandemics in humans. Influenza B is a disease of humans only, while
inluenza C causes milder illness in both humans and swine and occurs uncommonly.
Influenza A is subtyped based on viral envelope glycoproteins hemagglutinin (HA) and
neuraminidase (NA). There are 16 different HA antigens (H1 to H16) and 9 different NA antigens
(N1 to N9) for influenza A. Human disease has historically been related to 3 subtypes of HA (H1,
H2, and H3) and 2 subtypes of NA (N1 and N2).
Influenza A infects humans, birds, pigs, horses, whales, seals, and has recently been recognized in
felines. Avian influenza A can infect a variety of domestic and wild bird species. Avian influenza in
domestic chickens and turkeys is classified according to disease severity, with two recognized
forms: highly pathogenic avian influenza (HPAI), and low-pathogenic avian influenza (LPAI). Avian
influenza viruses that cause HPAI are highly virulent and mortality rates in infected flocks often
approach 100%. All known subtypes of influenza A can be found in birds, but only subtypes H5
and H7 have caused HPAI outbreaks.
AVIAN INFLUENZA AUGUST 2005
S.F.Dept. Public Health – Infectious Disease Emergencies AVIAN INFLUENZA, August 2005 Page 2/9
EPIDEMIOLOGY
Influenza Pandemics
Pandemics differ from seasonal outbreaks or “epidemics” of influenza, which are caused by
subtypes of influenza viruses that already exist among people. A pandemic is a global outbreak
that occurs when a new, highly pathogenic strain of influenza type A virus emerges in the human
population and spreads easily from person-to-person worldwide, aided by the lack of human
immunity to the novel strain.
Past influenza pandemics have led to high levels of illness, death, social disruption, and economic
loss. There were 3 influenza A pandemics during the 20
th
century:
1918-19, "Spanish flu," (H1N1), caused >500,000 deaths in the USA and >50,000,000 deaths
worldwide. Nearly half of those who died were young, healthy adults.
1957-58, "Asian flu," (H2N2), first identified in China in early 1957, it caused about 70,000
deaths in the USA by June 1957.
1968-69, "Hong Kong flu," (H3N2), caused about 34,000 deaths in the United States.
Influenza A (H3N2) viruses still circulate today.
Influenza in Bird Populations
All birds are believed susceptible to infection with avian influenza. Migratory waterfowl – most
notably wild ducks – are the natural reservoir of avian influenza viruses, however domestic poultry,
including chickens and turkeys, are particularly susceptible to epidemics of rapidly fatal influenza.
Recent research has shown that viruses of low pathogenicity can quickly mutate into highly
pathogenic viruses. For example, during a 1999–2001 avian influenza epidemic in Italy, the H7N1
virus, initially of low pathogenicity, mutated within 9 months to a highly pathogenic form. More
than 13 million birds died or were destroyed.
Standard control measures aimed at preventing spread of HPAI in a country’s poultry population
include quarantining of infected farms and destruction of infected or potentially exposed flocks.
In the absence of prompt control measures backed by good surveillance, epidemics can last for
years. For example, an epidemic of H5N2 avian influenza, which began in Mexico in 1992, started
with low pathogenicity, evolved to the highly fatal form, and was not controlled until 1995.
Mechanism of Transmission to Humans
Influenza A viruses are genetically labile and well adapted to elude host defenses. Influenza
viruses lack mechanisms for the “proofreading” and repair of errors that occur during replication.
As a result of these uncorrected errors, the genetic composition of a virus changes during passage
through humans and animals, and the existing strain is replaced with a new antigenic variant.
These changes in the antigenic composition of influenza A viruses are known as antigenic drift
.
S.F.Dept. Public Health – Infectious Disease Emergencies AVIAN INFLUENZA, August 2005 Page 3/9
Influenza A viruses, including subtypes from different species, can also swap or reassort genetic
materials. This process -- known as antigenic shift – creates a novel virus subtype that differs
genetically from both parent viruses. As populations will have no immunity to the new subtype,
and as no existing vaccines can confer protection, antigenic shift has historically resulted in highly
lethal pandemics. For this to happen, a subtype of avian influenza needs to acquire genes from
human influenza viruses that enable person-to-person transmission.
Conditions favorable for the emergence of antigenic shift are thought to involve humans living in
close proximity to domestic poultry and pigs. Because pigs are susceptible to infection with both
avian and mammalian viruses, including human strains, they can serve as a “mixing vessel” for the
scrambling of genetic material from human and avian viruses, resulting in the emergence of a
novel subtype. In addition, evidence is mounting that, for at least some avian influenza virus
subtypes circulating in bird populations, humans themselves can serve as the “mixing vessel”.
The Current H5N1 Threat
Of the avian influenza subtypes, currently the H5N1 subtype is of greatest pandemic concern for
the following reasons:
Rapid spread throughout poultry flocks in Asia; now appears to be endemic in eastern Asia
Mutates rapidly
Propensity to acquire genes from viruses infecting other animal species
Causes severe disease in humans, with a high case-fatality rate (approx. 70%)
There is ongoing exposure and infection of humans in rural Asia, where many households
keep free-ranging poultry flocks for income and food
The first documented infection of humans with an avian influenza virus occurred in Hong Kong in
1997, when the H5N1 strain caused severe respiratory disease in 18 humans, of whom 6 died. The
infection of humans coincided with an epidemic of HPAI, caused by the same strain, in Hong Kong’s
poultry population. Close contact with live infected poultry was the source of human infection, and
the virus was shown to have jumped directly from birds to humans. Transmission to health care
workers occurred, but did not cause severe disease. Rapid destruction of Hong Kong’s entire
poultry population, estimated at around 1.5 million birds, reduced opportunities for further direct
transmission to humans, and may have averted a pandemic.
Alarms have continued to mount since 2003, when an outbreak of HPAI caused by the H5N1 strain
spread rapidly through poultry farms in southeastern Asia. Areas currently affected by H5N1 avian
influenza in poultry include Cambodia, China (both Taiwan and the People’s Republic of China),
Hong Kong, Indonesia, Japan, Laos, Malaysia, Philippines, South Korea, Thailand, and Vietnam.
Over 140 million chickens have been slaughtered to halt spread of the virus.
The strain circulating in Asia appears highly pathogenic for humans, and immunity in the human
population is generally lacking. If H5N1 continues to circulate widely among poultry, the potential
S.F.Dept. Public Health – Infectious Disease Emergencies AVIAN INFLUENZA, August 2005 Page 4/9
for emergence of a pandemic strain remains high. Human cases of H5N1 have been reported
officially in Vietnam, Thailand, and Cambodia. Between December 26, 2003 and June 28, 2005,
the WHO has tallied 108 laboratory-confirmed cases of H5N1 influenza in humans (54 of them
fatal). Probable person-to-person transmission was identified in Thailand involving transmission
from an ill child to her mother and aunt. However, the strain has not yet been shown to be easily
transmitted between humans, and sustained person-to-person transmission has not occurred.
CLINICAL FEATURES
Typical clinical features of influenza A are shown in the following table.
In an outbreak of avian influenza among humans, the clinical picture of primary viral pneumonia
may predominate. However given that the virus responsible for human-to-human transmission will
be a novel strain, the specifics of its clinical presentation will not be known until the outbreak
actually occurs. A recent report of avian influenza A (H5N1) in 10 patients in Vietnam
demonstrated the following clinical features of the illness:
Incubation period was 2-4 days (mean 3 days)
10/10 presented with fever, shortness of breath, and cough
INFLUENZA A: CLINICAL FEAURES
Incubation Period
1-2 days
Signs & Symptoms
Initially, systemic symptoms predominate
Fever (duration 3 days, range 2-8 days)
Chills, malaise, headache, myalgias, arthralgias, anorexia, toxic appearance,
occasionally prostration
Dry cough, pharyngeal pain, rhinorrhea
Convalescent period lasts 1-3 weeks
Cough, lassitude, and malaise
Complications
Primary influenza pneumonia; rapid progression of fever, cough, dyspnea, cyanosis
Secondary bacterial pneumonia
Croup
Exacerbation of COPD
Myositis; myoglobinuria
Myocarditis; pericarditis
Toxic shock syndrome
Encephalitis (rare)
Laboratory
Findings
WBC often normal (unless secondary bacterial process: elevated with left shift)
Sputum gram stain unremarkable (unless secondary bacterial pneumonia)
CXR usually normal in uncomplicated influenza; usually shows infiltrates and/or
consolidation when pneumonia present
S.F.Dept. Public Health – Infectious Disease Emergencies AVIAN INFLUENZA, August 2005 Page 5/9
If you consider testing for Avian Influenza,
you should:
IMMEDIATELY notify SFDPH
Communicable Disease Control (24/7 Tel:
415-554-2830) to facilitate testing and
initiate the public health response. Testing
for H5N1 subtype of influenza A occurs as
specialized labs and requires SFDPH
authorization.
Inform your laboratory that Avian Influenza
is under suspicion, so that they may follow
the appropriate biosafety procedures.
5/10 reported sputum production; in 3 of these, sputum was blood-tinged.
7/10 reported diarrhea.
None complained of sore throat, conjunctivitis, rash, or a runny nose.
10/10 had abnormal CXR at the time of hospital admission (including extensive bilateral
infiltration, lobar collapse, focal consolidation, and air bronchograms).
10/10 had lymphopenia, and 9/10 had thrombocytopenia at presentation
10/10 received broad-spectrum antibiotics
5/10 were treated with oseltamivir (4 of whom died)
8/10 died
A recent case report of a 4-year-old Vietnamese child with H5N1 avian influenza who presented in
2004 with encephalitis demonstrated the following features:
The child presented with a 2-day history of fever, headache, vomiting, and severe diarrhea
Laboratory tests on admission were unremarkable and chest x-ray was normal.
On day 3, the child had a generalized convulsion and became comatose. He developed
respiratory failure and died on day 5.
H5N1 influenza A virus was isolated from CSF, fecal, throat, and serum specimens.
Acute encephalitis was reported as the cause of death
SURVEILLANCE AND DIAGNOSIS
As of this writing, CDC recommendations issued
February 2004 (and re-affirmed February,
2005) for enhanced surveillance of patients at
risk for avian influenza are still in effect.
These are:
1) Testing for influenza A(H5N1) in the USA is
indicated for hospitalized patients with:
Radiographically confirmed pneumonia,
acute respiratory distress syndrome
(ARDS), or other severe respiratory
illness for which an alternate diagnosis
has not been established, AND
History of travel within 10 days of
symptom onset to a country with documented H5N1 avian influenza in poultry and/or
humans. (List of H5N1-affected countries available at www.who.int/topics/avian_influenza)
2) Testing for influenza A(H5N1) should be considered on a case-by-case basis in consultation with
the local health department for hospitalized or ambulatory patients with:
Documented temperature of >38°C (>100.4°F), AND
S.F.Dept. Public Health – Infectious Disease Emergencies AVIAN INFLUENZA, August 2005 Page 6/9
Detailed guidelines for Avian Influenza treatment/prophylaxis have not yet been issued. For
updates and situational guidance in response to events, check www.sfdph.org/cdcp.
At least one: cough, sore throat, shortness of breath, AND
History of contact with domestic poultry (e.g., visited a poultry farm, household raising
poultry, or bird market) or a known or suspected human case of influenza A(H5N1) in an
H5N1-affected country within 10 days of symptom onset.
Clinical specimens from suspect influenza A(H5N1) cases may be tested by PCR assays under strict
biosafety precautions at public health reference laboratories. Virus isolation studies carry higher
risks of inadvertent transmission and require even more stringent precautions.
TREATMENT AND PROPHYLAXIS
Antiviral Agents
There are 2 key uncertainties that challenge planning for administration of antiviral agents in the
event of an avian influenza outbreak among humans. First, it is unclear how much antiviral drug
will be available in the event of a large-scale outbreak. Second, the influenza strain responsible for
the outbreak and its profile of antibiotic resistance may not be fully known in advance.
There are 2 classes of antiviral agents for influenza: adamantanes (amantadine and rimantadine),
and neuraminidase inhibitors (zanamivir and oseltamivir). The drugs differ in cost, routes of
administration, adverse events, contraindications, and potential for antiviral resistance.
CHARACTERISTICS OF ANTI-INFLUENZA ANTIVIRAL AGENTS
Adamantane Derivatives Neuraminidase Inhibitors
Amantadine Rimantadine Oseltamivir Zanamivir
Route
Oral
Oral
Oral
Inhalation
Treatment
License
>1 year old
>1 year old
>13 years old
Not FDA Approved
Prophylaxis
License
>1 year old
Adults
>1 year old
>7 years old
Selected
Adverse
Events
CNS (dizziness,
insomnia, seizures,
suicidality); GI
(nausea); some
reports cardiac
toxicity
CNS (e.g. insomnia,
dizziness), GI (e.g.
nausea, vomiting)
GI (principally
nausea, vomiting)
Poss. bronchospasm
and decrease in lung
function, esp. in
patients with
underlying airway
disease
Adapted from: DHHS Pandemic Influenza Response & Preparedness Plan, Aug. 26, 2004
S.F.Dept. Public Health – Infectious Disease Emergencies AVIAN INFLUENZA, August 2005 Page 7/9
These recommendations are current as of this document date. SFDPH will provide periodic
updates as needed and situational guidance in response to events (www.sfdph.org/cdcp).
Both classes of drugs reduce duration of uncomplicated influenza when started within 2 days of
illness onset. However, there are no controlled studies of patients infected with influenza A(H5N1).
Vaccine Development
Influenza vaccine must be both subtype- and strain-specific. Candidate vaccines against H5N1
subtype were developed during 2003 for protection against the strain that was isolated from
humans in Hong Kong in February of that year. However, the current strain is different. Clinical
trials of additional candidate H5N1 vaccines are currently under way. However, it is not clear if
prototype H5 vaccines will offer protection against an emergent pandemic strain, and WHO has
indicated that 4-6 months (minimum) would be needed to develop a vaccine against a novel strain.
INFECTION CONTROL
*
Poultry Workers
Birds that are infected with avian influenza viruses can shed virus in saliva, nasal secretions, and
feces. Activities that could result in exposure to avian influenza-infected poultry include
euthanasia, carcass disposal, and cleaning and disinfection of premises affected by avian influenza.
These activities are unlikely to occur in an urban area such as San Francisco. However, the CDC
has written interim guidance for protection of persons involved in control of avian influenza
outbreaks among poultry in the USA (www.cdc.gov/flu/avian/professional/protect-guid.htm).
Health Care Providers
Human influenza is transmitted primarily via large respiratory droplets, and isolation precautions
for typical human influenza include Standard plus Droplet Precautions. However, the CDC has
recommended additional precautions for healthcare workers involved in the care of patients with
documented or suspected avian influenza
, for the following reasons: 1) higher risk of serious
disease and increased mortality from HPAI; 2) each human infection represents an important
opportunity for avian influenza to further adapt to humans and gain the ability to transmit more
easily among people; and 3) any opportunities for human-to-human transmission of avian
influenza may increase opportunities for genetic reassortment and possible emergence of a
pandemic strain.
*
For description of Precautions, see Chapter on Infection Control
S.F.Dept. Public Health – Infectious Disease Emergencies AVIAN INFLUENZA, August 2005 Page 8/9
The most recent (May, 2004) CDC recommendations state:
All patients who present to a health-care setting with fever and respiratory symptoms
should be managed according to recommendations for respiratory hygiene and cough
etiquette (www.cdc.gov/flu/professionals/infectioncontrol) and questioned regarding their
recent travel history.
Patients with a history of travel within 10 days to a country with avian influenza activity
and who are hospitalized with a severe febrile respiratory illness, or are otherwise under
evaluation for avian influenza, should be managed using Standard plus Contact plus
Airborne Precautions. In addition, Eye Protection should be utilized when within 3
feet of the patient. These precautions should be continued for 14 days after onset of
symptoms or until either an alternative diagnosis is established or diagnostic test results
indicate that the patient is not infected with influenza A virus.
Patients managed as outpatients or hospitalized patients discharged before 14 days with
suspected avian influenza should be isolated in the home setting, following CDC guidelines
for home isolation of SARS patients (www.cdc.gov/ncidod/sars/guidance/i/pdf/i.pdf).
CDC guidance also recommends that healthcare workers who may come into contact with the H5N1
virus or with infected patients should be vaccinated with the most recent seasonal influenza
vaccine. Although this will not protect against H5N1 influenza A, it will help avoid simultaneous
infection with other influenza strains and may thereby decrease the risk of genetic reassortment.
REFERENCES
CDC. Interim Guidance for Protection of Persons Involved in U.S. Avian Influenza Outbreak Disease
Control and Eradication Activities. Feb. 17, 2004. (www.cdc.gov/flu/avian/professional/protect-
guid.htm)
CDC. Update on Influenza A(H5N1) and SARS: Interim Recommendations for Enhanced U.S.
Surveillance, Testing, and Infection Controls. February 3, 2004.
(www.cdc.gov/flu/avian/professional/han020302.htm)
CDC. Interim Recommendations for Infection Control in Health-Care Facilities Caring for Patients
with Known or Suspected Avian Influenza. May 21, 2004.
(www.cdc.gov/flu/avian/professional/infect-control.htm)
CIDRAP. Pandemic Influenza. July 1, 2005. (www.cidrap.umn.edu/cidrap/content/influenza)
CIDRAP. Avian Influenza (Bird Flu): Implications for Human Disease. July 13, 2005.
(www.cidrap.umn.edu/cidrap/content/influenza)
S.F.Dept. Public Health – Infectious Disease Emergencies AVIAN INFLUENZA, August 2005 Page 9/9
De Jong MD et al. Fatal avian influenza A (H5N1) in a child presenting with diarrhea followed by
coma. N Engl J Med 2005;352:686-91.
DHHS: Department of Health and Human Services Pandemic Influenza Response and
Preparedness Plan. August 26, 2004. (www.dhhs.gov/nvpo/pandemicplan)
Hien TT et al. Avian influenza (H5N1) in 10 patients in Vietnam. NEJM 2004;350(12):1179-88.
Treanor, JJ. Influenza Virus. In: Principles and Practice of Infectious Diseases, 6th edition; Gerald
Mandell et al, Eds. Elsevier, 2005.
WHO. Global Influenza Preparedness Plan 2005 (WHO/CDS/CSR/GIP/2005.5). Available at:
http://www.who.int/csr/disease/influenza/inforesources/en/
WHO. Avian influenza ("bird flu") and the significance of its transmission to humans. 15 January
2004. (www.who.int/mediacentre/factsheets/avian_influenza)
WHO. International conference draws up strategy to fight avian influenza. 06 July 2005
(www.wpro.who.int/health_topics/avian_influenza/)
WHO. Cumulative number of confirmed human cases of avian influenza A(H5N1) reported to WHO.
28 June 2005.
S.F. Dept Public Health – Infectious Disease Emergencies BOTULISM, July 2008 Page 1/11
Outline
Introduction
Epidemiology
Clinical Features
Differential Diagnosis
Laboratory Diagnosis
Treatment and Prophylaxis
Complications and Admission Criteria
Infection Control
Pearls and Pitfalls
References
Immediately report any suspected or
confirmed cases of botulism to:
SFDPH Communicable Disease Control
(24/7 Tel: 415-554-2830)
- By law, health care providers must report
suspected or confirmed cases of botulism to their
local health department immediately [within 1 hr].
- SFDPH Communicable Disease Control can
facilitate specialized testing and will initiate the
public health response as needed.
Also notify your:
Infection Control Professional
Clinical Laboratory
INTRODUCTION
Botulism is a disease caused by exposure to botulinum toxin produced from Clostridium species,
mainly Clostridium botulinum. Clinical forms of the disease include foodborne, inhalational, wound,
infant, adult intestinal toxemia, and iatrogenic. C. botulinum is a gram-positive, strictly anaerobic,
spore-forming bacillus naturally found in soil and aquatic sediments. There are seven types of the
toxin based on antigenic differences, labeled A through G. Types A, B, and E (and rarely, F) are
pathogenic in humans. Types C, D, and E cause illness in other mammals, birds, and fish.
Botulinum toxin lacks color, odor, and taste and is the most lethal toxin known. Death is caused
by doses of less than 1 µg. Antibiotics have no activity against the toxin itself.
1-3
In response to unfavorable environmental conditions (changes in pH, temperature, and water or
nutrient availability), C. botulinum bacteria sporulate. C. botulinum spores are hardy, resistant to
dessication, heat, ultraviolet (UV) light, and alcohols, and can survive boiling for up to 4 hours;
however, they are readily killed by chlorine-based disinfectants. Once spores encounter more
favorable conditions, such as are found in contact with human tissues, they germinate, thereby
producing growing cells that are capable of reproducing and elaborating toxin. .
1-3
The Working Group for Civilian Biodefense considers botulism to be a dangerous potential biological
weapon because of the pathogen’s “extreme potency and lethality; its ease of production,
transport, and misuse; and the need for prolonged intensive care among affected persons.” Use of
botulism as a biological weapon is expected to produce severe medical and public health
outcomes.
3-6
BOTULISM
S.F. Dept Public Health – Infectious Disease Emergencies BOTULISM, July 2008 Page 2/11
EPIDEMIOLOGY
Botulinum Toxin as a Biological Weapon
State-sponsored military programs have researched and weaponized botulinum toxin dating back
to the 1930s. Botulism has also been used as a weapon by a terrorist group. Unfortunately,
botulism is ubiquitous in nature and therefore access to it cannot be easily controlled.
3, 4
Likely modes of dissemination for toxin used as a weapon include:
3-6
Contamination of food or beverages. Possible food or beverage vehicles for botulism
toxin are those that are not heated at 85°C (185°F) for 5 minutes before consumption or
those that are contaminated after appropriate heating. Typical pasteurization does not
remove all toxin.
Dispersion of aerosolized toxin. Animal studies and rare cases of laboratory accidents
have confirmed the pathogenicity of aerosolized toxin. One study estimates that
aerosolizing 1 g of botulinum toxin could kill up to 1.5 million people; while another
estimates that a point source exposure could kill 10% of the population 500 meters
downwind. Technical factors make such dissemination difficult.
Contamination of a water supply. This is a possibility, but not likely because of the
quantity of toxin needed to effectively contaminate a water supply. Additionally, standard
drinking water treatment inactivates the toxin quickly and, in fresh water, it is inactivated
through natural mechanisms in 3 to 6 days.
An intentional release of botulinum toxin would have the following characteristics:
3, 4, 6
Clustering in time: multiple similarly presenting cases of rapidly progressing acute flaccid
symmetric paralysis with prominent bulbar palsies, generally 12 to 36 hours after release
Atypical host characteristics: cases of unusual botulinum toxin type (C, D, F, G, and possibly
E) or cases without typical gastrointestinal symptoms of nausea, vomitting, and diarrhea
Unusual geographic clustering: cases in geographic proximity during the week before
symptom onset, but lack common food exposure (aerosol exposure) or toxin type outside of
typical geographic range
Absent risk factors: multiple outbreaks without an association with a common food source
Naturally Occurring Botulism
Reservoirs
The sporulated form of the bacterium is commonly found in soils and aquatic sediments. Cistern
water, dust and foods, including honey, can become contaminated from contact with the soil.
1, 2, 7
Mode of Transmission
Botulism is caused by exposure to botulinum toxin. Humans can become infected in a number of
ways:
Inhalation of toxin (inhalational)
S.F. Dept Public Health – Infectious Disease Emergencies BOTULISM, July 2008 Page 3/11
Consumption of toxin (foodborne)
Consumption of C. botulinum spores (infant; adult intestinal toxemia)
Contamination of a tissue with C. botulinum spores (wound)
Contamination of a tissue with toxin (iatrogenic)
Worldwide occurrence
In the late 1700s, botulism emerged as a disease because of changes in sausage production in
Europe. In fact, botulus means sausage. Soon thereafter in the early 1800s, botulism associated
with consumption of fermented fish was recognized in Russia. Wound and infant botulism were
discovered much later in the mid to late 1900s. In 1999-2000, more than 2500 cases of foodborne
botulism were reported in Europe. The highest incidence is found in countries of the former Soviet
Union and in Asia and is related to improper food handling. Type B is more common in Europe,
whereas type E is more common in Scandinavia and Canada and is frequently linked to improper
storage of fish and marine mammals.
1, 8
United States Occurrence
In the United States, naturally occurring botulism is a rare disease with an annual incidence of
approximately 100 cases (infant: 71; food: 24; and wound: 3).
9
More than half of foodborne cases
occur in the Western states of California, Oregon, Washington, Alaska, and Colorado.
9
Type E is
more common among Alaskan natives because of their diet of fermented meat from aquatic
mammals and fish.
10
Type A is found mainly in Western states and type B is more common in the
East.
1
Most cases of wound botulism result from injection drug use with black tar heroin, which is
more common in the Western states.
11
Occurrence in California and San Francisco
From 1994 to 2006, 44 cases of foodborne botulism were reported in California, and one of these
occurred in San Francisco.
12-15
CLINICAL FEATURES
Regardless of the route of intoxication the same clinical neurologic syndrome develops.
1-3, 16
Botulism is an afebrile descending symmetric paralytic illness. Disease generally begins with
absorption of toxin by mucosal surfaces in the gastrointestinal system, the eye or nonintact skin.
Cranial nerve dysfunction ensues followed by muscle weakness beginning with the proximal muscle
groups. Severity of disease is variable, ranging from mild cranial nerve dysfunction to flaccid
paralysis. Both the severity of disease and the rapidity of onset correlate with the amount of toxin
absorbed into the circulation.
3, 6
Botulinum toxin blocks acetylcholine release at the neuromuscular junction of skeletal muscle
neurons and peripheral muscarinic cholinergic autonomic synapses. It binds irreversibly to
presynaptic receptors to inhibit the release of acetylcholine and cause neuromuscular weakness
and autonomic dysfunction. The effect lasts weeks to months, until the synapses and axonal
branches regenerate. Death from botulism results acutely from airway obstruction or paralysis of
respiratory muscles. .
1-3
S.F. Dept Public Health – Infectious Disease Emergencies BOTULISM, July 2008 Page 4/11
The case fatality rate was close to 60% prior to the advent of critical care. Even today, the
mortality rate is high if treatment is not immediate and proper. In an outbreak setting, the
mortality rate for the first case is 25 % and for all other cases is 4%. A shorter incubation period
has been linked to higher mortality, possibly reflecting a dose-dependent response. Fatality
doubles in persons above the age of 60.
1-3
Food-borne botulism occurs from the consumption of preformed botulinum toxin in food.
Waterborne botulism has not been seen. Toxin types A, B, and E account for most cases of
foodborne botulism. Minute amounts of toxin can cause disease. A case in which a
contaminated potato was spit out before being swallowed, resulted in 6 months of
hospitalization.
In order for foodborne botulism to occur:
1, 8
o C. botulinum spores must contaminate the food
o anaerobic, nonacidic, low sugar and salt, and warm conditions must be met during
the food preservation so that the spores can survive, germinate and produce toxin
o the food must not be reheated sufficiently to inactivate the heat-labile toxin before
the food is consumed (>85°C for 5 minutes).
Inhalational botulism does not occur in nature; however three human cases occurred in
1962 in lab technicians working with aerosolized botulinum toxin. It has also been produced
experimentally in laboratory animals.
Wound botulism is caused by toxin absorbed into the circulation through a wound. Most
cases are related to injection drug use, especially in association with use of black tar heroin
being injected into soft tissue ("skin popping").
11
Infant botulism occurs from the consumption of C. botulinum spores. The spores invade the
gastrointestinal tract, replicate, and release toxin, which is absorbed into the circulation. The
source of spores typically is unknown, although ingestion of corn syrup or raw honey accounts
for some cases.
Adult intestinal toxemia (or undefined) botulism occurs from the consumption of C.
botulinum spores. Characteristics include unknown source of toxin, presence of toxin in stool,
and abnormal gastrointestinal pathology (e.g., Billroth surgery, Crohn's disease, and peptic
ulcer disease) or antimicrobial drug use.
Iatrogenic botulism been noted very rarely after medical use or misuse of the botulinum
toxin. Purified, highly diluted, injectable botulinum toxin is used to treat a range of spastic or
autonomic muscular disorders. Toxin type A (Botox) is used in extremely minute doses for the
treatment of facial wrinkles and blepharospasm, cervical dystonia strabismus, glabellar lines,
and primary axillary hyperhidrosis. Toxin type B (Myobloc, Neurobloc) is used to treat cervical
dystonia. Dysphagia, limited paresis and other neuromuscular impairment of the toxin are
symtoms that have been seen.
17
S.F. Dept Public Health – Infectious Disease Emergencies BOTULISM, July 2008 Page 5/11
CLINICAL FEATURES: BOTULISM
1-3, 6, 16
Incubation Period
12-80 hours (range 2 hours to 8 days)
Transmission
Inhalation of toxin
Consumption of toxin or C. botulinum spores
Contamination of a tissue with toxin or C. botulinum spores
Signs and
Symptoms
Cardinal signs
Afebrile
Symmetrical neurological manifestations
Normal mental status, though may appear lethargic and have difficulty with
communication
Normal to slow heart rate without the presence of hypotension
Normal sensory nerve function, other than vision
Early presentation – cranial nerve abnormalities
Fatigue and vertigo
Double and blurred vision, intermittent ptosis and disconjugate gaze
Difficulty swallowing food
Later presentation – descending paralysis
Difficulty moving eyes and mild pupillary dilation and nystagmus
Tongue weakness, decreased gag reflex, indistinct speech, dysphagia, dysphonia
Symmetrical, descending progressive muscular weakness, especially arms and legs
Unsteady gait
Extreme weakness, including postural neck muscles and occasional mouth
breathing
Autonomic nerve dysfunction; may include urinary retention, orthostasis
Constipation
Ingestional:
Dry mouth and dysarthria
Nausea and vomiting, except when exposure is purified toxin
Inhalational:
Mucus in throat
Serous nasal discharge, salivation
Infant:
Inability to suck and swallow
Constipation
Weakened voice
Floppy neck
Progression and
Complications
Respiratory failure and possible aspiration pneumonia
Residual fatigue, dry mouth or eyes, dyspnea on exertion several years later
Laboratory and
Radiographic
Findings
Normal CSF values
Normal CBC
Normal imaging of brain and spine (CT scan or MRI)
Characteristic EMG findings include:
Decremented response to repetitive nerve stimulation at low frequency (3 Hz)
Facilitated response to repetitive nerve stimulation at high frequencies (10-50 Hz)
Low compound muscle action pontential
CBC, complete blood count; CSF, cerebrospinal fluid, CT, computed tomographic; EMG, electromyogram; MRI,
magnetic resonance imaging.
S.F. Dept Public Health – Infectious Disease Emergencies BOTULISM, July 2008 Page 6/11
DIFFERENTIAL DIAGNOSIS
Diagnosis of botulism during the initial stages requires a high index of suspicion because of the lack
of readily available rapid confirmatory tests.
Important questions to ask include:
recent history of eating
o home-canned or home-prepared vegetable, fruit, including foil-wrapped baked
potato
o lightly preserved or fermented meat and fish products, including seafood products
from Alaska, Canada or the Great Lakes
other known individuals with similar symptoms
recent history of injection drug use, particularly with black tar heroin or cocaine
Key features that distinguish botulism are the constellation of:
afebrile illness
normal mental status
cranial nerves prominently involved
descending paralysis
symmetric bilateral impairment
absence of paresthesias
normal CSF studies
characteristic EMG findings
Other conditions to consider are:
Guillain-Barre syndrome
(especially Miller-Fisher syndrome)
myasthenia gravis
stroke or CNS tumor
CNS infections (particularly of
brainstem)
Lambert-Eaton syndrome
tick paralysis
sudden infant death syndrome
hyperemesis gravidarum
saxitoxin (paralytic shellfish
poisoning)
tetrodotoxin (puffer fish poisoning)
laryngeal trauma
diabetic neuropathy
poliomyelitis/West Nile acute flaccid paralysis
psychiatric illness (i.e., conversion paralysis)
inflammatory myopathy
streptococcal pharyngitis
viral syndrome
hypothyroidism
overexertion
diphtheria
Wernicke's encephalopathy
intoxication with CNS depressants (atropine,
aminoglycoside, magnesium, ethanol,
organophosphates, nerve gas, carbon
monoxide)
S.F. Dept Public Health – Infectious Disease Emergencies BOTULISM, July 2008 Page 7/11
These recommendations are current as of this document date. SFDPH will provide periodic updates as needed
and situational guidance in response to events (www.sfcdcp.org).
If you are testing or considering testing for
botulism, you should:
IMMEDIATELY notify
SFDPH Communicable Disease Control
(24/7 Tel: 415-554-2830).
SFDPH can authorize and facilitate testing, and
will initiate the public health response as needed.
Inform your lab that botulism is under
suspicion.
LABORATORY DIAGNOSIS
Routine laboratory and radiographic findings
for specific clinical presentations of botulism
are listed in the clinical features table.
Although laboratory confirmation should be
initiated as soon as possible if testing
facilities are available, the clinical
presentation should guide clinical
management and public health interventions.
Laboratory confirmation is challenging, but
can be achieved in most cases by detection
of botulinum toxin in serum, respiratory secretions, and stool via mouse bioassay, in which mice
are injected with the patient sample and observed for the development of characteristic symptoms.
Serum specimens must be taken before antitoxin treatment to demonstrate the presence of
botulinum toxin. The test requires 1-4 days to complete and is performed only at reference
laboratories. Electromyography provides diagnostic information more rapidly. Repetitive nerve
stimulation at 20 to 50 Hz differentiates between various etiologies of acute flaccid paralysis.
Electromyography is not recommended for infants.
3, 6, 16
Because the laboratory diagnosis of botulism may take several days to complete, health
department officials can authorize the release of antitoxin prior to laboratory confirmation on the
basis of clinical findings and may be able to provide other rapid detection tests that are currently
investigational (e.g., time-resolved fluorescence assay, toxin micronanosensor, ganglioside-
liposome immunoassay, enzyme-linked immunosorbent assay [ELISA]).
TREATMENT AND PROPHYLAXIS
Treatment
Outcome is based on early diagnosis and treatment. Supportive care (including airway protection,
mechanical ventilation, and feeding by central tube or parenteral nutrition) and timely
administration of equine botulinum antitoxin are keys to the successful management of botulism.
2,
3, 18
Establish a means of communication early because sometimes conditions such as debilitating
headaches are not communicated after the onset of paralysis.
Antitoxin
Antitoxin administration should not be delayed for laboratory confirmation because antitoxin does
not reverse disease or existing paralysis, but only stops progression of disease.
S.F. Dept Public Health – Infectious Disease Emergencies BOTULISM, July 2008 Page 8/11
Patients given antitoxin within the first 24 hours after symptom onset had shorter hospital stays,
shorter duration of ventilatory support, and a lower fatality rate (10%) than those given antitoxin
more than 24 hours after onset (15%) or those who did not receive antitoxin at all (46%).
19, 20
Antitoxin is provided by the Centers for Disease Control and Prevention (CDC) but is available for
release only by the state or local health departments. Delivery can be expected within 12 hours of
request.
Consult public health authorities regarding dosage, because recommendations change. Currently,
the CDC recommends immediate intravenous administration of the trivalent antitoxin (one vial
diluted 1:10 over 30 minutes). If it is suspected that the exposure was to an extremely high
dosage of toxin, the serum may be tested after treatment for the presence of remaining toxin.
16
Because antitoxin is of equine origin, hypersensitivity reactions can occur. From 1967 to 1977, 9%
of persons treated with botulinal antitoxin had a nonfatal hypersensitivity reaction.
21
In recent
years, when the recommended dosage has decreased 2- to 4-fold, less than 1% have experienced
hypersensitivity reactions.
2
A skin test may be valuable in patients with allergies, previous
anaphylaxis, or prior receipt of equine antitoxins. If skin testing is positive, consider desensitizing
over several hours before administering the complete dose of antitoxin or pretreat with
antihistamines, steroids, and epinephrine infusions. Diphenhydramine, epinephrine, and airway
equipment should be easily accessible during any administration.
Human botulism immune globulin is used to treat infants, which is administered intravenously.
22
Supportive care
Ventilatory support may be required for several weeks or more. One study found the mean time
on a ventilator for botulism cases was 58 days.
20
With modern intensive care methods, case fatality rates for botulism in the United States have
dropped to less than 10%. In a mass casualty setting, measurement and management of
ventilatory function may pose challenges because of limited ventilator capacities. Local health
departments can request supplemental laryngoscopes, endotracheal tubes, and Ambu bags from
the CDC. If personnel are limited, consider recruiting health civilians for bag ventilation.
A reverse Trendelenburg positioning with cervical vertebral support has been beneficial in terms of
respiratory mechanics and airway protection in nonventilated infants with botulism, but has not
been tested in adults. In adults, especially those with obesity, a 20- to 25-degree angle may be
beneficial.
3
Utilize physical therapy and physical turning to minimize intensive care complications.
Secondary infections
Antibiotics may be used for treatment of secondary infections; however, aminoglycosides and
clindamycin are contraindicated because they may exacerbate the neuromuscular blockade.
3
S.F. Dept Public Health – Infectious Disease Emergencies BOTULISM, July 2008 Page 9/11
These recommendations are current as of this document date. SFDPH will provide periodic updates as needed
and situational guidance in response to events (www.sfcdcp.org).
Post-exposure prophylaxis
There is currently no available postexposure prophylaxis for asymptomatic exposed persons.
3, 16
Such persons should be educated regarding the signs and symptoms of clinical botulism and
instructed to seek medical care immediately if symptoms occur. Not all exposed persons will
develop clinical symptoms. Exposed persons and their families may experience anxiety and/or
somatic symptoms that may include neurologic symptoms. These patients should be carefully
assessed. Antitoxin supplies are limited, and therapy will be reserved for patients with compatible
neurological findings.
Vaccine
Preexposure immunization with botulinum toxoid is restricted to certain laboratory and military
personnel. Supplies are extremely limited and would not be available for the public.
3, 16
COMPLICATIONS AND ADMISSION CRITERIA
In patients with botulism, cranial nerve dysfunction progresses inexorably to a symmetric,
descending muscle weakness or paralysis. Respiratory failure occurs in 40-70% of botulism
patients because of declining upper airway and ventilatory muscle strength. Additional
complications of botulism include secondary infection of the respiratory system and sequelae
related to intubation and mechanical ventilation, prolonged immobilization, and autonomic
dysfunction. Diminished respiratory muscle function and easy fatigability were described by
botulism patients 2 years after recovery.
Hospital admission is required for protection of the airway, mechanical ventilatory support, and
fluid and nutritional management until normal muscular function returns.
INFECTION CONTROL
Clinicians should notify local public health authorities and their laboratory of any suspected
botulism case. Health authorities may conduct epidemiologic investigations and implement disease
control interventions to protect the public. Both HICPAC (Hospital Infection Control Practices
Advisory Committee) of the CDC and the Working Group for Civilian Biodefense recommend
Standard Precautions for botulism patients in a hospital setting without the need for isolation.
Person-to-person transmission does not occur.
3, 16, 23
Decontamination
After exposure to toxin, wash clothes and skin with soap and water. Inactivation of the toxin in the
environment can take 2 days; however, changes in temperature and humidity can affect the rate of
decomposition. Contaminated surfaces and spills of cultures or toxin can be disinfected with
S.F. Dept Public Health – Infectious Disease Emergencies BOTULISM, July 2008 Page 10/11
sodium hypochlorite (0.1% which is a 1:50 dilution of household bleach) or sodium hydroxide
(0.1N). Moist heat at 120°C for at least 15 minutes destroys spores.
3
PEARLS AND PITFALLS
1. Botulism is often misdiagnosed as a polyradiculopathy (Guillain-Barre syndrome or Miller-
Fisher syndrome), myasthenia gravis, or other diseases of the central nervous system.
Botulism is distinguished from other flaccid paralyses by its initial presentation with
prominent cranial neuropathy, its subsequent descending, symmetrical paralysis, and its
absence of sensory nerve deficits.
2. In the United States, botulism is more likely than Guillain-Barre syndrome, chemical
poisoning, or poliomyelitis to cause a cluster of cases of acute flaccid paralysis.
3. Botulism antitoxin neutralizes freely circulating toxin but does not dislodge toxin already
bound to presynaptic receptors. Early administration of antitoxin can help to inhibit further
paralysis, but does not reverse paralysis that has already occurred.
4. Botulism antitoxin is limited in quantity and is available only through public health
authorities. Since the laboratory diagnosis of botulism requires an in-vivo assay and may
take several days to complete, health department officials often authorize the release of
antitoxin prior to laboratory confirmation, on the basis of clinical findings.
REFERENCES
1. Bleck TP. Clostridium botulinum. In: Mandell GL, Bennett JE, Dolin R, eds. Principles and
practice of infectious diseases. 6 ed. New York: Churchill Livingstone; 2005:2822-2828.
2. Sobel J. Botulism. Clin Infect Dis. Oct 15 2005;41(8):1167-1173.
3. Arnon SS, Schechter R, Inglesby TV, et al. Botulinum toxin as a biological weapon: medical
and public health management. Jama. Feb 28 2001;285(8):1059-1070.
4. Franz DR, Jahrling PB, Friedlander AM, et al. Clinical recognition and management of patients
exposed to biological warfare agents. Jama. Aug 6 1997;278(5):399-411.
5. Bleck TP. Botulinum Toxin as a Biological Weapon. In: Mandell GL, Bennett JE, Dolin R, eds.
Principles and practice of infectious diseases. 6 ed. New York: Churchill Livingstone;
2005:3624-3625.
6. CIDRAP. Botulism: Current, comprehensive information on pathogenesis, microbioloby,
epidemiology, diagnosis, and treatment. Center for Infectious Disease Research and Policy,
University of Minnesota. May 23. Available at:
http://www.cidrap.umn.edu/cidrap/content/bt/botulism/biofacts/botulismfactsheet.html
.
7. Huss HH. Distribution of Clostridium botulinum. Appl Environ Microbiol. Apr 1980;39(4):764-
769.
8. Peck MW. Clostridium botulinum and the safety of minimally heated, chilled foods: an
emerging issue? J Appl Microbiol. Sep 2006;101(3):556-570.
9. Shapiro RL, Hatheway C, Swerdlow DL. Botulism in the United States: a clinical and
epidemiologic review. Ann Intern Med. Aug 1 1998;129(3):221-228.
10. Sobel J, Tucker N, Sulka A, McLaughlin J, Maslanka S. Foodborne botulism in the United
States, 1990-2000. Emerg Infect Dis. Sep 2004;10(9):1606-1611.
S.F. Dept Public Health – Infectious Disease Emergencies BOTULISM, July 2008 Page 11/11
11. Werner SB, Passaro D, McGee J, Schechter R, Vugia DJ. Wound botulism in California, 1951-
1998: recent epidemic in heroin injectors. Clin Infect Dis. Oct 2000;31(4):1018-1024.
12. CDPH. California Monthly Summary Report Selected Reportable Diseases. Division of
Communicable Disease Control, California Department of Public Health. Available at:
http://www.cdph.ca.gov/data/statistics/Pages/CD_Tables.aspx
.
13. SFDPH. San Francisco Communicable Disease Report, 1986-2003. San Francisco Department
of Public Health. May. Available at: http://www.sfcdcp.org/publications
.
14. SFDPH. Annual Report of Communicable Diseases in San Francisco, 2004-2005. San Francisco
Department of Public Health. August. Available at: http://www.sfcdcp.org/publications
.
15. SFDPH. Annual Report of Communicable Diseases in San Francisco, 2006. San Francisco
Department of Public Health. January. Available at: http://www.sfcdcp.org
.
16. CDC. Botulism: Information and Guidance for Clinicians. Centers for Disease Control and
Prevention. Available at: http://www.bt.cdc.gov/agent/Botulism/clinicians/
.
17. Chertow DS, Tan ET, Maslanka SE, et al. Botulism in 4 adults following cosmetic injections
with an unlicensed, highly concentrated botulinum preparation. Jama. Nov 22
2006;296(20):2476-2479.
18. Kongsaengdao S, Samintarapanya K, Rusmeechan S, et al. An outbreak of botulism in
Thailand: clinical manifestations and management of severe respiratory failure. Clin Infect Dis.
Nov 15 2006;43(10):1247-1256.
19. Tacket CO, Shandera WX, Mann JM, Hargrett NT, Blake PA. Equine antitoxin use and other
factors that predict outcome in type A foodborne botulism. Am J Med. May 1984;76(5):794-
798.
20. Horowitz BZ. Botulinum toxin. Crit Care Clin. Oct 2005;21(4):825-839, viii.
21. Black RE, Gunn RA. Hypersensitivity reactions associated with botulinal antitoxin. Am J Med.
Oct 1980;69(4):567-570.
22. Arnon SS, Schechter R, Maslanka SE, Jewell NP, Hatheway CL. Human botulism immune
globulin for the treatment of infant botulism. N Engl J Med. Feb 2 2006;354(5):462-471.
23. Siegel JD, Rhinehart E, Jackson M, Chiarello L, HICPAC. Guideline for Isolation Precautions:
Preventing Transmission of Infectious Agents in Healthcare Settings, 2007. Centers for
Disease Control and Prevention. Available at:
http://www.cdc.gov/ncidod/dhqp/gl_isolation.html
.
S.F. Dept. Public Health – Infectious Disease Emergencies BRUCELLOSIS, August 2005 Page 1/6
Outline
Agent
Epidemiology
Clinical Features
Differential Diagnosis
Laboratory Diagnosis
Treatment and Prophylaxis
Infection Control
References
By law, health care providers must report
suspected or confirmed Brucellosis to the local
health department immediately (within 1 hr).
Even a single case of Brucellosis is considered
an outbreak and is a public health emergency.
To report: call SFDPH communicable disease
control (24/7 Tel: 415-554-2830).
Upon receipt, SFDPH will initiate the public health
response and can facilitate lab testing.
AGENT
Brucellosis is a zoonotic disease of domestic and wild animals, caused by the non-motile, non-
sporulating, small, gram-negative coccobacilli bacteria of the genus Brucella. Four species can be
pathogenic in humans: B. melitensis, B. abortus, B. canis and B. suis. They are highly infectious,
especially B. melitensis and B. suis.
Brucellae contain lipopolysaccharide (LPS) in the outer cell membrane, however this LPS is
structurally different from that of the Enterobacteriaceae, and this feature may underlie the
reduced pyrogenicity (less than 1/100th) of Brucella LPS compared with E. coli LPS.
EPIDEMIOLOGY
Brucellosis as a Biological Weapon
The US military developed B. suis as a biological weapon in the 1950’s, but terminated this
program in 1967. Their transmissibility by aerosol suggests that Brucella organisms might be a
candidate for use as a bioweapon. Fewer than 100 organisms could constitute an infectious
aerosol. The CDC considers brucellosis a lesser threat than agents such as anthrax and smallpox:
its incubation period is rather long, many infections are asymptomatic, and the mortality is low.
However, it might be used as an incapacitating agent as it often causes a protracted illness.
The most likely form of intentional release would be via infectious aerosols; however food-borne
exposure is also possible. Any large-scale outbreak of brucellosis would suggest deliberate release
of Brucella organisms. Bioterrorism might also be suggested by clusters of brucellosis cases
without a travel history to endemic areas, without relevant foodborne or occupational exposures, or
where the cases are linked in time and place (e.g. geographically related cases following a wind
direction pattern).
BRUCELLOSIS AUGUST 2005
S.F. Dept. Public Health – Infectious Disease Emergencies BRUCELLOSIS, August 2005 Page 2/6
Naturally Occurring Brucellosis
Brucella species infect mainly ruminant mammals, including cattle, sheep, goats, pigs, and camels,
in which they cause genital infection, abortion, and fetal death. Additional animal reservoirs
include elk, caribou, bison, deer, and wild and domestic canines. Animals may transmit Brucella
organisms during septic abortion, at the time of slaughter, and in their milk. Humans are usually
infected incidentally in one of three ways:
Direct contact with the tissues of infected animals. Occupational exposures include those
of veterinarians, shepherds, ranchers, and slaughterhouse workers, who are believed to
become infected through skin abrasions, cuts, or conjunctival exposure.
Ingestion of contaminated food or water. Consumption of contaminated milk products is
the most common mode of acquisition worldwide. Pasteurization of dairy products
prevents transmission and has drastically reduced the incidence of brucellosis in the
developed world. Meat products are rarely the source of infection because they are not
usually eaten raw and the number of organisms in muscle tissue is low.
Inhalation of infectious aerosols. The inhalational route is of consequence for occupational
exposures listed above, particularly slaughterhouse workers, and may also constitute a risk
factor for laboratory workers who culture Brucella bacteria.
Naturally occurring exposures to brucellosis are unusual in the USA and tend to be isolated. Fewer
than 200 total cases per year are reported in the United States, most of these in Texas and
California. During the period 1994-2003 there were 275 total cases reported in California; of
these, 2 occurred in San Francisco. The epidemiology of brucellosis in Texas and California has
changed from a disease associated with exposure to cattle to one linked to the ingestion of
unpasteurized goat milk products (“queso fresco”) imported from Mexico.
Disease incidence is much higher in the Middle East and Mediterranean regions, and in China,
India, and Latin America.
CLINICAL FEATURES
The brucellae are facultative intracellular pathogens that can survive and multiply within the
phagocytic cells of the host. After entering the human body and being taken up by local tissue
lymphocytes, brucellae are transferred through regional lymph nodes into the circulation and are
subsequently seeded throughout the body, with tropism for the reticuloendothelial system.
Clinical manifestations of brucellosis are diverse and often non-specific, and the course of the
disease is variable. For most exposures, the clinical syndrome does not clearly relate to the portal
of entry of the organism; however those exposed via the aerosol route may have increased
frequency of respiratory symptoms. B. melitensis tends to cause more severe, systemic illness
than the other brucellae; B. suis is more likely to cause localized, suppurative disease.
S.F. Dept. Public Health – Infectious Disease Emergencies BRUCELLOSIS, August 2005 Page 3/6
BRUCELLOSIS: CLINICAL FEATURES
Incubation Period
2-4 weeks (range 5 days to several months)
Signs & Symptoms
Fever always occurs; spiking or “undulant” pattern may be apparent
May have acute, subacute, or chronic presentation
Other consitutional symptoms: malaise, anorexia, back pain, myalgias,
arthralgias, headache
“Malodorous perspiration”
Mild lymphadenopathy (10-20%)
Hepatomegaly or splenomegaly (20-30%)
Nonspecific skin lesions (papules, ulcers, e. nodosum, petechiae) in 5%
Weight loss among chronically infected
Almost any organ system can be involved
Most affected persons recover in 3-12 months, however a minority may
develop one or more of the complications below
Complications
Skeletal: osteomyelitis (most common); also sacroiliitis, spondylitis,
peripheral arthritis
Reproductive: spontaneous abortion; epididymo-orchitis
GI: acute ileitis, hepatitis, liver abscess, liver granuloma
CNS: meningitis, encephalitis, brain abscess, myelitis
CV: endocarditis, pericarditis
Pulmonary: bronchitis, pneumonia, lung nodules, abscess, hilar adenopathy,
pleural effusion/empyema, lung abscess
Uveitis
Laboratory Findings
Mild leukopenia with relative lymphocytosis
Mild anemia and thrombocytopenia may be present; DIC is rare
Other abnormalities are related to the organ system involved
DIFFERENTIAL DIAGNOSIS
Due to the non-specific presentation and numerous, varied complications of brucellosis in humans,
the differential diagnosis is vast and will not be addressed in detail here. A high index of suspicion
is necessary to diagnose brucellosis, due both to the non-specific presentation and to the relatively
long latency period between inoculation and the development of symptoms.
Key clinical questions that help to suggest naturally-acquired brucella infection include:
History of contact with ruminant mammals, via occupational or recreational exposures
(veterinarians, slaughterhouse workers, ranchers, shepherds, laboratory workers, visitors
to dairy farms or petting zoos)
Consumption of unpasteurized milk products (e.g. “queso fresco”)
Travel to areas where brucellae are established in the animal population
In the setting of intentional attack using brucella, these exposures may be notably absent.
S.F. Dept. Public Health – Infectious Disease Emergencies BRUCELLOSIS, August 2005 Page 4/6
If you are testing or considering testing for
Brucellosis, you should:
IMMEDIATELY notify SFDPH Communicable
Disease Control (24/7 Tel: 415-554-2830)
Notify the lab that Brucellosis is suspected, as
the organism may pose a risk to personnel.
Neither CDC nor the Working Group on Civilian
Biodefense has issued bioterrorism-specific
treatment/prophylaxis recommendations for
Brucellosis. SFDPH will provide situational
guidance in response to events
(www.sfdph.org/cdcp).
LABORATORY DIAGNOSIS
Definitive diagnosis of brucellosis is made
when brucellae are recovered from infected
tissues, typically blood or bone marrow.
The rate of isolation ranges from 15-70%.
The organism has also been recovered from
urine, CSF, synovial fluid, and biopsies of
liver and lymph nodes. Brucella species
often require several weeks to grow in
culture, so this method is not useful for
rapid identification.
A presumptive diagnosis can be made using
specific antibody titers. The serum
agglutination test (SAT) is based on
antibody against lipopolysaccharide. Most cases of active infection have a single titer of 1:160 or
higher. Drawbacks of the SAT include the inability to diagnose B. canis infection, cross-reaction
with other gram-negative organisms, and the lack of seroconversion in some cases. Also, SAT are
not suitable for patient follow-up since titers can remain elevated for a prolonged period. The
ELISA test for brucellosis relies on cytoplasmic antigens and is both more sensitive and more
specific than SAT. However, like SAT, titers can remain elevated for prolonged periods. A number
of variations of PCR tests are becoming available, but standardization is still lacking.
TREATMENT AND PROPHYLAXIS
Treatment
Generally accepted principles of brucellosis treatment are that the antibiotics used must penetrate
macrophages, and that monotherapy has a higher rate of relapse compared with combined therapy
regimens.
BICHAT, the European Commission’s Task Force on Biological and Chemical Agent Threats, has
recommended as first-line therapy: Doxycline 100 mg IV/PO twice daily, combined with either
streptomycin 1 gm IM once or twice daily for up to 2 weeks; OR rifampin 600-900 mg PO daily for
6 weeks; OR gentamicin 5 mg/kg/day IV in 2 divided doses for up to 2 weeks. This regimen,
dosage-adjusted to body weight, is also first-line treatment for children >8 years old. Treatment
with trimethoprim-sulfamethoxazole (TMP-SMX) plus rifampin is recommended for pregnant
women and for children <8 years of age. Quinolones have been used with success against
Brucellae, while macrolide antibiotics are not effective. Complications of brucellosis are also ed
with 2-drug regimens, while neurobrucellosis has generally been treated with 3 agents.
S.F. Dept. Public Health – Infectious Disease Emergencies BRUCELLOSIS, August 2005 Page 5/6
These recommendations are current as of this document date. SFDPH will provide periodic
updates as needed and situational guidance in response to events (www.sfdph.org/cdcp).
Relapses occur in about 10% of cases, usually during the first year after infection, and are often
milder in severity than the initial disease. Relapse has been managed with a repeated course of
the usual antibiotic regimens. Most cases of relapse are felt to be caused by inadequate treatment.
Post-Exposure Prophylaxis
There is little evidence to support the utility of post-exposure prophylaxis against brucellosis in
humans. BICHAT has recommended a 3-6 week course of doxycycline OR TMP-SMX, with the
addition of rifampin to either drug. In the event of outbreak, SFDPH will provide updated,
situational guidelines for prophylaxis (www.sfdph.org/cdcp).
Vaccination
There is currently no licensed vaccine available for brucellosis. Some limited clinical data exist on a
live, attenuated vaccine candidate, but licensing and production of this vaccine are not anticipated.
INFECTION CONTROL
*
Person-to-person transmission of brucellosis is extremely rare. Standard Precautions are
considered adequate for patients with brucellosis.
Brucella is sensitive to exposure to heat and most disinfectants but can survive in the
environment for up to two years under specific conditions, becoming a continuing threat to
both humans and animals.
REFERENCES
Bossi P et al. Bichat Guidelines for the Clinical Management of Brucellosis and Bioterrorism-related
Brucellosis. Eurosurveillance 2004; 9(12):1-5.
CDC. Suspected Brucellosis Case Prompts Investigation of Possible Bioterrorism-Related Activity.
MMWR 2000, June 16; 49(23):509-512.
Franz DR, Jahrling PB, Friedlander AM, et al. Clinical recognition and management of patients
exposed to biological warfare agents. JAMA 1997;278(5):399-411
*
For description of Precautions, see Chapter on Infection Control.
S.F. Dept. Public Health – Infectious Disease Emergencies BRUCELLOSIS, August 2005 Page 6/6
Friedlander AM & Hoover DL. Brucellosis. Textbook of Military Medicine: Medical Aspects of
Chemical and Biological Warfare. Online at www.vnh.org/MedAspChemBioWar
LA County DHS. Terrorism Agent Information and Treatment Guidelines for Clinicians and
Hospitals. June 2003. (labt.org/Zebra.asp)
Pappas G et al. Brucellosis. N Engl J Med 2005;352:2325-36.
WHO. Health aspects of chemical and biological weapons. Ed 2. Geneva, Switzerland: World Health
Organization, 2004:250-4
S.F. Dept Public Health – Infectious Disease Emergencies PLAGUE, June 2008 Page1/13
Outline
Introduction
Epidemiology
Clinical Features
Differential Diagnosis
Laboratory Diagnosis
Treatment and Prophylaxis
Complications and Admission Criteria
Infection Control
Pearls and Pitfalls
References
Immediately report any suspected or
confirmed cases of plague to:
SFDPH Communicable Disease Control
(24/7 Tel: 415-554-2830)
- By law, health care providers must report
suspected or confirmed cases of plague to their
local health department immediately [within 1 hr].
- SFDPH Communicable Disease Control can
facilitate specialized testing and will initiate the
public health response as needed.
Also notify your:
Infection Control Professional
Clinical Laboratory
INTRODUCTION
Plague is an acute bacterial infection caused by Yersinia pestis, a member of the family
Enterobacteriaceae. Y. pestis is a pleomorphic, nonmotile, nonsporulating, intracellular, Gram-
negative bacillus that has a characteristic bipolar appearance on Wright, Giemsa, and Wayson’s
stains. There are three virulent biovars: antiqua, medievalis, and orientalis and a fourth avirulent
biovar, microtu.
1, 2
The orientalis biovar is thought to have originated in southern China and caused
the most recent pandemic.
3
The Working Group for Civilian Biodefense considers plague to be a potential biological weapon
because of the pathogen’s availability “around the world, capacity for its mass production and
aerosol dissemination, difficulty in preventing such activities, high fatality rate of pneumonic
plague, and potential for secondary spread of cases during an epidemic.” Of the potential ways
that Y. pestis could be used as a biological weapon, aerosol release would be most likely. This
method has been successfully demonstrated to cause disease in Rhesus macaques.
4
EPIDEMIOLOGY
Plague as a Biological Weapon
In the 20
th
century, countries including the United States, the former Soviet Union, and Japan
developed ways for using Y. pestis as a weapon.
5-7
Creating aerosolized plague is technically
challenging; however, if an intentional release of aerosolized plague were to take place, an
outbreak of pneumonic plague would be likely. This would be of serious concern because of the
high case-fatality rate and the potential for person-to-person transmission.
4, 7, 8
PLAGUE JUNE 2008
S.F. Dept Public Health – Infectious Disease Emergencies PLAGUE, June 2008 Page2/13
An outbreak of disease caused by an intentional release of Y. pestis would have the following
characteristics:
Clustering in time: multiple similarly presenting cases of severe, progressive multilobar
pneumonia, generally 2-4 days after release (range of 1 to 6 days)
Atypical host characteristics: unexpected, unexplained cases of acute illness in previously
healthy persons who rapidly develop severe, progressive multilobar pneumonia with
hemoptysis and gastrointestinal symptoms
Unusual geographic clustering: multiple cases in an urban area where naturally occurring
plague is not endemic
Absence of risk factors: patients lack plague exposure risk factors (e.g., recent flea bite;
exposure to rodents, especially rabbits, squirrels, wood rat, chipmunk, or prairie dogs;
scratches or bites from infected domestic cats)
Intentionally-released Y. pestis strains may be altered to have enhanced virulence, antimicrobial
resistance, or increased ability to evade vaccines and diagnostic tests.
4, 7
Naturally Occurring Plague
Reservoirs
The natural reservoir for Y. pestis is primarily wild rodents. Around the world, the domestic rat has
been associated with the most human cases; however in the western United States, burrowing
rodents (e.g., ground squirrels, rock squirrels, and prairie dogs) are the most important reservoir.
9
A recent study has found soil to be a potential reservoir, with Y. pestis persisting from months to
several years n association with wild rodents.
10, 11
Other mammals that act as hosts include cats,
goats, sheep, camels, and humans.
10, 12-17
Human plague cases often follow epizootics in local
rodent populations.
10, 18, 19
Mode of Transmission
Humans can become infected in a number of ways:
4, 13, 14, 16, 17, 20
Bite of infected rat flea
Direct contact with infected draining buboes
Direct contact (including bites or scratches) with infected animals
Inhalation of respiratory droplets from pneumonic plague-infected humans or animals
(within 2 meters)
Ingestion of bacteria (e.g., eating infected meat)
Human plague cases in nature are most commonly acquired from animal reservoirs via bites of the
Oriental rat flea.
9, 21
Worldwide occurrence
The first recorded plague pandemic was the Justinian plague (541-767 AD) which caused ~100
million deaths and is thought to have contributed to the demise of the Roman Empire. The second
pandemic, also known as the Black Death, lasted from the 14
th
to the 19
th
centuries and was
estimated to have killed between a third and a half of Europe's population. The third and most
recent pandemic began in 1894 in China and caused an estimated 12 million deaths.
14, 19, 22, 23
Recent outbreaks in humans have included India (1994), Zambia (1996), Indonesia (1997), Algeria
S.F. Dept Public Health – Infectious Disease Emergencies PLAGUE, June 2008 Page3/13
(2003), Uganda (2004), and the Congo (2005).
24-28
Approximately 1,800 worldwide cases of
plague are reported annually to the WHO, from all continents except Europe and Australia.
10
Occurrence in the United States
Ships carrying infected rats introduced plague to the Americas via the ports on the Pacific Ocean
and Gulf of Mexico in the early 1900s. In San Francisco, urban rats passed along the disease to
native rodent populations. Eventually, plague spread across the western half of the United States
and has been found in the native rodent population, their fleas, and their predators. Naturally
occurring human plague generally occurs during the summer months in persons exposed to the
reservoir.
27, 29
The last urban plague outbreak in the US occurred in Los Angeles in 1925.
25-27, 30
From 1990 to 2005, a median of 7 cases of plague per year were reported in the US.
26
Based on
provisional data, in 2006, there were 17 cases, and in 2007, 7 cases.
31
Occurrence in California and San Francisco
From 1994 to 2007, 9 cases of plague were reported in California, and none of these occurred in
San Francisco.
32-35
CLINICAL FEATURES
Human plague occurs in many forms, determined primarily by the route of infection. The most
common forms of plague in humans are bubonic plague, septicemic plague, and pneumonic plague.
These are presented in detail below.
Plague infection is a severe clinical illness that can be life-threatening. Case fatality rates vary
based on the route of infection. Mortality was historically much higher with nearly 100% mortality
for untreated septicemic and pneumonic plague and 50-60% mortality for untreated bubonic
plague cases. Administration of appropriate antibiotic treatment within the first 18 to 24 hours has
decreased mortality rates to 30-50% for septicemic plague, 5-15% for pneumonic plague, and less
than 5% for bubonic plague.
4
Thus, early administration of appropriate antibiotic treatment is
critical, as poor outcomes occur with delays in seeking care and/or instituting effective
antimicrobial treatment.
Pneumonic Plague
Primary pneumonic plague occurs when the organism is inhaled in respiratory droplets from
infected humans or animals or in infectious aerosols accidentally or intentionally produced (e.g.,
spilled lab specimen or bioterrorism related release). Secondary pneumonic plague occurs when
there is hematogenous spread of the organism to the lung. Primary pneumonic plague causes a
more acute and fulminant disease. Pneumonic plague is not highly contagious but transmission
can occur with prolonged close contact (within 2 meters) with a coughing patient in the end stage
of illness. In a recent outbreak in Uganda, 1.3 pneumonic plague transmissions per pneumonic
plague case were reported.
24
If untreated, pneumonic plague can spread and progress to
septicemic plague.
S.F. Dept Public Health – Infectious Disease Emergencies PLAGUE, June 2008 Page4/13
PNEUMONIC PLAGUE
Incubation period 1-4 days, with a maximum of 6 days
Transmission Inhalation of contaminated aerosol
Inhalation of respiratory droplets from pneumonic plague-infected humans or animals
(within 2 meters)
Secondary hematogenous spread to the lung
Signs and symptoms Acute fever, chills, malaise, myalgia, headache
Productive cough, with sputum becoming more and more bloody
Chest pain, dyspnea, cyanosis
Tachypnea in children
Gastrointestinal symptoms
Progression and
complications
Refractory pulmonary syndrome
Adult respiratory distress syndrome
Septicemia
Laboratory and
Radiographic Findings
Leukocytosis with left shift
Gram-negative bipolar bacilli on sputum smear
Elevated creatinine and abnormally high liver enzymes
CXR findings include alveolar infiltrates progressing to lobar consolidation, pleural
effusion
Rarely, mediastinal widening on CXR due to adenopathy
Bubonic Plague
Yersinia pestis can cause bubonic plague in humans via the bite of an infected rodent flea. Y.
pestis survives in the flea midgut after a blood meal from an infected host. The organism is
transmitted to a new host when the flea regurgitates during its next feeding. Y. pestis migrates to
regional lymph nodes where it causes hemorrhagic lymphadenitis, creating the swollen, painful
buboes that are characteristic of bubonic plague. The organisms often enter the bloodstream,
causing hemorrhagic lesions in distant lymph nodes and organs. If untreated, bubonic plague can
spread and progress to pneumonic or septicemic plague. Approximately, 80% of cases develop
bacteremia, 25% develop clinical septicemia and 10% develop pneumonia as a complication.
BUBONIC PLAGUE
Incubation period 1-8 days
Transmission Bite of infected rat flea
Direct contact with infected draining buboes
Direct contact (including bites or scratches) with infected animals
Signs and symptoms Major
Sudden onset of chills, high fever, headache, lethargy
Buboes - Swollen, red, painful lymph nodes in areas proximal to the inoculation site
(e.g., inguinal, axillary or cervical areas)
Rapid pulse
Hypotension
Other
Gastrointestinal discomfort
Restlessness, confusion, lack of coordination
Skin lesion at the site of the flea bite occurs in < 10% of cases
Buboes may rupture and suppurate in second week
S.F. Dept Public Health – Infectious Disease Emergencies PLAGUE, June 2008 Page5/13
Progression and
complications
Septicemia
Secondary pneumonic plague
Meningitis (rare)
Laboratory findings Leukocytosis with left shift
Gram-negative bipolar bacilli on bubo aspirate smear
Elevated creatinine and abnormally high liver enzymes
Septicemic Plague
In primary septicemic plague there is systemic sepsis caused by Y. pestis, but without noticeable,
preceding lymph node or pulmonary involvement. Up to 25% of naturally-occurring plague cases
may present with primary septicemic plague.
19
Secondary septicemic plague occurs commonly
with either bubonic or pneumonic plague.
Septicemic plague causes a Gram-negative sepsis syndrome with multi-organ involvement,
disseminated intravascular coagulation (DIC), and shock. In the late stages of infection, high-
grade bacteremia often occurs, with identifiable organisms on peripheral blood smear.
18
Meningitis
can occur and is characterized by cerebrospinal fluid (CSF) with many polymorphonuclear
leukocytes.
9
SEPTICEMIC PLAGUE
Incubation period 1-4 days
Transmission Site of primary infection may be unknown
Signs and symptoms Acute fever, chills, weakness, malaise
Gastrointestinal symptoms
Purpuric skin lesions and gangrene of the distal digits
Progression and
complications
Disseminated intravascular coagulation (DIC)
Shock
Multi-organ failure
Laboratory findings Leukocytosis with left shift and toxic granulation
Gram-negative bipolar bacilli on blood smear
Disseminated intravascular coagulation (DIC)
Elevated creatinine and abnormally high liver enzymes
Other syndromes caused by Y. pestis infection include:
Plague meningitis. Although it is generally a complication of other forms of plague, it
can be the presenting clinical syndrome. Plague meningitis results from hematogenous
spread of Y. pestis organisms and is characterized by CSF with many polymorphonuclear
leukocytes.
9
Plague pharyngitis. Plague pharyngitis generally results from direct inoculation of the
pharynx. Eating raw infected meat is a risk factor. Clinically, plague pharyngitis presents
as a severe pharyngitis or tonsillitis with cervical adenitis.
Pestis minor. Pestis minor is a milder form of bubonic plague. Lymph nodes drain and
patients convalesce without treatment.
10
S.F. Dept Public Health – Infectious Disease Emergencies PLAGUE, June 2008 Page6/13
DIFFERENTIAL DIAGNOSIS
The diagnosis of plague during the initial stages requires a high index of suspicion because of the
nonspecific, flu-like picture early in the disease. Early diagnosis is critical because prompt
administration of antibiotics can decrease mortality.
Differential: Pneumonic Plague
Consider pneumonic plague in any case of severe Gram-negative pneumonia.
Key features that may help to distinguish plague pneumonia are:
Primary pneumonic plague:
Rapid onset and rapid progression
Secondary pneumonic plague:
Presence of painful adenitis (buboes)
Primary or secondary pneumonic plague:
No response to typical antibiotic therapy for community-acquired pneumonia
Hemoptysis in late stages of disease
Other conditions to consider are:
bacterial pneumonia (Mycoplasma,
Legionella, Staphylococcus,
Streptococcus, Haemophilus, Klebsiella,
Moraxella)
viral pneumonia (influenza, respiratory
syncytial virus [RSV], cytomegalovirus
[CMV], hantavirus, severe acute
respiratory syndrome [SARS])
Chlamydia infection
Q fever
inhalation anthrax
tularemia
ricin
rickettsial infections
aerosolized exposure to staphylococcal
enterotoxin B
Differential: Bubonic Plague
A key feature that may help to distinguish bubonic plague is:
Presence of painful adenitis (buboes) progressing to systemic disease
Other conditions to consider are:
cat scratch disease (Bartonella)
ulceroglandular tularemia
adenitis due to staphylococcal,
streptococcal, or filarial infection
tuberculosis
non-tuberculosis mycobacterial
infection
lymphogranuloma venereum
Capnocytophaga canimorsus infection
chancroid
primary genital herpes
primary or secondary syphilis
appendicitis
strangulated inguinal or femoral hernia
lymphadenopathy (secondary lymphoma,
Kikuchi’s lymphadenitis, systemic lupus
erythematosus, toxoplasmosis, infectious
mononucleosis)
S.F. Dept Public Health – Infectious Disease Emergencies PLAGUE, June 2008 Page7/13
If you are testing or considering testing for
plague, you should:
IMMEDIATELY notify
SFDPH Communicable Disease Control
(24/7 Tel: 415-554-2830).
SFDPH can authorize and facilitate testing, and
will initiate the public health response as needed.
Inform your lab that plague is under
suspicion. Some commercial bacterial test
systems cannot reliably identify Y. pestis.
Differential: Septicemic Plague
Key features that may help to distinguish septicemic plague are:
Primary septicemic plague:
Absence of painful adenitis (buboes) or pulmonary involvement
Secondary septicemic plague:
Presence of painful adenitis (buboes)
Other conditions to consider are:
Gram-negative sepsis
Gram-positive sepsis (Staphylococcus)
meningococcemia
rickettsial infections
malaria
louse-borne relapsing fever
appendicitis
LABORATORY DIAGNOSIS
Routine laboratory and radiographic findings for
specific clinical presentations of plague are
listed in the clinical features tables.
Initial identification of the organism relies on
microscopic evaluation of infected tissue (blood,
sputum, CSF, or fluid aspirated from a bubo or
skin lesion scraping). Staining of the infected
tissue may reveal Gram-negative bacilli (Gram
stain) and bipolar staining (Wright, Giemsa, or
Wayson stain).
4, 9
Order a Gram stain, culture,
and Giemsa, Wright or Wayson stain of the
material. Store and transport blood at room temperature. Transport other samples at room
temperature, but store under refrigeration if transport time will be > 2 hours.
Although recommended, culture and isolation may be difficult. Blood and site-specific specimens
should be collected prior to antibiotic administration as sterilization can occur rapidly. Y. pestis is
slow-growing in culture and may not demonstrate growth until 48 hours after inoculation. Also,
many commercial bacterial identification systems may misidentify Y. pestis.
4
To improve yield and
ensure biosafety precautions, clinicians should notify laboratory personnel when plague is
suspected.
Although rapid diagnostic tests are not widely available, the public health laboratory system may
have access to rapid diagnostic testing on clinical specimens (e.g., polymerase chain reaction [PCR]
or direct fluorescent antibody testing for Y. pestis F1 antigen).
S.F. Dept Public Health – Infectious Disease Emergencies PLAGUE, June 2008 Page8/13
These recommendations are current as of this document date. SFDPH will provide periodic updates as needed
and situational guidance in response to events (www.sfcdcp.org).
TREATMENT AND PROPHYLAXIS
Treatment
Supportive care and timely administration of antibiotics are the keys to successful management of
plague. Plague pneumonia is often fatal if antibiotics are not begun within 12-24 hours of
symptoms. Many patients will require intensive care with respiratory support because of
complications of Gram-negative sepsis.
Resistant strains may occur either naturally or be engineered. In 1995, 2 distinct strains of
naturally-occurring antibiotic-resistant Y. pestis were isolated from human cases of bubonic plague
in Madagascar. One strain was resistant to all drugs recommended for plague treatment and
prophylaxis and the other had high-level resistance to streptomycin. Both patients recovered with
oral trimethoprim-sulfamethoxazole and intramuscular injections of streptomycin.
36, 37
In addition,
in vitro resistance to imipenem and rifampin has been seen.
10
Contained casualty setting: The Working Group recommends parenteral antimicrobial therapy
when individual medical management is available. Antibiotics should be administered to all
patients for 10 days. Therapy may be switched to oral antimicrobials when clinically indicated.
Mass casualty setting: Replacement of parenteral antibiotics with oral antibiotics may be
indicated if the number of patients exceeds the medical care system’s capacity to administer
parenteral antibiotics.
S.F. Dept Public Health – Infectious Disease Emergencies PLAGUE, June 2008 Page9/13
PLAGUE - TREATMENT AND POST-EXPOSURE PROPHYLAXIS RECOMMENDATIONS
A
Contained Casualty Setting Mass Casualty
Setting
Post-Exposure
Prophylaxis
Duration of Rx
10 days 10 days 7 days
Preferred
Streptomycin, 1 gm IM q12 hrs or
Gentamicin,
B
5 mg/kg IM or IV q24 hrs, or 2
mg/kg loading dose followed by 1.7 mg/kg IM or IV
q8 hrs
Doxycycline, 100 mg orally twice daily or
Ciprofloxacin, 500 mg orally twice daily
Adult
Alternative
H
Doxycycline
5,6
, 100 mg IV q12 hrs or 200 mg IV
q24 hrs or
Ciprofloxacin, 400 mg IV q12 hrs or
Chloramphenicol
C
, 25 mg/kg IV q6 hrs (max 4
g/day)
Chloramphenicol
C
, 25 mg/kg orally 4 times
daily (max 4 g/day)
Preferred
Streptomycin, 15 mg/kg IM q12 hrs (max 2
g/day) or
Gentamicin
B
, 2.5 mg/kg IM or IV q8 hrs
Doxycycline
E,F
:
>
45 kg, give adult dosage
<45 kg, give 2.2 mg/kg orally twice daily (max
200 mg/day) or
Ciprofloxacin
E,G
, 20 mg/kg orally twice daily
(max 1 g/day)
Children
Alternative
H
Doxycycline
E,F
:
>
45 kg, give adult dosage
<45 kg, give 2.2 mg/kg IV q12 hrs (max 200
mg/day) or
Ciprofloxacin
E,G
, 15 mg/kg IV q12 hrs (max 1
g/day) or
Chloramphenicol
C,D
, 25 mg/kg IV q6 hrs (max 4
g/day)
Chloramphenicol
C,D
, 25 mg/kg orally 4 times
daily (max 4 g/day)
Preferred
Gentamicin
B
, 5 mg/kg IM or IV q24 hrs or 2 mg/kg
loading dose followed by 1.7 mg/kg IM or IV q8 hrs
Doxycycline
E,F
, 100 mg orally twice daily or
Ciprofloxacin
E
, 500 mg orally twice daily
Pregnant Women
Alternative
H
Doxycycline
E,F
, 100 mg IV q12 hrs or 200 mg IV
q24 hrs or
Ciprofloxacin
E
, 400 mg IV q12 hrs
Chloramphenicol
C,D
, 25 mg/kg orally 4 times
daily
For plague meningitis, pleuritis, or myocarditis: Chloramphenicol should be used for 21 days for conditions when
tissue penetration is important
4
. Irreversible marrow aplasia is rare (1 in 40,000 patients).
A
Treatment recommendations come from the Working Group of Civilian Biodefense and may not necessarily be
approved by the US Food and Drug Administration. Table adapted from JAMA. 2000;283:2281-2290.
B
Aminoglycoside doses must be further adjusted for newborns, and according to renal function.
C
Therapeutic concentration is 5 - 20 mcg/mL; concentrations >25 mcg/mL can cause reversible bone marrow
suppression.
D
According to the Working Group on Civilian Biodefense, children younger than 2 years of age should not receive
chloramphenicol due to risk of ‘gray baby syndrome’; however, the American Academy of Pediatrics has
recommended chloramphenicol as the drug of choice for plague meningitis in children.
E
Tetracycline and quinolone antibiotics are generally not recommended during pregnancy or childhood; however their
use may be indicated for life-threatening illness.
F
Ciprofloxacin may be preferred in pregnant women and children up to 8 years of age because of the known adverse
event profile of doxycycline (e.g., tooth discoloration).
G
Doxycycline may be preferred in children 8 years and older because of the adverse event profile of ciprofloxacin
(e.g., arthropathies).
H
Trimethoprim-sulfamethoxazole has been successfully used to treat plague; however the Working Group considers
this a second tier choice.
S.F. Dept Public Health – Infectious Disease Emergencies PLAGUE, June 2008 Page10/13
Post-Exposure Prophylaxis
Post-exposure prophylaxis is the administration of antibiotics after suspected exposure to plague
has occurred but before symptoms are present. If symptoms are present, see section above on
treatment. Persons thought to have had an infective exposure should receive post-exposure
prophylaxis. Infective exposures include household, hospital, or other close contact (less than 2
meters) with a person suspected or confirmed to have pneumonic plague who has received no
treatment, less than 48 hours of antimicrobial therapy, or more than 48 hours of antimicrobial
therapy without clinical improvement. Post-exposure prophylaxis may be recommended for
persons exposed to intentional aerosol releases. In such an event, public health authorities will
provide guidance. Regardless of whether post-exposure prophylaxis is recommended or taken,
persons potentially exposed should be observed for fever or cough for 7 days after exposure. Any
potentially-exposed person who develops a fever or cough should seek prompt medical attention
and begin treatment. Quarantine is not currently recommended.
4, 7, 9
Vaccination
Current killed whole cell vaccines have been in use for military personnel and have been shown to
generate cell-mediated responses lasting at least 15 years; however, they require repeat dosing
with adjuvants, have questionable protection against respiratory infections, and are reactogenic.
Vaccine production has been discontinued in the US. Microencapsulated subunit vaccines (of F1
and V proteins) requiring only single dose administration are under development and show the
most promise against aerosol exposures.
4, 38, 39
COMPLICATIONS AND ADMISSION CRITERIA
Whereas primary pneumonic plague results from direct inhalation of plague bacilli, secondary
pneumonic plague can manifest as a complication in patients with bubonic plague. Hematogenous
dissemination of Y. pestis results in plague septicemia,
which can be complicated by septic shock,
disseminated intravascular coagulation, necrosis of small vessels, and purpuric skin lesions. Plague
meningitis due to hematogenous seeding of the meninges occurs infrequently.
Patients with suspect or confirmed pneumonic or bubonic plague require hospitalization for
intravenous antibiotics, supportive care, and close monitoring for decompensation and signs of
toxemia.
S.F. Dept Public Health – Infectious Disease Emergencies PLAGUE, June 2008 Page11/13
These recommendations are current as of this document date. SFDPH will provide periodic updates as needed
and situational guidance in response to events (www.sfcdcp.org).
INFECTION CONTROL
Clinicians should notify local public health authorities, their institution’s infection control
professional and their laboratory of any suspected plague cases. Public health authorities may
conduct epidemiological investigations and implement disease control interventions to protect the
public. Infection control professionals will guide and enforce implementation of infection control
precautions within the healthcare setting. Laboratory personnel will take appropriate biosafety
precautions.
Although not highly contagious, plague can be transmitted person-to person via respiratory
droplets when the disease is end stage.
30
Both the Healthcare Infection Control Practices Advisory
Committee of the CDC and the Working Group on Civilian Biodefense recommend Droplet and
Standard precautions for patients with suspected or confirmed pneumonic plague. These
precautions should be maintained until 48 hours of appropriate antibiotics have been administered
AND the patient shows clinical improvement. Close contacts of pneumonic plague patients should
be identified, assessed for prophylaxis and monitored for symptoms. For patients with suspected
or confirmed bubonic plague or other non-pneumonic plague syndromes, Standard precautions
are recommended. Aerosol-generating procedures should be avoided if possible. Routine
laboratory procedures should be carried out under Biosafety level-2 conditions; however,
manipulation of cultures or other activities that may produce aerosol or droplets (e.g., centrifuging,
grinding, vigorous shaking, and animal studies) require Biosafety level -3 conditions.
4, 40
Decontamination
In general, environmental decontamination following an aerosol event has not been recommended,
since experts have estimated that an aerosol of Y. pestis organism would be infectious for only
about 1 hour.
4, 40
A recent study demonstrated that Y. pestis can survive on selected
environmental surfaces for at least several days; however the potential for re-aerosolization of
these organisms was not addressed.
41
Commercially available bleach or 0.5% hypochlorite
solution (1:10 dilution of household bleach) is considered adequate for cleaning and
decontamination. All persons exposed to an aerosol containing Y. pestis should be instructed to
wash body surfaces and clothing with soap and water.
PEARLS AND PITFALLS
Bubonic plague is not transmitted directly from one human to another in the absence of
lymph node suppuration and drainage. Persons with bubonic plague become more
infectious as Y. pestis organisms reach the lungs via hematogenous spread. Once
pneumonic plague develops, transmission occurs via direct contact with respiratory
secretions or inhalation of respiratory droplets.
S.F. Dept Public Health – Infectious Disease Emergencies PLAGUE, June 2008 Page12/13
Clinical clues pointing toward a diagnosis of primary pneumonic plague are sudden onset of
headache, malaise, and fever, fulminant pneumonitis with rapid progression from dry
cough to tachypnea, dyspnea, and productive cough, and in the late stage of disease,
hemoptysis with copious amounts of bright red sputum.
30
REFERENCES
1. Zhou D, Han Y, Yang R. Molecular and physiological insights into plague transmission, virulence and etiology.
Microbes Infect 2006;8:273-84.
2. Devignat R. Varietes de l'espece Pasteurella pestis. Nouvelle hyphothese. Bull WHO 1951;4:247-63.
3. Wren BW. The Yersinia—a model genus to study the rapid evolution of bacterial pathogens. Nat Rev
Microbiol 2003;1.
4. Inglesby TV, Dennis DT, Henderson DA, et al. Plague as a biological weapon: medical and public health
management. Working Group on Civilian Biodefense. Jama 2000;283:2281-90.
5. Davis CJ. Nuclear blindness: An overview of the biological weapons programs of the former Soviet Union and
Iraq. Emerg Infect Dis 1999;5:509-12.
6. Alibek K, Handelman S. Biohazard. New York, NY: Random House; 1999.
7. Borio LL. Plague as an Agent of Bioterrorism. In: Mandell GL, Bennett JE, Dolin R, eds. Principles and
Practice of Infectious Diseases. Philadelphia: Elselvier Churchill Livingstone; 2005:3601 - 5.
8. Franz DR, Jahrling PB, Friedlander AM, et al. Clinical recognition and management of patients exposed to
biological warfare agents. JAMA 1997;278:399 - 411.
9. Butler T, Dennis DT. Yersinia Species, Including Plague. In: Mandell GL, Bennett JE, Dolin R, eds. Principles
and Practice of Infectious Diseases. 6th ed. Philadelphia: Elsevier Churchill Linvingstone; 2005:2691 - 701.
10. Plague: Current, comprehensive information on pathogenesis, microbiology, epidemiology, diagnosis, and
treatment. Center for Infectious Disease Research and Policy, University of Minnesota, 2008. (Accessed at
http://www.cidrap.umn.edu/cidrap/content/bt/plague/biofacts/plaguefactsheet.html.)
11. Drancourt M, Houhamdi L, Raoult D. Yersinia pestis as a telluric, human ectoparasite-borne organism. Lancet
Infect Dis 2006;6:234 - 41.
12. Dennis DT, Hughes JM. Multidrug resistance in plague. N Engl J Med 1997;337:702-4.
13. Christie AB, Chen TH, Elberg SS. Plague in camels and goats: their role in human epidemics. J Infect Dis
1980;141:724-6.
14. Gage KL, Dennis DT, Orloski KA, et al. Cases of cat-associated human plague in the Western US, 1977-
1998. Clin Infect Dis 2000;30:893-900.
15. Palmer DL, Kisch AL, Williams RC, Jr., Reed WP. Clinical features of plague in the United States: the 1969-
1970 epidemic. J Infect Dis 1971;124:367-71.
16. Reed WP, Palmer DL, Williams RC, Jr., Kisch AL. Bubonic plague in the Southwestern United States. A
review of recent experience. Medicine (Baltimore) 1970;49:465-86.
17. von Reyn CF, Barnes AM, Weber NS, Quan T, Dean WJ. Bubonic plague from direct exposure to a naturally
infected wild coyote. Am J Trop Med Hyg 1976;25:626-9.
18. Butler T. Plague. In: Strickland GT, ed. Tropical medicine. Philadelphia, Pa: WB Saunders; 1991:408-16.
19. Perry RD, Fetherston JD. Yersinia pestis--etiologic agent of plague. Clin Microbiol Rev 1997;10:35-66.
20. Bin Saeed AA, Al-Hamdan NA, Fontaine RE. Plague from Eating Raw Camel Liver. Emerg Infect Dis
2005;11:1456 - 7.
21. Titball RW, Hill J, Lawton DG, Brown KA. Yersinia pestis and plague. Biochem Soc Trans 2003;31:104-7.
22. Koirala J. Plague: Disease, Management, and Recognition of Act of Terrorism. Infect Dis Clin North Am
2006;20:273 - 87.
23. Guiyoule A, Grimont F, Iteman I, Grimont PA, Lefevre M, Carniel E. Plague pandemics investigated by
ribotyping of Yersinia pestis strains. J Clin Microbiol 1994;32:634-41.
24. Begier EM, Asiki G, Anywaine Z, et al. Penumonic Plage Cluster, Uganda, 2004. Emerg Infect Dis
2006;12:460 - 7.
25. Anderson ET. Plague in the continental United States, 1900-76. Public Health Rep 1978;93:297-301.
26. CDC. Human plague--four states, 2006. Morbity and Mortality Weekly Report 2006;55:940-3.
27. Caten JL, Kartman L. Human plague in the United States, 1900-1966. Jama 1968;205:333-6.
28. Hull HF, Montes JM, Mann JM. Septicemic plague in New Mexico. J Infect Dis 1987;155:113-8.
29. Kaufmann AF, Boyce JM, Martone WJ. From the Center for Disease Control. Trends in human plague in the
United States. J Infect Dis 1980;141:522-4.
30. Kool JL. Risk of person-to-person transmission of pneumonic plague. Clin Infect Dis 2005;40:1166-72.
31. CDC. Notifiable Disease Tables. Morbity and Mortality Weekly Report 2008;57:636.
S.F. Dept Public Health – Infectious Disease Emergencies PLAGUE, June 2008 Page13/13
32. CDPH. Monthly Summary Report Selected Reportable Diseases: California Department of Public Health;
2008.
33. San Francisco Communicable Disease Report, 1986-2003. San Francisco Department of Public Health, 2005.
(Accessed at http://www.sfcdcp.org/publications.)
34. Annual Report of Communicable Diseases in San Francisco, 2004-2005. San Francisco Department of Public
Health, 2006. (Accessed at http://www.sfcdcp.org/publications.)
35. Annual Report of Communicable Diseases in San Francisco, 2006. San Francisco Department of Public
Health, 2008. (Accessed at http://www.sfcdcp.org/publications.)
36. Guiyoule A, Gerbaud G, Buchrieser C, et al. Transferable plasmid-mediated resistance to streptomycin in a
clinical isolate of Yersinia pestis. Emerg Infect Dis 2001;7:43-8.
37. Rasoamanana B, Rahalison L, Raharimanana C, Chanteau S. Comparison of Yersinia CIN agar and mouse
inoculation assay for the diagnosis of plague. Trans R Soc Trop Med Hyg 1996;90:651.
38. Elvin SJ, Eyles JE, Howard KA, et al. Protection against bubonic and pneumonic plague with a single dose
microencapsulated sub-unit vaccine. Vaccine 2006;24:4433 - 9.
39. Reed DS, Martinez MJ. Respiratory immunity is an important component of protection elicited by subunit
vaccination against pneumonic plague. Vaccine 2006;24:2283-9.
40. Siegel JD, Rhinehart E, Jackson M, Chiarello L, HICPAC. 2007 Guideline for Isolation Precautions: Preventing
Transmission of Infectious Agents in Healthcare Settings 2007. In: CDC; 2007:1 - 219.
41. Rose LJ, Donlan R, Banerjee SN, Arduino MJ. Survival of Yersinia pestis on Environmental Surfaces. Applied
and Environmental Microbiology 2003;69:2166 - 71.
S.F. Dept Public Health – Infectious Disease Emergencies SMALLPOX, July 2008 Page 1/14
Outline
Introduction
Epidemiology
Clinical Features
Differential Diagnosis
Laboratory Diagnosis
Treatment and Prophylaxis
Complications and Admission Criteria
Infection Control
Pearls and Pitfalls
References
Immediately report any suspected or
confirmed cases of smallpox to:
SFDPH Communicable Disease Control
(24/7 Tel: 415-554-2830)
- By law, health care providers must report suspected
or confirmed cases of smallpox to their local health
department immediately [within 1 hr].
- SFDPH Communicable Disease Control can
facilitate specialized testing and will initiate the public
health response as needed.
Also notify your:
Infection Control Professional
Clinical Laboratory
INTRODUCTION
Smallpox is caused by variola viruses, which are large, enveloped, single-stranded DNA viruses of
the Poxvirus family and the Orthopoxvirus genus. Variola major strains cause three forms of
disease (ordinary, flat type, and hemorrhagic), whereas variola minor strains cause a less severe
form of smallpox. Vaccination with vaccinia virus, another member of the Orthopoxvirus genus,
protects humans against smallpox because of the high antibody cross-neutralization between
orthopoxviruses.
1-4
The Working Group for Civilian Biodefense considers smallpox to be a dangerous potential
biological weapon because of “its case-fatality-rate of 30% or more among unvaccinated persons
and the absence of specific therapy.” Of the potential ways in which smallpox could be used as a
biological weapon, an aerosol release is expected to have the most severe medical and public
health outcomes because of the virus’ stability in aerosol form, low infectious dose, and high rate
of secondary transmission. A single case of smallpox would be a public health emergencey.
2
EPIDEMIOLOGY
Smallpox as a Biological Weapon
Smallpox has been used as a biological weapon in the distant past. More recently it has been a
focus of bioweapons research. In the 18
th
century, British troops in North America gave smallpox-
infected blankets to their enemies, who went on to suffer severe outbreaks of smallpox. Defecting
Russian scientists describe covert Russian operations during the 1970s and 1980s that focused on
SMALLPOX
S.F. Dept Public Health – Infectious Disease Emergencies SMALLPOX, July 2008 Page 2/14
bioweapons research and development including creation of more virulent smallpox strains and
development of missiles and bombs that could release smallpox.
2, 4, 5
Aerosol release of virus (such as into a transportation hub) would likely result in a high number of
cases. Other possibilities include use of "human vectors" (i.e., persons who have been deliberately
infected with smallpox) and use of fomites (e.g., contamination of letters sent through the mail).
2, 5
Smallpox is of concern as a biological weapon because:
2
much of the population (80%) is susceptible to infection
the virus has a low infectious dose and carries a high rate of morbidity and mortality
a vaccine that lacs significant side effects is not yet available for general use
experience has shown that introduction of the virus creates havoc and panic
An intentional release of smallpox would have the following characteristics:
6
Clustering in time: Multiple similarly presenting cases of fever and rash in mouth and on
face, arms, and legs generally 4 days after release
Naturally Occurring Smallpox
Reservoirs
The natural reservoir for smallpox was humans with disease; there was no chronic carrier state. In
1980, the World Health Organization (WHO) declared smallpox eradicated from the world and
recommended destruction or transfer all remaining stocks to one of two WHO reference labs, the
Centers for Disease Control and Prevention (CDC) in Atlanta, Georgia, and the former Institute of
Virus Preparations (later transferred to the Vector Institute) in Russia. Since eradication, there is
no natural reservoir for smallpox. Presently, smallpox is only officially found in these designated
WHO reference laboratories.
2, 5, 6
Mode of Transmission
Historically, humans were able to be infected in a number of ways:
1, 2, 6
Inhalation of droplet nulcei or aerosols originating from the mouth of smallpox-infected
humans
Direct contact with skin lesions or infected body fluids of smallpox-infected humans
Direct contact with contaminated clothing or bed linens
Worldwide occurrence
In 1967, a WHO-led international campaign of mass vaccination, surveillance and outbreak
containment was started in order to eradicate smallpox globally. In 1977, the last community-
acquired smallpox case was reported in Somalia, and in 1978, a laboratory accident in England
caused the last human case.
3
Occurrence in the United States
The last case of smallpox in the United States occurred in the Rio Grande Valley of Texas in 1949.
The risk of disease was low enough to end routine vaccination of the US population in 1971.
3
S.F. Dept Public Health – Infectious Disease Emergencies SMALLPOX, July 2008 Page 3/14
Vaccination is currently required for most military personnel and is recommended for select health
care and emergency workers, described below. Because of the relative frequency and seriousness
of vaccine-related complications and the low risk of smallpox outbreak in the United States, routine
vaccination is not recommended for the vast majority of healthcare workers or for the general U.S.
population.
7
In 2002, the CDC recommended pre-event vaccination for local smallpox response teams,
consisting of public health, medical, nursing, and public safety personnel, who would conduct
investigation and management of initial smallpox cases. As of July 31, 2004, 39,608 healthcare
workers and first responders had been vaccinated nationally.
6
CLINICAL FEATURES
Historically, smallpox has been divided into variola major and variola minor based on severity of
clinical disease. Variola major was more common and caused more severe disease relative to
variola minor. The case mortality was 15 to 45% for variola major and 1% for variola minor.
The infectious dose for smallpox is a few virions. The virus typically enters the body via respiratory
or oral mucosa and is carried by macrophages to regional lymph nodes from which a primary
asymptomatic viremia develops on the 3
rd
or 4
th
day after infection. The reticuloendothelial organs
are invaded and overwhelmed leading to a secondary viremia around the 8
th
to 12
th
day after
infection. Toxemia and fever onset follow. Seven to 17 days following infection, fever, malaise,
and extreme exhaustion begin. A maculopapular rash first presents on the face, mouth, pharynx,
and forearms and spreads to the trunks and legs. The rash progresses to a vesicular and pustular
stage (round and deeply embedded). Scabs form on the 8
th
day of the rash. Scars are formed
from sebaceous gland destruction and granulation tissue shrinking and fibrosis.
1-4, 6
Although most data supports communicability with rash onset, some low level of communicablity is
present prior to rash onset because viral shedding from oral lesions occurs during the 1 to 2 days
of fever preceding rash onset. However, secondary transmission peaks 3 to 6 days after fever
onset (1
st
week after rash onset), and 91.1% of secondary cases occurred by the 9
th
day after
fever onset.
8
The period of communicability ends when all the scabs have fallen off. Scabs are not
very infectious because the tight binding of the fibrin matrix retains the virions; however secondary
cases have been documented through transmission from direct contact with contaminated clothing
and bedding.
1-4, 6
Secondary bacterial infection and other organ involvement are uncommon. Encephalitis is a
possible complication. Mortality is most commonly associated with toxemia of circulating immune
complexes and soluble variola antigens and is seen in the second week of illness. Approximately
30 to 80% of unvaccinated close contacts will develop the disease. In addition, 3.5 to 6
transmissions per smallpox case are estimated.
6
S.F. Dept Public Health – Infectious Disease Emergencies SMALLPOX, July 2008 Page 4/14
Variola Major
Variola major is associated with the most severe disease, and presents as:
ordinary (80% or more of cases: mortality is 30% in unvaccinated and 3% in vaccinated
patients)
flat (4 to 6% of cases: mortality is 95% in unvaccinated and 66% in vaccinated patients)
hemorrhagic (2 to 3% of cases: mortality is 99% in unvaccinated and 94% in vaccinated
patients)
modified (13% of cases and low risk of death)
variola sine eruptione (30 to 50% of vaccinated contacts of smallpox and low risk of death)
CLINICAL FEATURES: ORDINARY VARIOLA MAJOR.
1-4, 6
Incubation Period
10-13 days (range 7-19 days)
Transmission
Inhalation of droplet nuclei or aerosols originating from the mouths of
smallpox-infected humans
Direct contact with skin lesions or infected body fluids of smallpox-infected
humans
Direct contact with contaminated clothing or bed linens
Signs and Symptoms
Prodromal phase
2-4 days of fever, chills, headache, backache, and often GI symptoms
Rash phase
Enanthem (papules, vesicles, then ulcers) of oropharyngeal mucosa beginning
1 day before skin lesions appear
First skin lesions ("herald spots") are often on the face
Lesions spread centrifugally: trunk to proximal extremities to distal extremities
Palms and soles are usually involved, and truncal rash is usually sparse
Lesion progression: maculopapular (days 1-2), vesicular (days 3-5), pustular
(days 7-14)
Vesicles and pustules are frequently umbilicated
Pustules can be like small, embedded hard balls or “shotty”
Lesions tend to progress at same rate
Lesions may be discrete, semiconfluent, or confluent
Lesions are typically painful and cause pitted scars as they heal
Lesions gradually scab over during days 13-18
Progression and
Complications
Viral bronchitis or pneumonitis
Third spacing of fluid with resulting electrolyte and renal abnormalities
Skin desquamation
Secondary bacterial infection, particularly skin and pulmonary
Spontaneous abortion, stillbirth
Rarely: blindness, keratitis, corneal ulceration, encephalitis, osteomyelitis or
arthritis, orchitis
Death may occur during 2
nd
week of illness, from high-level viremia and
circulating immune complexes
Laboratory Findings
Lymphocytopenia and/or granulocytopenia
GI, gastrointestinal
S.F. Dept Public Health – Infectious Disease Emergencies SMALLPOX, July 2008 Page 5/14
Other forms of smallpox caused by variola major infection include:
Flat-type smallpox (also known as malignant smallpox) occurred in about 4 to 6% of cases
and more frequently in children. It is associated with a late, deficient cellular immune response. It
is characterized by a short incubation period, prostrating prodromal illness, severe systemic toxicity
and high mortality (90-97%). The lesions do not progress to the pustular stage, instead remaining
soft, velvety and flattened. If the patient survives, the lesions will resolve by desquamation
without scabs or scarring.
Hemorrhagic smallpox occurred in about 2 to 3% of cases. Pregnant women are highly
susceptible. Similar to flat-type smallpox, it is associated with a defective immune response. It is
characterized by a short incubation period, prostrating prodromal illness, severe systemic toxicity,
and high mortality (96%). The rash begins as a dusky erythema, followed by extensive petechiae,
mucosal hemorrhage, and intense toxemia. Thrombocytopenia and coagulopathy may be present.
These patients usually died during week 1 of illness, often before the development of the typical
pox lesions.
Modified smallpox occurred in about 13% of cases. It occurred in persons with some immunity.
The pre-eruptive illness is typical in duration and severity as ordinary smallpox; however, during
the eruption, fever is absent and the skin lesions are superficial, pleomorphic, fewer in number,
and evolve rapidly.
Variola sine eruption occurred in about 30 to 50% of vaccinated contacts of smallpox cases. It is
characterized by a sudden onset of fever, headache, occasional backache which resolves within 48
hours, influenza-like symptoms and no rash.
Variola Minor
Variola minor, caused by different strains of variola, is a milder form of smallpox. Compared with
variola major, there are milder constitutional symptoms, discrete lesions that evolve a bit more
rapidly, lower rates of hemorrhagic disease, and only rare fatal outcomes (<1%). The illness may
be difficult to distinguish clinically from modified smallpox and variola without eruption. In the
1890s, variola minor spread from South Africa to Florida. In the early 1900s, variola minor
became prevalent in the United States, Latin America, and Europe.
DIFFERENTIAL DIAGNOSIS
The characteristic features of smallpox need to be differentiated from other illnesses that present
with vesicular or pustular rash. One disease that could be confused with smallpox is chickenpox.
These may be differentiated clinically, as follows:
S.F. Dept Public Health – Infectious Disease Emergencies SMALLPOX, July 2008 Page 6/14
CLINICAL DIFFERENTIATION OF VARIOLA VS. VARICELLA
9
Feature Variola Varicella
Prodrome
Duration: 2-4 days
Fever, chills, headache, backache, often
GI symptoms
Commonly does not occur
If present, mild symptoms and duration
of 1 day
Rash
Distribution
Centrifugal: more dense on face and
distal extremities
Frequently involves palms and soles
More involvement of back than abdomen
Centripetal: more dense on trunk
Spares palms and soles
Back and abdomen equally involved
Lesion
Evolution
Usually appear on oropharyngeal mucosa
first, then all over within 1-2 days
Progress at same rate; at any point in
time, lesions are at same stage of
evolution
Lesions progress slowly (7-14 days) from
macules to papules to vesicles to
pustules to scabs
Lesions appear in crops
At any point in time, crops of lesions are
at different stages of evolution
Lesions progress quickly (1-2 days) from
macules to papules to vesicles to scabs
Lesion
Attributes
May be semiconfluent or confluent
Deep
May be umbilicated
Often painful; pruritic only as scabs
Usually discrete
Superficial
Rarely found of palms and soles
Do not umbilicate or dimple
Typically painless; intensely pruritic
GI, gastrointestinal
Monkeypox is another disease that could be confused with smallpox. In 2003, an outbreak of
monkeypox, associated with prarie dog contact, took place in the midwestern United States.
Monkeypox in humans presents similarly to ordinary smallpox. However, monkeypox is milder and
has prominent lymphadenopathy and a shorter duration of rash.
The CDC has outlined criteria for determining the risk of smallpox when evaluating patients with
generalized vesicular or pustular rash:
9, 10
Risk of Smallpox in Patients with Generalized Vesicular or Pustular Rash
High
All three major criteria present:
a) Febrile prodrome 1-4 days before rash onset, with fever >101°F, plus 1 or more of the
following: prostration, headache, backache, chills, vomiting, severe abdominal pain
b) Classic smallpox lesions present (vesicles or pustules that are deep-seated, firm or hard,
round, and well-circumscribed; sharply raised and feel like BB pellets under the skin; may
become umbilicated or confluent as they evolve)
c) Lesions on any one part of the body are in the same stage of development
Moderate
Febrile prodrome as in (a) above, plus either (b) or (c) above
S.F. Dept Public Health – Infectious Disease Emergencies SMALLPOX, July 2008 Page 7/14
OR
Febrile prodrome as in (a) above, plus at least four of the following minor criteria:
Centrifugal distribution
First lesions appeared on the oral mucosa/palate, face, or forearms
Patient appears toxic or moribund
Slow evolution of lesions from macules to papules to pustules over several days
Lesions on the palms and soles
Low
No viral prodrome
OR
Febrile prodrome as in (a) above, plus < 4 minor criteria above
Additional considerations in the differential diagnosis of smallpox include:
Macular/papular stage
measles
scarlet fever
rubella
Vesicular/pustular stage:
disseminated herpes zoster
disseminated herpes simplex
Mmolluscum contagiosum
bullous pemphigoid
impetigo (Streptococcus, Staphylococcus)
human monkey pox
Either stage:
erythema multiforme major
- (Stevens-Johnson syndrome)
miscellaneous drug eruptions
secondary syphilis
enteroviral infection (hand, foot &
mouth disease)
chickenpox
contact dermatitis
generalized vaccinia
acne
scabies/insect bites
Hemorrhagic smallpox may resemble:
meningococcemia
rickettsial infections
Gram-negative septicemia
Flat-type smallpox may resemble:
hemorrhagic chickenpox
S.F. Dept Public Health – Infectious Disease Emergencies SMALLPOX, July 2008 Page 8/14
If you are testing or considering testing for
smallpox, you should:
IMMEDIATELY notify
SFDPH Communicable Disease Control
(24/7 Tel: 415-554-2830).
SFDPH can authorize and facilitate testing, and
will initiate the public health response as needed.
Inform your lab that smallpox is under
suspicion.
These recommendations are current as of this document date. SFDPH will provide periodic updates as needed
and situational guidance in response to events (www.sfcdcp.org).
LABORATORY DIAGNOSIS
The diagnosis of smallpox requires a high index of
suspicion because the disease has been eradicated
and its clinical presentation is similar to other pox
viruses. Routine laboratory findings for specific
clinical presentations of smallpox are listed in the
clinical features table. Radiographic findings do
not assist in identification of smallpox.
Diagnosis of smallpox will be clinical initially, but
followed by laboratory confirmation. Once
smallpox has been confirmed in a geographic
area, additional cases can be diagnosed clinically, and specimen testing can be reserved for specific
cases in which the clinical presentation is unclear or to assist with law enforcement activities.
Clinicians should use the CDC-developed tools to assess the likelihood that patients with acute
generalized vesicular or pustular rash illnesses have smallpox.
9
CDC has also developed algorithms for
laboratory evaluation of suspect smallpox cases based on the likelihood of disease.
10
If a patient is
determined to be at high risk for smallpox, clinicians should call their local public health authorities
immediately and obtain photos of the patient. Public health will provide guidance on specimen
collection and packaging and will facilitate transport of specimens to the appropriate public health
laboratory.
Multiple tests will be used to evaluate for smallpox. Polymerase chain reaction (PCR) testing will be an
important method; however, other methods will also be used including: electron microscopic
examination of vesicular or pustular fluid or scabs, direct examination of vesicular or pustular material
looking for inclusion bodies (Guarnieri’s bodies), culture on egg chorioallantoic membrane, tissue
culture, strain analysis with a restriction fragment length polymorphism assay, and serology.
Definitive laboratory identification and characterization of the variola virus requires several days.
TREATMENT AND PROPHYLAXIS
Treatment
The management of confirmed or suspected cases of smallpox consists of supportive care, with careful
attention to electrolyte and volume status, and ventilatory and hemodynamic support. General
supportive measures include ensuring adequate fluid intake (difficult because of the enanthem),
alleviation of pain and fever, and keeping skin lesions clean to prevent bacterial superinfection.
1-4, 6
Currently there are no FDA approved antiviral agents with proven activity against smallpox in humans.
S.F. Dept Public Health – Infectious Disease Emergencies SMALLPOX, July 2008 Page 9/14
Antiviral agents that have shown some activity in vitro against poxviruses may be available from the
CDC under an investigational protocol. ST-246 is a novel agent that is currently undergoing safety and
efficacy testing.
6, 11
Additionally, cidofovir, a nucleoside analogue DNA polymerase inhibitor, might be
useful if administered within 1-2 days after exposure; however, there is no evidence that it would be
more effective than vaccination, and it has to be administered intravenously and causes renal toxicity.
Immunity from prior vaccination
Protection from smallpox is estimated to last between 11.7 to 28.4 years after primary vaccination
and longer for variola minor than for variola major. Those who were previously vaccinated may retain
some protection that could decrease the severity of the disease and allow for greater mobility thereby
complicating public health response.
12
Postexposure prophylaxis
Postexposure prophylaxis for smallpox is the administration of vaccinia vaccine after suspected
exposure to smallpox has occurred but before symptoms are present. Immunity generally develops
within 8 to 11 days after vaccination with vaccinia virus. Because the incubation period for smallpox
averages about 12 days, vaccination within 4 days may confer some immunity to exposed persons and
reduce the likelihood of a fatal outcome. Postexposure vaccination may be particularly important for
those vaccinated in the past, provided that revaccination is able to boost the anamnestic immune
response. In addition to vaccination, exposed persons should be monitored for symptoms.
Temperature should be checked once a day, preferably in evening, for 17 days after exposure for
fever (over 38°C).
2-4, 6, 7
If a case or cases of smallpox occur, public health authorities will conduct surveillance and implement
containment strategies. Ring vaccination will be important and includes identification of contacts of
cases and provision of prophylaxis and guidance on monitoring for symptoms. Large-scale voluntary
vaccination may be offered to low-risk populations to supplement and address public concerns.
Vaccine Supply, Administration, and Efficacy
The smallpox vaccine used in the United States (formerly Dryvax, now ACAM2000) is a lyophilized
(freeze-dried) preparation of live attenuated vaccinia virus, an Orthopoxvirus closely related to
cowpox that induces antibodies that are protective against smallpox. The ACAM2000 uses vaccinia
virus derived from the Dryvax vaccine via plaque purification cloning. The virus is then grown in
African green monkey (Vero) cells. The ACAM2000 preparation also contains HEPES, human serum
albumin, mannitol and trace amounts of neomycin and polymixin B. The diluent contains glycerin and
a phenol preservative.
13
Production of the Dryvax vaccine stopped in the 1980s. Acambis currently makes the ACAM2000
vaccine which received FDA approval in September 2007. By that time 192.5 million doses of
ACAM2000 were already in the United States stockpile. All lots of Dryvax vaccine expired February 20,
2008, and were destroyed by March 31, 2008.
6, 14
Technique. The Dryvax vaccine should be administered by trained, vaccinated personnel using a
bifurcated needle that is stroked against the skin until blood appears. Vaccinees are instructed to
S.F. Dept Public Health – Infectious Disease Emergencies SMALLPOX, July 2008 Page 10/14
keep the site dry and covered, to avoid touching the site, and to thoroughly launder or carefully
discard any materials that come into contact with the site. Should smallpox vaccination be
deemed necessary, it will be coordinated by local, state and federal health agencies. For
additional information on vaccine administration, see
http://www.bt.cdc.gov/agent/smallpox/vaccination.
7
Vaccine Contraindications and Complications
The ACAM2000 and Dryvax vaccines have similar safety profiles.
14
Both have serious complications.
Likelihood of adverse effects is 3 to 4-fold higher in infants and in primary recipients. Based on the
U.S. Vaccine Adverse Events Reporting System of recently vaccinated people, there was a rate of 26.4
deaths per 10,000 vaccinees. Adverse events included the following: 33% cardiac, 25% nonspecific
chest pain, 21% neurological, 14% infection, 3% malignancy, 3% pulmonary (noninfectious), and 1%
normal vaccination response.
13, 15, 16
Vaccination during the pre-exposure period is contraindicated for certain persons. During a
smallpox emergency, however, all contraindications would be reviewed in the context of
the risk of smallpox exposure, and updated recommendations would be issued by public
health authorities. Current contraindications to vaccination are as follows (see
www.bt.cdc.gov/agent/smallpox/vaccination for further description):
7, 13
past or present eczema or atopic dermatitis (risk of eczema vaccinatum)
other acute or chronic exfoliative skin conditions (e.g. burns, impetigo, chicken pox,
contact dermatitis, shingles, herpes, severe acne, psoriasis), until the condition resolves
immunodeficiency states, due to disease or treatment of disease
pregnancy (vaccination may offer partial protection for mother, but increases risk of fetal
vaccinia)
breastfeeding
hypersensitivity to vaccine components
under 18 years of age in nonemergency situations
having a household contact who is immunodeficient, who has past or present eczema or
atopic dermatitis, or who has an acute, chronic, or exfoliative skin condition
physician-diagnosed cardiac disease, or 3 or more major risk factors for cardiac disease
Well-documented adverse reactions to vaccination
are listed below:
1, 2, 7, 13
tenderness, erythema, or other localized reactions at the injection site
systemic symptoms of fever, malaise, myalgias, local lymphadenopathy
dermatologic reactions, including erythema multiforme and Stevens-Johnson syndrome,
urticaria, exanthems, contact dermatitis, and erythematous papules
secondary bacterial infections at injection site
focal and generalized suppurative folliculitis (without evidence of viral infection; may be
mistaken for generalized vaccinia)
inadvertent autoinoculation of another body site (most common sites are face, eyelid,
nose, mouth, genitalia, rectum)
generalized vaccinia: vesicles or pustules appearing distant from the vaccination site
S.F. Dept Public Health – Infectious Disease Emergencies SMALLPOX, July 2008 Page 11/14
eczema vaccinatum: localized or dissemination of vaccinia virus; usually mild but may be
severe and fatal
vaccinia keratitis
progressive vaccinia: progressive necrosis in vaccination area, often with metastatic sites;
can be severe and fatal
postvaccinial encephalitis
fetal vaccinia: occurs when mother is vaccinated during during pregnancy; usually results
in premature birth or miscarriage
myopericarditis, identified among military personnel vaccinated between December 2002
and December 2003
death: 1.1 deaths per 1 million primary vaccine recipients
contact vaccinia: transmission of vaccinia virus from newly vaccinated persons to
susceptible unvaccinated contacts
The primary therapy for adverse reactions to smallpox vaccination is vaccinia immunoglobulin
(VIG).
7
However VIG is contraindicated in vaccinia keratitis and provides no benefit in
postvaccinial encephalitis. VIG is manufactured from the plasma of persons vaccinated with
vaccinia vaccine. An intravenous preparation (VIGIV) was recently licensed by the FDA.
17
Cidofovir
and topical ophthalmic antiviral agents are also recommended by some experts.
7
Cidofovir use
requires an Investigational New Drug (IND) protocol, and topical ophthalmic agent use is off-label.
COMPLICATIONS AND ADMISSION CRITERIA
Before smallpox was eradicated worldwide, viral bronchitis and pneumonitis were the most
frequent complications of ordinary-type smallpox. Cutaneous complications included
desquamation, massive subcutaneous fluid accumulation with electrolyte abnormalities and renal
failure, or, less commonly, secondary bacterial infection of smallpox lesions. Infrequently,
smallpox patients experienced encephalitis, osteomyelitis, corneal ulceration, or ocular keratitis.
Ordinary-type smallpox with confluent lesions, rather than discrete lesions, carried a much higher
risk of massive exfoliation, tissue destruction, bacterial sepsis, and death. Hemorrhagic-type and
flat-type smallpox were nearly always fatal.
1-4, 6
Many patients do not require hospitalization. Those with discrete lesions, nonhemorrhagic and non-
flat-type, are less likely to become critically ill or require much supportive care and can be more
easily managed outside the hospital. These people should be isolated and monitored at home or in
a nonhospital facility, and smallpox vaccination should be provided to caregivers and household
members. Patients with evidence of severe disease or presentations that suggest progression to
severe disease is likely should be considered for admission to a negative-pressure environment
with strict maintenance of Airborne Precautions.
2, 6
S.F. Dept Public Health – Infectious Disease Emergencies SMALLPOX, July 2008 Page 12/14
These recommendations are current as of this document date. SFDPH will provide periodic updates as needed
and situational guidance in response to events (www.sfcdcp.org).
INFECTION CONTROL
Clinicians should notify local public health authorities, their institution’s infection control
professional, and their laboratory of any suspected smallpox cases. Public health authorities may
conduct epidemiological investigations and will implement disease control interventions to protect
the public. Infection control professionals will implement infection control precautions within the
healthcare setting. Laboratory personnel should take appropriate safety precautions.
Smallpox is transmissible from person to person by exposure to respiratory secretions and by
direct contact with pox lesions and fomites. Airborne and Contact Precautions in addition to
Standard Precautions should be implemented for patients with suspected smallpox and until all
scabs have separated. Healthcare workers caring for patients with suspected smallpox should be
vaccinated immediately.
18, 19
Decontamination
Survival of the virus in the environment is inversely proportional to temperature and humidity. All
bedding and clothing of smallpox patients should be minimally handled to prevent re-aerosolization
and autoclaved or laundered in hot water with bleach. Standard disinfection and sterilization
methods are deemed to be adequate for medical equipment used with smallpox patients and
cleaning surfaces and rooms potentially contaminated with the virus. Airspace decontamination
(fumigation) is not required.
2, 19
PEARLS AND PITFALLS
1. The CDC has developed a number of clinical diagnostic tools to assist with the visual
recognition, differential diagnosis, and initial management of suspected smallpox.
These resources are available at: http://www.bt.cdc.gov/agent/smallpox/index.asp.
20
2. Hemorrhagic smallpox is rare but can be confused with invasive meningococcal
disease, rickettsial infections, or gram-negative sepsis because of the patient’s ill
appearance, petechial and purpuric lesions, and hemorrhagic manifestations.
3. Smallpox is most often transmitted through direct contact with respiratory droplets as
a result of close (within 2 meters) or face-to-face contact. Viruses can also travel over
greater distances as airborne particles, particularly in cases with coughing.
Transmission has occasionally been linked to fomites carried on clothing or bedding
that has been contaminated by dried respiratory secretions or draining skin lesions.
S.F. Dept Public Health – Infectious Disease Emergencies SMALLPOX, July 2008 Page 13/14
4. Since 2003, many health departments have established smallpox preparedness teams
consisting of providers who have been pre-vaccinated against smallpox who can
asssist with the response to a suspected case of smallpox.
REFERENCES
1. Damon I. Orthopoxviruses: Vaccinia (Smallpox Vaccine), Variola (Smallpox),
Monkeypox, and Cowpox. In: Mandel GL, Bennett JE, Dolin R, eds. Principles and
practice of infectious diseases. 6 ed. New York: Churchill Livingstone; 2005:1742-1751.
2. Henderson DA, Inglesby TV, Bartlett JG, et al. Smallpox as a biological weapon:
medical and public health management. Working Group on Civilian Biodefense. Jama.
Jun 9 1999;281(22):2127-2137.
3. Breman JG, Henderson DA. Diagnosis and management of smallpox. N Engl J Med. Apr
25 2002;346(17):1300-1308.
4. Moore ZS, Seward JF, Lane JM. Smallpox. Lancet. Feb 4 2006;367(9508):425-435.
5. Rotz LD, Cono J, Damon I. History of Smallpox and Bioterrorism. In: Mandel GL,
Bennett JE, Dolin R, eds. Principles and practice of infectious diseases. 6 ed. New York:
Churchill Livingstone; 2005:3612-3617.
6. CIDRAP. Smallpox: Current, comprehensive information on pathogenesis, microbiology,
epidemiology, diagnosis, treatment, and prophylaxis. Center for Infectious Disease
Research and Policy, University of Minnesota. Available at:
http://www.cidrap.umn.edu/cidrap/content/bt/smallpox/biofacts/smllpx-summary.html.
7. CDC. Smallpox Vaccination, Information for Health Professionals. Centers for Disease
Control and Prevention. Available at: www.bt.cdc.gov/agent/smallpox/vaccination.
8. Nishiura H, Eichner M. Infectiousness of smallpox relative to disease age: estimates
based on transmission network and incubation period. Epidemiol Infect. Oct
2007;135(7):1145-1150.
9. CDC. Smallpox: Diagnosis/Evaluation. Centers for Disease Control and Prevention.
Available at: http://www.bt.cdc.gov/agent/smallpox/diagnosis/.
10. CDC. Acute, Generalized Vesicular or Pustular Rash Illness Testing Protocol in the
United States. Centers for Disease Control and Prevention. November 14. Available at:
http://emergency.cdc.gov/agent/smallpox/diagnosis/pdf/poxalgorithm11-14-07.pdf.
11. Jordan R, Tien D, Bolken TC, et al. Single-dose safety and pharmacokinetics of ST-246,
a novel orthopoxvirus egress inhibitor. Antimicrob Agents Chemother. May
2008;52(5):1721-1727.
12. Nishiura H, Schwehm M, Eichner M. Still protected against smallpox? Estimation of the
duration of vaccine-induced immunity against smallpox. Epidemiology. Sep
2006;17(5):576-581.
13. FDA. Produce Approval Information: Smallpox (Vaccinia) vaccine. U.S. Food and Drug
Administration. August 31. Available at:
http://www.fda.gov/cber/products/acam2000.htm
.
14. CDC. Notice to Readers: Newly Licensed Smallpox Vaccine to Replace Old Smallpox
Vaccine. MMWR. February 29 2008;57(08):207-208.
S.F. Dept Public Health – Infectious Disease Emergencies SMALLPOX, July 2008 Page 14/14
15. Casey CG, Iskander JK, Roper MH, et al. Adverse events associated with smallpox
vaccination in the United States, January-October 2003. Jama. Dec 7 2005;294(21):2734-
2743.
16. Sejvar JJ, Labutta RJ, Chapman LE, Grabenstein JD, Iskander J, Lane JM. Neurologic
adverse events associated with smallpox vaccination in the United States, 2002-2004.
Jama. Dec 7 2005;294(21):2744-2750.
17. FDA. FDA approves new plasma-derived product to treat complications of smallpox
vaccination. U.S. Food and Drug Administration. February 18. Available at:
http://www.fda.gov/bbs/topics/ANSWERS/2005/ANS01341.html
.
18. CDC. Guide F: Environmental Control of Smallpox Virus, Smallpox Response Plan.
Centers for Control and Prevention. March 20. Available at:
www.bt.cdc.gov/agent/smallpox/response-plan/files/guide-f.doc
.
19. Siegel JD, Rhinehart E, Jackson M, Chiarello L, HICPAC. Guideline for Isolation
Precautions: Preventing Transmission of Infectious Agents in Healthcare Settings, 2007.
Centers for Disease Control and Prevention. Available at:
http://www.cdc.gov/ncidod/dhqp/gl_isolation.html.
20. CDC. Smallpox. Centers for Disease Control and Preventino. Available at:
http://www.bt.cdc.gov/agent/smallpox/index.asp.
S.F. Dept Public Health – Infectious Disease Emergencies TULAREMIA, July 2008 Page 1/13
Outline
Introduction
Epidemiology
Clinical Features
Differential Diagnosis
Laboratory Diagnosis
Treatment and Prophylaxis
Complications
Infection Control
Pearls and Pitfalls
References
Immediately report any suspected or
confirmed cases of tularemia to:
SFDPH Communicable Disease Control
(24/7 Tel: 415-554-2830)
- By law, health care providers must report suspected or
confirmed cases of tularemia to their local health
department immediately [within 1 hr].
- SFDPH Communicable Disease Control can facilitate
specialized testing and will initiate the public health
response as needed.
Also notify your:
Infection Control Professional
Clinical Laboratory
INTRODUCTION
Tularemia is a zoonotic disease caused by Francisella tularensis, a non-sporulating, non-motile,
aerobic, gram-negative coccobacillus. There are multiple subspecies of F. tularensis, with the
biovars tularensis (type A) and holarctica (type B) occurring most commonly in the United States.
The clinical syndromes caused by tularemia depend on the route of infection and subspecies of the
infecting organism. Tularemia is highly infectious, requiring inhalation or inoculation of as few as 10
to 50 organisms to cause disease.
1
Although its virulence factors are not well characterized, type A
is generally thought to be the more virulent subspecies.
2-4
However, the virulence of type A
subspecies may vary between geographic regions within the US, with the mid-western and eastern
states having more severe infections.
5, 6
The Working Group for Civilian Biodefense considers tularemia to be a dangerous potential
biological weapon because of its “extreme infectivity, ease of dissemination, and its capacity to
cause illness and death.” Of the potential ways that F. tularensis could be used as a biological
weapon, an aerosol release is expected to have the most severe medical and public health
outcomes.
3
EPIDEMIOLOGY
Tularemia as a Biological Weapon
Weaponized F. tularensis was developed and stockpiled by the US military, though the supply was
destroyed in the 1970’s. The Soviet Union is reported to have developed antibiotic- and vaccine-
resistant strains of weaponized F. tularensis.
3
TULAREMIA JULY 2008
S.F. Dept Public Health – Infectious Disease Emergencies TULAREMIA, July 2008 Page 2/13
Experts believe that an aerosolized release is the most likely intentional use of F. tularensis
organisms. Exposure to aerosolized F. tularensis would cause:
2
Via inhalation:
o primary pneumonic tularemia (majority of patients)
o typhoidal tularemia (nonspecific febrile illness of varying severity)
o oropharangeal tularemia
Via contact with eyes: oculoglandular tularemia
Via contact with broken skin: glandular or ulceroglandular disease
An intentional release of tularemia would have the following characteristics:
Multiple similarly presenting cases clustering in time:
o acute non-specific febrile illness with onset 3 to 5 days after the initial release
(range 1-14 days)
o community-acquired atypical pneumonia unresponsive to typical antimicrobials
Atypical host characteristics: unexpected, unexplained cases of acute illness in previously
healthy persons who rapidly develop pleuropneumonia and systemic infection, especially if
patients develop pleural effusions
and hilar lymphadenopathy
Unusual geographic clustering: multiple cases in an urban area, where naturally occurring
tularemia is not endemic
Absence of risk factors: patients lack tularemia exposure risk factors (e.g. outdoor field
work or recreational activity, contact with tissues of potentially infected animals)
Intentionally released F. tularensis strains may be altered to have enhanced virulence or
antimicrobial resistance.
3
Naturally Occurring Tularemia
Reservoir
The natural reservoirs for F. tularensis are small and medium-sized mammals. In the United
States these are primarily lagomorphs (rabbits, hares) but may include beaver, squirrels,
muskrats, field voles, and rats. Incidental hosts include some species of mammals (e.g. humans,
cats, dogs, cattle), birds, fish, and amphibians. Organisms can survive for weeks in moist
environments, including water, mud, and decaying animal tissue.
2
There is some evidence that the
protozoa Acanthamoeba castellanii, may be an important reservoir for F. tularensis.
7
Mode of Transmission
The primary vectors for infection in the United States are ticks (dog ticks, wood ticks) and flies,
such as the deerfly. Humans become infected by a number of mechanisms:
3
bites by infected arthropods (majority of cases)
contact with infectious animal tissues or fluids, during for example hunting or butchering
ingestion of contaminated food, water, or soil
inhalation of infectious aerosols, including aerosols generated during landscaping activities
(e.g., lawn mowing, using a power blower, and brush cutting)
S.F. Dept Public Health – Infectious Disease Emergencies TULAREMIA, July 2008 Page 3/13
exposure in the laboratory (accidental inhalation of aerosol, direct contact with an
infectious specimen including accidental parenteral inoculation, or ingestion)
Tularemia is not spread from person to person.
Worldwide Occurrence
Worldwide, human cases of tularemia occur throughout North America, Europe, and Asia.
Infections with type A strain are generally only seen in North America. Within Europe and Asia, the
greatest numbers of human cases are reported in Scandinavian countries and countries of the
former Soviet Union.
3
Recent significant outbreaks of tularemia in humans include: Sweden (2000,
2003, 2006), Kosovo (2002), France (2004), Turkey (2004-2005), Bulgaria (1997-2005) and Spain
(2007).
2, 8-10
United States Occurrence
Nationwide, incidence of tularemia has declined from approximately 2000 annually reported cases
during the first half of the 20
th
century to an average of 124 cases per year during the 1990s.
Most cases occur in rural or semirural environments, during the summer months, with the greatest
number of cases occurring in Missouri, Oklahoma, South Dakota, Montana, and Martha’s Vineyard,
Massachusetts. From 1990 to 2000, incidence of tularemia in the United States was highest in
children 5-9 years and adults 75 years and older. Regardless of age, males had a higher incidence
of tularemia, potentially because of participation in activities more likely to cause exposures, such
as hunting, trapping, butchering, and farming.
11
Recent significant outbreaks include:
In 1978 and 2000, outbreaks of the rare primary pneumonic tularemia occurred on
Martha’s Vineyard, Massachusetts. These represent the only outbreaks of primary
pneumonic tularemia in the United States. Additional cases of tularemia have been
reported each year in Martha’s Vineyard (2000-2006). Exposure is most likely from
breathing infectious aerosols generated during landscaping activities.
12
The reservoir in
these outbreaks is still unclear, but may involve skunks and raccoons.
13
In 2002, tularemia was responsible for a die-off of several hundred prairie dogs caught in
the wild in South Dakota and then commercially distributed widely throughout the USA.
One human case occurred in an animal handler who cared for the infected animals.
14
In 2003, low levels of F. tularensis were identified in a biodetection air-monitoring system
in Houston, Texas. No human cases occurred. An investigation supported contamination of
the filters by naturally occurring F. tularensis organisms from an unidentified environmental
reservoir.
15
Occurrence in California and San Francisco
From 2000 to 2007, 16 cases of plague were reported in California, and one of these occurred in
San Francisco.
16-19
CLINICAL FEATURES
Human tularemia occurs in six recognized forms, determined primarily by route of infection.
Tularemia infection can range from mild to severe clinical illness and can be life-threatening.
S.F. Dept Public Health – Infectious Disease Emergencies TULAREMIA, July 2008 Page 4/13
Overall case-fatality rates have declined from 5-15% in the pre-antibiotic era to approximately 2%
currently. Mortality was historically much higher with pneumonic and typhoidal tularemia, with
case-fatality as high as 30-60% if untreated.
3
Administration of appropriate antibiotic treatment
typically leads to general symptom improvement within 24-48 hours. Recognition of tularemia as a
potential etiologic agent is critical, because poor outcomes have been associated with delays in
seeking care and/or instituting effective antimicrobial treatment.
20
Pneumonic Tularemia
Pneumonic tularemia causes the most severe disease, and presents as a non-specific febrile illness
with progression to pleuropneumonitis and systemic infection.
PNEUMONIC TULAREMIA
Incubation Period
3-5 days (range 1-14 days)
Transmission
Inhalation of contaminated aerosols
Secondary hematogenous spread to the lung
Signs and Symptoms
Initial presentation as atypical CAP unresponsive to routine antibiotic therapy,
which can progress slowly OR rapidly to severe disease
Fever (abrupt onset), headache, cough, minimal or no sputum production,
dyspnea, pleuritic chest pain, myalgias (often prominent in lower back),
bronchiolitis and/or pharyngitis may be present
Generalized maculopapular rash with progression to pustules or erythema-
nodosum type rash occurs in 20%
Nausea, vomiting, diarrhea is not uncommon
Hemoptysis (not common)
Progression and
Complications
Respiratory failure, ARDS
Severe pneumonia
Lung abscess or cavitary lesions
Sepsis
Laboratory Findings
Lobar, segmental, or subsegmental opacities on CXR, pleural effusion, pleural
adhesions, Hilar adenopathy
Leukocytosis; differential may be normal
Liver enzymes and/or CK may be abnormal
Sputum gram stain usually nonspecific
ARDS, acute respiratory distress syndrome; CAP, community-acquired pneumonia; CK, creatine kinase; CXR,
chest x-ray.
Glandular and Ulceroglandular Tularemia
Glandular and ulceroglandular tularemia account for the majority of naturally occurring cases of
tularemia. In the ulceroglandular form, an ulcer is formed at the site of inoculation, with
subsequent lymphadenopathy in the proximal draining lymph nodes. Occasionally,
lymphadenopathy occurs without an ulcer, leading to the designation of glandular disease.
S.F. Dept Public Health – Infectious Disease Emergencies TULAREMIA, July 2008 Page 5/13
GLANDULAR AND ULCEROGLANDULAR TULAREMIA
Incubation Period
3-5 days (range 1-14 days)
Transmission
Bite of an infective arthropod
Direct contact with infectious material (i.e., contaminated carcass, settled
infectious aerosol)
Signs and Symptoms
Ulceroglandular form – local skin involvement at site of exposure that
develops into a painful cutaneous papule with subsequent ulceration within
several days. Papule becomes necrotic and scars.
Glandular form – no cutaneous lesion occurs
Enlarged and tender regional lymphadenopathy that can persist for months
Fever, chills, malaise, myalgias, arthralgias, headache, anorexia, GI
symptoms are common
Progression and
Complications
Lymph node suppuration
Secondary pneumonia
Hematogenous spread to other organs
Sepsis
Laboratory Findings
Leukocytosis; differential may be normal
Liver enzymes and/or CK may be abnormal
CK, creatine kinase; GI, gastrointestinal.
Oculoglandular Tularemia
Oculoglandular tularemia results either from ocular inoculation from the hands after contact with
contaminated material or from splashes or aerosols generated during handling of infective material
(e.g., animal carcasses). This form of tularemia could occur in a bioterrorism setting as a result of
an aerosol exposure. Organisms spread from the conjunctiva to regional nodes, where they cause
focal necrosis and lesions.
2-4
After an incubation period of 3-5 (range 1-14) days, oculoglandular tularemia presents as a painful
“red eye” with purulent exudation, chemosis, vasculitis, and painful regional lymphadenopathy.
Additional signs and symptoms may include photophobia, lacrimation, itching, local edema, and
changes in visual acuity. There is a potential for lymph node suppuration, hematogenous
dissemination, and development of sepsis.
2-4
Laboratory values are generally nonspecific, and Gram stain of conjunctival scrapings may or may
not demonstrate organisms.
2
Oropharyngeal Tularemia
Oropharyngeal or gastrointestinal tularemia occurs via ingestion of contaminated food including
undercooked meat, contaminated water or droplets, and oral inoculation from the hands after
contact with contaminated material.
3, 4
S.F. Dept Public Health – Infectious Disease Emergencies TULAREMIA, July 2008 Page 6/13
After an incubation period of 3-5 (range 1-14) days, oropharyngeal tularemia presents either as
acute pharyngitis with cervical lymphadenopathy or as ulcerative gastrointestinal lesions with
fever, abdominal pain, diarrhea, nausea, vomiting, mesenteric lymphadenopathy, and
gastrointestinal bleeding. Severity can range from mild diarrhea to overwhelming ulceration with
frank gastrointestinal bleeding and sepsis. A large inoculum (approximately 10
8
organisms) is
required to transmit disease orally. There is a potential for lymph node suppuration,
hematogenous dissemination, and development of sepsis.
2-4
Routine tests are generally
nonspecific. Leukocytosis may or may not be present.
2
Typhoidal Tularemia
Typhoidal (septicemic) tularemia is an acute, nonspecific febrile illness associated with F. tularensis
without prominent lymphadenopathy.
TYPHOIDAL TULAREMIA
Incubation Period
3-5 days (range 1-14 days)
Transmission Site of primary infection usually unknown
Signs and Symptoms
Fever, chills, headache, malaise, weakness, myalgias, arthralgias, cough
Prostration, dehydration, hypotension, pharyngitis
Watery diarrhea, anorexia, nausea, vomiting, abdominal pain (children may
have more severe GI involvement)
Generalized maculopapular rash with progression to pustules or erythema-
nodosum type rash may occur
Splenomegaly and hepatomegaly (not common)
Progression and
Complications
Secondary pneumonia
Hematogenous spread to other organs – osteomyelitis, pericarditis, peritonitis,
endocarditis, meningitis
Sepsis
Rhabdomyolysis
Cholestasis with jaundice
Renal failure
Debilitating illness lasting several months
Laboratory Findings
Pleural effusions
Leukocytosis; differential may be normal
Liver enzymes and/or CK may be abnormal
Sterile pyuria may occur
CK, creatine kinase; GI, gastrointestinal.
DIFFERENTIAL DIAGNOSIS
A high index of suspicion is required to diagnose tularemia because there are no readily available
rapid and specific confirmatory tests. In addition, the various forms of tularemia can have a
nonspecific appearance and/or resemble a wide range of much more common illnesses.
S.F. Dept Public Health – Infectious Disease Emergencies TULAREMIA, July 2008 Page 7/13
Differential: Pneumonic Tularemia
The following are clinical syndromes that can appear similar to the pneumonic form of tularemia:
bacterial pneumonia (Mycoplasma,
Staphylococcus, Streptococcus,
Haemophilus, Klebsiella, Moraxella
Legionella)
Chlamydia infection
Q fever
tuberculosis
inhalational anthrax
pneumonic plague
fungal pulmonary disease
(histoplasmosis, coccidiodomycosis)
Viral pneumonia (influenza, hantavirus,
RSV, CMV)
severe acute respiratory syndrome
(SARS)
other causes of atypical or chronic
pneumonias
Differential: Glandular and Ulceroglandular Tularemia
The following are clinical syndromes that can appear similar to the glandular and ulceroglandular
forms of tularemia:
pyogenic bacterial infections
cat-scratch disease (Bartonella)
syphilis
chancroid
lymphogranuloma venereum
tuberculosis
nontuberculosis mycobacterial infection
toxoplasmosis
sporotrichosis
rat-bite fever
anthrax
plague
herpes simplex virus infection
adenitis or cellulitis (Staphylococcus
or Streptococcus)
Pasteurella infections
rickettsial infections
orf virus infection
Differential: Oculoglandular Tularemia
The following are clinical entities that can appear similar to the oculoglandular form of tularemia:
pyogenic bacterial infections
adenoviral infection
syphilis
cat-scratch disease
herpes simplex virus infection
varicella-Zoster virus infection
sporotrichosis
coccidioidomycosis
tuberculosis
Differential: Oropharyngeal Tularemia
The following are causes of syndromes that appear similar to the oropharyngeal form of tularemia:
Streptococcus pharyngitis
infectious mononucleosis
adenoviral infection
diphtheria
GI anthrax
Differential: Typhoidal Tularemia
The following are causes of syndromes that can appear similar to typhoidal forms of tularemia:
Salmonella spp. infection
brucellosis
endocarditis
leptospirosis
S.F. Dept Public Health – Infectious Disease Emergencies TULAREMIA, July 2008 Page 8/13
If you are testing or considering testing for
tularemia, you should:
IMMEDIATELY notify
SFDPH Communicable Disease Control
(24/7 Tel: 415-554-2830).
SFDPH can authorize and facilitate testing, and
will initiate the public health response as needed.
Inform your lab that tularemia is under
suspicion. F. tularensis may pose a risk to
lab personnel.
These recommendations are current as of this document date. SFDPH will provide periodic updates as needed
and situational guidance in response to events (www.sfcdcp.org).
Legionella infection
Chlamydia infection
Q fever
disseminated mycobacterial or fungal
infection
rickettsial infections
malaria
meningococcemia
septicemic plague
septicemia caused by other gram-
negative bacteria
Staphylococcus or Streptococcus toxic
shock syndrome
other causes of prolonged fever without
localizing signs
LABORATORY DIAGNOSIS
The diagnosis of tularemia requires a high
index of suspicion because the disease often
presents with nonspecific symptoms and
nonspecific results of routine lab tests.
Although recommended, microscopy and
culture are difficult and often not fruitful. The
organism is rarely seen on stained clinical
specimens and is difficult to isolate using
routine culture media and conditions.
However, isolation is possible from a variety of
clinical specimens if culture conditions are
optimized. Even still, some strains may require up to a week to develop visible colonies, especially
if the patient has been placed on bacteriostatic antibiotic therapy. Because of the need for
nonroutine laboratory methods and because F. tularensis is a risk to laboratory personnel,
clinicians should notify the laboratory when tularemia is suspected.
3, 21
Diagnosis is most commonly confirmed by serologic testing. Antibody detection assays include tube
agglutination, microagglutination, hemoagglutination, and enzyme-linked immunosorbent assay
(ELISA). Significant antibodies appear around the end of the 2nd week of illness, peak at 4-5
weeks, and can persist indefinitely. A single titer of 1:160 or greater (by tube agglutination) or
1:128 or greater (by microagglutination) is a presumptive positive; a four-fold rise in titer is
required for definitive serologic diagnosis.
3, 21
Although rapid diagnostic tests are not widely available, the public health laboratory system may
be able to provide timely testing of certain clinical specimens (e.g., polymerase chain reaction
(PCR) testing).
TREATMENT AND PROPHYLAXIS
S.F. Dept Public Health – Infectious Disease Emergencies TULAREMIA, July 2008 Page 9/13
Treatment
First-line treatment for tularemia is streptomycin or gentamicin.
2, 20, 22, 23
Flouroquinolone
antibiotics have also been effective
24, 25
. Other alternatives are tetracyclines and chloramphenicol.
However these drugs are bacteriostatic and their uses have produced more relapses than
treatment with aminoglycosides.
20, 26
Clinicians should be aware that F. tularensis strains released
intentionally may be resistant to antimicrobials.
3
Supportive care, including fluid management and hemodynamic monitoring, should be considered
in all patients. Intensive care with respiratory support may be necessary in patients with
complications.
2
Contained casualty setting: The Working Group recommends parenteral antimicrobial therapy
when individual medical management is available (Table 1). Therapy may be switched to oral
antimicrobials when clinically indicated.
3
Mass casualty setting: Use of oral antibiotics may be necessary if the number of patients
exceeds the medical care capacity for individual medical management (Table 2).
3
TABLE 1. TREATMENT OF TULAREMIA IN THE CONTAINED CASUALTY SETTING
3
Patient Category Therapy Recommendation*
Adults:
Preferred Choices
Streptomycin, 1 gm IM BID for 10 daysठOR
Gentamicin, 5 mg/kg IM or IV QD for 10 days‡
Adults:
Alternative Choices
Doxycycline, 100 mg IV BID for 14-21 days OR
Chloramphenicol, 15 mg/kg IV QID for 14-21 days** OR
Ciprofloxacin, 400 mg IV BID for 10 days
Children:
Preferred Choices
Streptomycin, 15 mg/kg IM BID (max 2 gm/day) for 10 days‡ OR
Gentamicin, 2.5 mg/kg IM or IV TID for 10 days‡
Children:
Alternative Choices
Doxycycline,
>45 kg, give adult dosage for 14-21 days
<45 kg, give 2.2 mg/kg IV BID for 14-21 days OR
Chloramphenicol, 15 mg/kg IV QID for 14-21 days** OR
Ciprofloxacin, 15 mg/kg IV BID (max 1 gm/day) for 10 days
* These treatment recommendations reflect those of the Working Group on Civilian Biodefense and may not
necessarily be approved by the Food and Drug Administration.
Acceptable for pregnant women.
§ Streptomycin is not as acceptable as gentamicin for use in pregnant women because irreversible deafness in children
exposed in utero has been reported with streptomycin use.
‡ Aminoglycosides must be adjusted according to renal function.
** Concentration should be maintained between 5 and 20 цg/mL; concentrations >25 цg/mL can cause reversible
bone marrow suppression.
S.F. Dept Public Health – Infectious Disease Emergencies TULAREMIA, July 2008 Page 10/13
TABLE 2. TREATMENT OF TULAREMIA IN THE MASS CASUALTY SETTING AND FOR
POSTEXPOSURE PROPHYLAXIS*
3
Patient Category Therapy Recommendation*
Adults (Including
Pregnant Women)
Doxycycline, 100 mg PO BID for 14 days OR
Ciprofloxacin, 500 mg PO BID for 14 day
Children
Doxycycline,
>45 kg, give adult dosage for 14 days
<45 kg, give 2.2 mg/kg PO BID for 14 days OR
Ciprofloxacin, 15 mg/kg PO BID (max 1 gm/day) for 10 days
* These treatment recommendations reflect those of the Working Group on Civilian Biodefense and may not
necessarily be approved by the Food and Drug Administration.
‡ Although fetal toxicity may occur with doxycycline use, the Working Group recommended doxycycline or
ciprofloxacin for postexposure prophylaxis of pregnant women or for treatment of infection of pregnant women in
the mass casualty setting.
Postexposure Prophylaxis
Antibiotic prophylaxis should begin as soon as possible and preferably within 24 hours after
exposure to an infectious aerosol containing F. tularensis (Table 2). Postexposure prophylactic
antibiotic treatment of close contacts
of tularemia patients is not recommended because human to
human
transmission of F. tularensis is not known to occur.
3
Vaccination
A live, attenuated vaccine was developed and has been used in the United States to protect
laboratory personnel who work with F. tularensis. This vaccine is currently under review by the
Food and Drug Administration and is unavailable.
3
Clinical trials to develop a new tularemia
vaccine are underway but it is not likely that a vaccine will be widely available in the near future.
COMPLICATIONS AND ADMISSION CRITERIA
Disease manifestations and complications are typically related to the portal of entry of F.
tularensis. Pneumonic tularemia may result in severe pneumonia, lung abscess, or acute
respiratory distress syndrome (ARDS). Glandular and ulceroglandular tularemia may progress to
lymph node suppuration and secondary pneumonia. Oculoglandular tularemia can cause localized
lymph node suppuration, whereas oropharyngeal tularemia has been associated with mesenteric
lymphadenitis, gastrointestinal (GI) ulceration, and GI bleeding. Typhoidal tularemia not
uncommonly progresses to secondary pneumonia. All forms of human tularemia carry the
potential for hematogenous dissemination of the organism to other organs such as bone,
pericardium, and peritoneum, and for progression to sepsis and multiorgan failure.
2-4
Admission to the hospital is generally advisable for patients with any form of tularemia, in order to
administer antibiotics intravenously and to monitor for disease progression.
S.F. Dept Public Health – Infectious Disease Emergencies TULAREMIA, July 2008 Page 11/13
These recommendations are current as of this document date. SFDPH will provide periodic updates as needed
and situational guidance in response to events (www.sfcdcp.org).
INFECTION CONTROL
Clinicians should notify local public health authorities, their institution’s infection control
professional, and their laboratory of any suspected tularemia cases. Public health authorities may
conduct epidemiologic investigations and implement disease control interventions to protect the
public. Both the Hospital Infection Control Practices Advisory Committee (HICPAC) of the CDC and
the Working Group for Civilian Biodefense recommend Standard Precautions for patients with
tularemia in a hospital setting without the need for isolation. Routine laboratory procedures should
be carried out under Biosafety Level 2 (BSL-2) conditions; however, manipulation of cultures or
other activities that may produce aerosol or droplets (e.g., centrifuging, grinding, vigorous
shaking) require BSL-3 conditions.
3
Decontamination
Contaminated surfaces can be disinfected with commercially available bleach or a 1:10 dilution of
household bleach and water. All persons exposed to an aerosol containing F. tularensis should be
instructed to wash body surfaces and clothing with soap and water.
3
.
PEARLS AND PITFALLS
1. The onset of tularemia is usually abrupt, with fever, headache, chills and rigors,
generalized body aches, and coryza. A pulse-temperature dissociation has been noted in as
many as 42% of patients.
3
2. Clinicians should familiarize themselves with the local epidemiology of tularemia. The
occurrence of human cases may follow a local tularemia epizootic (outbreak of disease in
an animal population). Occurrence of pneumonic tularemia in a low-incidence area should
prompt consideration of bioterrorism.
3
3. The diagnosis of tularemia relies heavily on clinical suspicion. Routine laboratory tests are
usually nonspecific. The organism is usually not apparent on gram-stained smears or
tissue biopsies and usually does not grow on standard culture plates. However, F.
tularensis may be recovered from blood and body fluids using special supportive media.
Because of this and its potential hazards to laboratory personnel, the laboratory should be
notified if tularemia is suspected.
S.F. Dept Public Health – Infectious Disease Emergencies TULAREMIA, July 2008 Page 12/13
REFERENCES
1. FRANZ DR, JAHRLING PB, FRIEDLANDER AM, ET AL. CLINICAL RECOGNITION AND
MANAGEMENT OF PATIENTS EXPOSED TO BIOLOGICAL WARFARE AGENTS. JAMA. AUG 6
1997;278(5):399-411.
2. CIDRAP. TULAREMIA: CURRENT, COMPREHENSIVE INFORMATION ON PATHOGENESIS,
MICROBIOLOGY, EPIDEMIOLOGY, DIAGNOSIS, TREATMENT, AND PROPHYLAXIS. CENTER FOR
INFECTIOUS DISEASE RESEARCH AND POLICY, UNIVERSITY OF MINNESOTA. AVAILABLE AT:
HTTP://WWW.CIDRAP.UMN.EDU/CIDRAP/CONTENT/BT/TULAREMIA/BIOFACTS/TULAREMIAFAC
TSHEET.HTML.
3. DENNIS DT, INGLESBY TV, HENDERSON DA, ET AL. TULAREMIA AS A BIOLOGICAL WEAPON:
MEDICAL AND PUBLIC HEALTH MANAGEMENT. JAMA. JUN 6 2001;285(21):2763-2773.
4. PENN RL. CHAPTER 224 - FRANCISELLA TULARENSIS (TULAREMIA). IN: MANDELL GL, BENNETT
JE, DOLIN R, EDS. PRINCIPLES AND PRACTICE OF INFECTIOUS DISEASES, ED 6. NEW YORK, NY:
CHURCHILL LIVINGSTONE; 2005:2674-2685.
5. FARLOW J, WAGNER DM, DUKERICH M, ET AL. FRANCISELLA TULARENSIS IN THE UNITED
STATES. EMERG INFECT DIS. DEC 2005;11(12):1835-1841.
6. STAPLES JE, KUBOTA KA, CHALCRAFT LG, MEAD PS, PETERSEN JM. EPIDEMIOLOGIC AND
MOLECULAR ANALYSIS OF HUMAN TULAREMIA, UNITED STATES, 1964-2004. EMERG INFECT
DIS. JUL 2006;12(7):1113-1118.
7. ABD H, JOHANSSON T, GOLOVLIOV I, SANDSTROM G, FORSMAN M. SURVIVAL AND GROWTH OF
FRANCISELLA TULARENSIS IN ACANTHAMOEBA CASTELLANII. APPL ENVIRON MICROBIOL. JAN
2003;69(1):600-606.
8. CELEBI G, BARUONU F, AYOGLU F, ET AL. TULAREMIA, A REEMERGING DISEASE IN
NORTHWEST TURKEY: EPIDEMIOLOGICAL INVESTIGATION AND EVALUATION OF
TREATMENT RESPONSES. JPN J INFECT DIS. AUG 2006;59(4):229-234.
9. KANTARDJIEV T, IVANOV I, VELINOV T, ET AL. TULAREMIA OUTBREAK, BULGARIA, 1997-2005.
EMERG INFECT DIS. APR 2006;12(4):678-680.
10. REINTJES R, DEDUSHAJ I, GJINI A, ET AL. TULAREMIA OUTBREAK INVESTIGATION IN KOSOVO:
CASE CONTROL AND ENVIRONMENTAL STUDIES. EMERG INFECT DIS. JAN 2002;8(1):69-73.
11. CDC. TULAREMIA - UNITED STATES, 1990-2000. MMWR MORB MORTAL WKLY REP. MARCH 8
2002;51(9):181-184.
12. FELDMAN KA, STILES-ENOS D, JULIAN K, ET AL. TULAREMIA ON MARTHA'S VINEYARD:
SEROPREVALENCE AND OCCUPATIONAL RISK. EMERG INFECT DIS. MAR 2003;9(3):350-354.
13. BERRADA ZL, GOETHERT HK, TELFORD SR, 3RD. RACCOONS AND SKUNKS AS SENTINELS FOR
ENZOOTIC TULAREMIA. EMERG INFECT DIS. JUN 2006;12(6):1019-1021.
14. AVASHIA SB, PETERSEN JM, LINDLEY CM, ET AL. FIRST REPORTED PRAIRIE DOG-TO-HUMAN
TULAREMIA TRANSMISSION, TEXAS, 2002. EMERG INFECT DIS. MAR 2004;10(3):483-486.
15. BARNS SM, GROW CC, OKINAKA RT, KEIM P, KUSKE CR. DETECTION OF DIVERSE NEW
FRANCISELLA-LIKE BACTERIA IN ENVIRONMENTAL SAMPLES. APPL ENVIRON MICROBIOL. SEP
2005;71(9):5494-5500.
16. CDPH. CALIFORNIA MONTHLY SUMMARY REPORT SELECTED REPORTABLE DISEASES.
DIVISION OF COMMUNICABLE DISEASE CONTROL, CALIFORNIA DEPARTMENT OF PUBLIC
HEALTH. AVAILABLE AT:
HTTP://WWW.CDPH.CA.GOV/DATA/STATISTICS/PAGES/CD_TABLES.ASPX
.
17. SFDPH. SAN FRANCISCO COMMUNICABLE DISEASE REPORT, 1986-2003. SAN FRANCISCO
DEPARTMENT OF PUBLIC HEALTH. MAY 2005. AVAILABLE AT:
HTTP://WWW.SFCDCP.ORG/PUBLICATIONS
.
18. SFDPH. ANNUAL REPORT OF COMMUNICABLE DISEASES IN SAN FRANCISCO, 2004-2005. SAN
FRANCISCO DEPARTMENT OF PUBLIC HEALTH. AUGUST, 2006. AVAILABLE AT:
HTTP://WWW.SFCDCP.ORG/PUBLICATIONS
.
19. SFDPH. ANNUAL REPORT OF COMMUNICABLE DISEASES IN SAN FRANCISCO, 2006. SAN
FRANCISCO DEPARTMENT OF PUBLIC HEALTH. JANUARY 2008. AVAILABLE AT:
HTTP://WWW.SFCDCP.ORG/PUBLICATIONS
.
20. EVANS ME, GREGORY DW, SCHAFFNER W, MCGEE ZA. TULAREMIA: A 30-YEAR EXPERIENCE
WITH 88 CASES. MEDICINE. JUL 1985;64(4):251-269.
21. MITCHELL CL, PENN RL. CHAPTER 322 - FRANCISELLA TULARENSIS (TULAREMIA) AS AN
AGENT OF BIOTERRORISM. IN: MANDELL GL, BENNETT JE, DOLIN R, EDS. PRINCIPLES AND
PRACTICE OF INFECTIOUS DISEASES, ED 6. NEW YORK, NY: CHURCHILL LIVINGSTONE; 2005.
S.F. Dept Public Health – Infectious Disease Emergencies TULAREMIA, July 2008 Page 13/13
22. AAP. TULAREMIA. IN: PICKERING L, ED. RED BOOK: REPORT ON THE COMMITTEE ON
INFECTIOUS DISEASES. 25 ED. ELK GROVE VILLAGE, ILL: AMERICAN ACADEMY OF
PEDIATRICS; 2000:618-620.
23. HASSOUN A, SPERA R, DUNKEL J. TULAREMIA AND ONCE-DAILY GENTAMICIN. ANTIMICROB
AGENTS CHEMOTHER. FEB 2006;50(2):824.
24. LIMAYE AP, HOOPER CJ. TREATMENT OF TULAREMIA WITH FLUOROQUINOLONES: TWO
CASES AND REVIEW. CLIN INFECT DIS. OCTOBER 1999;29:922-924.
25. PEREZ-CASTRILLON JL, BACHILLER-LUQUE P, MARTIN-LUQUERO M, MENA-MARTIN FJ,
HERREROS V. TULAREMIA EPIDEMIC IN NORTHWESTERN SPAIN: CLINICAL DESCRIPTION AND
THERAPEUTIC RESPONSE. CLIN INFECT DIS. AUGUST 15 2001;33:573-576.
26. OVERHOLT EL, TIGERTT WD, KADULL PJ, ET AL. AN ANALYSIS OF FORTY-TWO CASES OF
LABORATORY-ACQUIRED TULAREMIA. TREATMENT WITH BROAD SPECTRUM ANTIBIOTICS.
AM J MED. MAY 1961;30:785-806.
S.F. Dept Public Health – Infectious Disease Emergencies VIRAL HEMORRHAGIC FEVER, July 2008 Page 1/18
Outline
Introduction
Epidemiology
Clinical Features
Differential Diagnosis
Laboratory Diagnosis
Treatment and Prophylaxis
Complications
Infection Control
Pearls and Pitfalls
References
Immediately report any suspected or confirmed
cases of viral hemorrhagic fevers to:
SFDPH Communicable Disease Control
(24/7 Tel: 415-554-2830)
- By law, health care providers must report suspected or
confirmed cases of viral hemorrhagic fever to their local
health department immediately [within 1 hr].
- SFDPH Communicable Disease Control can facilitate
specialized testing and will initiate the public health response
as needed.
Also notify your:
Infection Control Professional
Clinical Laboratory
INTRODUCTION
Viral hemorrhagic fevers (VHFs) refer to a group of illnesses caused by several families of viruses,
including:
Filoviridae (Ebola and Marburg viruses)
Arenaviridae (Lassa fever and New World hemorrhagic fever)
Bunyaviridae (Rift Valley fever, Crimean-Congo fever, and “agents of hemorrhagic fever
with renal syndrome”)
Flaviviridae (yellow fever, Omsk hemorrhagic fever, Kyasanur Forest disease, and dengue)
Many VHF viruses are virulent, and some are highly infectious (e.g., filoviruses and arenaviruses)
with person-to-person transmission from direct contact with infected blood and bodily secretions.
Effective therapies and prophylaxis are extremely limited for VHF; therefore, early detection and
strict adherence to infection control measures are essential.
The Working Group for Civilian Biodefense considers some hemorrhagic fever viruses to pose a
more serious threat as potential biological weapons based on risk of morbidity and mortality,
feasibility of production, and ability to cause infection through aerosol dissemination. These include
Ebola, Marburg, Lassa fever, New World arenaviruses, Rift Valley fever, yellow fever, Omsk
hemorrhagic fever, and Kyasanur Forest disease.
1
Therefore, this chapter will focus only on these
VHF viruses and will not include a discussion of dengue fevers, hemorrhagic fever with renal
syndrome (e.g., hantavirus), and Crimean-Congo hemorrhagic fevers.
VIRAL HEMORRHAGIC FEVER JULY 2008
S.F. Dept Public Health – Infectious Disease Emergencies VIRAL HEMORRHAGIC FEVER, July 2008 Page 2/18
EPIDEMIOLOGY
VHF viruses as Biological Weapons
Of the potential ways in which VHFs could be used as a biological weapon, an aerosol release is
expected to have the most severe medical and public health outcomes.
An intentional release of a VHF virus would have the following characteristics:
2
Multiple similarly presenting cases clustering in time:
o acute nonspecific febrile illness with onset 2 to 21 days after the initial release
(may include fever, myalgias, rash, and encephalitis)
o severe illness with a fever and hemorrhagic manifestations
Atypical host characteristics: unexpected, unexplained cases of acute illness in previously
healthy persons, or people with hemorrhagic symptoms who have no conditions
predisposing for hemorrhagic illness
Unusual geographic clustering: cases occurring in an area where naturally occurring VHF is
not endemic
Absence of risk factors: patients lack VHF exposure risk factors (e.g., travel to a VHF
endemic country such as South America, Africa, or Asia; handling animal carcasses;
contact with people sick with VHF).
In the event of an intentional release, some VHFs could infect susceptible animals and potentially
lead to establishment of the disease in the environment.
Naturally Occurring Viral Hemorrhagic Fever
All of the VHF agents cause sporadic disease or epidemics in areas of endemicity. The routes of
transmission are variable, but most are zoonotic with spread via arthropod bites or contact with
infected animals. Person-to-person spread is a major form of transmission for many of the viruses.
Epidemiologic characteristics for each virus are described in the tables below.
S.F. Dept Public Health – Infectious Disease Emergencies VIRAL HEMORRHAGIC FEVER, July 2008 Page 3/18
EPIDEMIOLOGIC CHARACTERISTICS OF VHF VIRUSES
Virus Worldwide Occurrence Reservoir/
Vector
Transmission
Ebola
Identified in 1976 during outbreaks in the
Democratic Republic of Congo (formerly
known as Zaire) and Sudan. Four species
of Ebola virus are recognized and named
after the region where they were
discovered: Ivory Coast, Sudan, Zaire, and
Reston
Reported cases of naturally occurring
infections have occurred in Africa:
Democratic Republic of Congo (1976,
1995), Sudan (1976, 1979, 2004), Gabon
(1994, 1996, 2001-02), Ivory Coast
(1994), Uganda (2000-01), Republic of
Congo (2001-02, 2003-04, 2005)
Laboratory-acquired infections have
occurred in England (1976)
Ebola has been introduced to quarantine
facilities in United States (1989, 1990,
1996), Italy (1992), Philippines (1996)
unknown
A
/
unknown
Filoviruses
Marburg
Identified in 1967 in Germany when
laboratory staff handling tissues from
African green monkeys became infected
Reported cases of naturally occurring
infections have occurred in: South Africa
D
(1975), Western Kenya (1987, 1980),
Democratic Republic of Congo (1998-
2000), Angola (2004-2005)
Laboratory-acquired infections have
occurred in Germany (1967)
unknown
E
/
unknown
Person-to-person transmission
B
occurs via:
Contact with blood, secretions,
or tissue of infected patient
C
(sexual transmission may
occur up to 3 months after
clinical illness ends)
Contact with cadaver
Airborne transmission
(suspected)
Parenteral inoculation
(unsterilized needles,
accidental needle sticks)
Contact with blood, secretions,
or tissue of infected nonhuman
primate
Exposure in laboratory
Lassa
Identified in 1969 in Nigeria
Lassa fever is endemic in West African
countries between Nigeria and Senegal.
There are an estimated 100,000-300,000
annual infections in West Africa.
Nosocomial outbreaks and endemic
transmission are more common during the
dry season (January – April). Outbreaks
have occurred in Sierra Leone, Guinea,
Liberia, and Nigeria.
Lassa fever is occasionally imported to
other countries through travel
multimmamate
mouse/ none
Arenaviruses
New
World HF
New World HFs (or South American HF)
include Junin, Machupo, Guanarito, and
Sabia
Reported cases of naturally occurring
infections have occurred in South America:
Argentina, Bolivia, Venezuela, Brazil
An additional New World HF, Whitewater
Arroyo, was isolated from 3 cases in
California
rodents (mouse,
wood rat)/ none
Inhalation of aerosols of
rodent excreta,
Ingestion of food contaminated
with rodent excreta
Contact of rodents OR rodent
excreta with open skin or
mucous membranes
Person-to-person transmission
via:
Contact with infectious blood
and bodily fluids
Parenteral inoculation
(unsterilized needles accidental
needlesticks)
Airborne transmission
(suspected)
Exposure in laboratory
S.F. Dept Public Health – Infectious Disease Emergencies VIRAL HEMORRHAGIC FEVER, July 2008 Page 4/18
Bunyavirus
Rift Valley
Fever
Reported cases of naturally occurring
infections have occurred in Sub-Saharan
Africa, Egypt (1977-8, 1993), Kenya &
Somalia (1997-8), Saudi Arabia (2000-01),
Yemen (2000-01), Tanzania (2006)
3
ruminants
(sheep, cattle,
goats, buffalo)/
mosquito
Bite of an infected mosquito
Direct contact with infected
animal tissue (ruminants)
Inhalation of aerosol from
infected animal carcasses
(ruminants)
Transmission by ingestion of
contaminated raw animal milk
(suspected)
Exposure in laboratory
Yellow
Fever
Yellow fever is endemic in Sub-Saharan
Africa and tropical regions of South
America (mostly in forested regions). From
2000-2004 there were 2570 cases reported
in Africa and 629 in South America.
4
Most outbreaks occur in:
- West Africa and Central Africa – in
Savanna zones during the rainy season
- Urban and Jungle regions of sub-Saharan
Africa
- South America – forested areas of Bolivia,
Brazil, Columbia, Ecuador, Peru,
Venezuela, French Guiana, Guyana
primate/
Aedes and
Haemagogus
mosquitoes
Bite of an infected mosquito
Exposure in laboratory
Kyasanur
Forest
disease
virus
First identified in 1957 from a sick monkey
from the Kyasanur forest in the Karnataka
State, India. Recently, a similar virus was
discovered in Saudi Arabia.
Kyasanur Forest disease is only found in
the Karnataka State in India, where 400-
500 cases are reported annually.
5
Vertebrates
F
/
Tick
G
Bite of an infected tick;
Inhalation of aerosols by
laboratory workers during
cultivation of these viruses
Flavivirus
Omsk HF
Omsk HF (OHF) was first identified in 1947
in Omsk, Russia. Epizootics began
occurring in western Siberia among newly
introduced muskrats (for fur trade) and
caused large outbreaks in humans from
1945-1958.
6
Cases of Omsk HF have been reported in
central Asia (western Siberian regions of
Omsk, Novosibirsk, Kurgan, Tyumen).
From 1988-1997, there were 165 cases of
Omsk reported from these regions.
7, 8
Naturally occurring infections peak in
spring/early summer and autumn.
6
Few
cases have occurred in recent years.
9
rodents (vole,
muskrat) –
possibly water-
borne/tick
Bite of an infected tick
Contact with blood, secretions,
or tissue of an infected animal
Inhalation of aerosols by
laboratory workers during
cultivation of these viruses
Ingestion of contaminated raw
goat milk
Waterborne (suspected)
Airborne (suspected)
A
Fruit bats are currently a candidate reservoir. Asymptomatic infections occur in bats within the geographical range of
human Ebola outbreaks.
10
B
The initial transmission of Marburg and Ebola viruses from animals to humans is not understood.
C
Risk of transmission is greatest during the latter stages of illness when viral loads are highest, while transmission rarely
(if ever) occurs before the onset of symptoms.
D
Case most likely exposed in Zimbabwe, traveling nurse also became infected.
E
Fruit bats are currently a candidate reservoir. Serological evidence of infections has been noted in fruit bats in the areas
of human Marburg cases.
11
F
Not well understood – vertebrate hosts include: rodents, bats, small mammals, monkeys
G
Not well understood
S.F. Dept Public Health – Infectious Disease Emergencies VIRAL HEMORRHAGIC FEVER, July 2008 Page 5/18
OCCURRENCE OF VHF VIRUSES IN THE UNITED STATES
Virus United States Occurrence
Ebola Ebola-Reston virus has been introduced into quarantine by monkeys imported from the
Philippines on three occasions. In two of the three incidents (1989, 1990), four humans were
infected with Ebola-Reston but did not become ill (developed antibodies).
Marburg NA
Lassa Fever Lassa fever is rarely encountered in the United States. In 2004, a case of imported Lassa fever
occurred in a New Jersey resident who became infected while traveling in West Africa. None of
the contacts of the patient developed any symptoms compatible with Lassa fever within the
incubation period. This was the first reported case of Lassa fever imported into the United
States since 1989.
12
New World HF Three cases of Whitewater Arroyo virus were reported in California in 1999-2000; all were
fatal. Whitewater Arroyo has been isolated from woodrats in North America, but these were
the first reported cases of human disease.
Rift Valley virus NA
Yellow Fever Virus spread from West Africa to United States through slave trade vessels, caused significant
outbreaks, including:
Philadelphia (1793) – 10% of population died
Mississippi (1878) – 100,000 cases
Yellow fever has been imported into the United States by non-immunized travelers to yellow-
fever endemic countries 3 times since 1924:
1996 (Brazil to Tennessee)
13
1999 (Venezuela to Marin County, CA)
4
2002 (Brazil to Texas)
14
All cases were fatal
Kyasanur Forest
disease virus
NA
Omsk HF NA
CLINICAL FEATURES
The clinical features of VHF vary according to the virus and are detailed by disease below.
However, in the case of bioterrorism, the virus may not initially be known; therefore, clinical
features of VHFs, in general, are also provided.
CLINICAL FEATURES OF VIRAL HEMORRHAGIC FEVER
A
Early Signs High fever, headache, malaise, fatigue, arthralgias/ myalgias, prostration,
nausea, abdominal pain, nonbloody diarrhea
Mild hypotension, relative bradycardia, tachypnea, conjunctival involvement ,
pharyngitis, rash or flushing
Progression (1-2 weeks) Hemorrhagic manifestations (e.g., petechiae, hemorrhagic or purpuric rash,
epistaxis, hematemesis, melena, hemoptysis, hematochezia, hematuria)
CNS dysfunction (e.g., delirium, convulsions, cerebellar signs, coma)
Hepatic involvement (e.g., jaundice, hepatitis)
Complications and Sequelae Shock, DIC, multi-system organ failure
Illness-induced abortion in pregnant women
Transverse myelitis
Uveitis
Pericarditis
Orchitis
S.F. Dept Public Health – Infectious Disease Emergencies VIRAL HEMORRHAGIC FEVER, July 2008 Page 6/18
Parotitis
Pancreatitis
Hearing or vision loss
Impaired motor coordination
Convalescence may be prolonged or complicated by weakness, fatigue,
anorexia, cachexia, alopecia, arthralgias
Laboratory Findings Leukopenia (except in Lassa)
Leukocytosis
Thrombocytopenia
Elevated liver enzymes
Anemia or hemoconcentration
Coagulation abnormalities (e.g., prolonged bleeding time, prothrombin time,
and activated partial thromboplastin time, elevated fibrin degradation
products, and increased fibrinogen)
Proteinuria, hematuria, oliguria, and azotemia
A
Adapted from
1
CNS, central nervous system; DIC, disseminated intravascular coagulation.
Ebola/Marburg:
Case-fatality of Ebola ranges from 50% to 90% and that for Marburg, from 23% to 70%.
Considerations for pregnant women: High mortality from Ebola infection in pregnant women
(95.5%), as well as high rates of fetal and neonatal loss (100%) have been reported.
15
CLINICAL FEATURES OF EBOLA/MARBURG
Incubation
Period
Prominent Clinical Features Laboratory Findings
2-21 days Acute onset of fever, myalgias/arthralgias, headache, prostration,
fatigue (< 1 week)
Nausea, vomiting, abdominal pain, diarrhea, chest pain, cough,
pharyngitis, hiccups
Maculopapar rash (day 5 after symptom onset)
Hemorrhagic manifestations
Photophobia, conjunctival inflammation, lymphadenopathy,
hepatitis, pancreatitis (common)
CNS dysfunction
Shock with DIC and organ failure (week 2 after symptom onset)
Complications and sequelae: arthralgias, ocular disease, parotitis,
orchitis, hearing loss, pericarditis, transverse myelitis
Leukopenia (early)
Leukocytosis (late)
Thrombocytopenia (early)
Elevated liver enzymes
Elevated amylase
Lab features of DIC
Lassa Fever
Most people infected with Lassa fever have a mild or subclinical presentation (80%). Severe
disease occurs in 12-20%, with overall case-fatality around 1% (10-25% mortality in hospitalized
patients). During an outbreak, a clinical combination of fever, pharyngitis, retrosternal pain, and
proteinuria was predictive of laboratory-confirmed disease in 70% of cases. Findings associated
S.F. Dept Public Health – Infectious Disease Emergencies VIRAL HEMORRHAGIC FEVER, July 2008 Page 7/18
with death include hypotension, peripheral vasoconstriction, oliguria, edema, pleural effusions, and
ascites. Lassa fever requires a high index of suspicion because clinical features are nonspecific and
vary from patient to patient. Recovery generally begins around day 10 but may be accompanied
by prolonged weakness and fatigue.
16
Considerations for children: Clinical features of Lassa fever infection in children may be even
more difficult to diagnose due because of heterogeneous presentation. One syndrome in children
less than 2 years old is marked by severe generalized edema, abdominal distension, and bleeding
manifestations (this is associated with high case fatality of 75%.
16
)
Considerations for pregnant women: Case-fatality in pregnant women is higher than in
nonpregnant women, and risk of death increases in the third trimester (30%). Evacuation of
uterus (i.e., delivery, spontaneous abortion, or evacuation of retained products of conception) can
significantly reduce risk of death in the pregnant woman. Lassa virus infection leads to a high rate
of fetal and neonatal death (>80%).
17
CLINICAL FEATURES OF LASSA FEVER
A
Incubation
Period
Prominent Clinical Features Laboratory Findings
3-16 Gradual onset of fever, weakness, pain, arthralgias
Chest and back pain, exudative pharyngitis, cough, abdominal pain,
vomiting (very common)
Diarrhea and proteinuria (common)
Facial and pulmonary edema, mucosal bleeding, pleural effusions,
neurological involvement (encephalopathy, coma, seizures), ascites,
shock (less common)
Illness-induced abortion among pregnant women
Complications & Sequelae: 8
th
cranial nerve damage with hearing loss,
pericarditis
Leukocyte & platelet
counts often normal
Elevated liver
enzymes may occur
A
Adapted from
1, 2, 6
New World Hemorrhagic Fevers
The New World hemorrhagic fevers (Junin, Machupo, Guanarito, Sabia) have similar clinical
features and progression. Mortality ranges from 15% to 30% and recovery generally takes 2-3
weeks. Sequelae are not common.
6
Considerations for pregnant women: Case-fatality from New World Hemorrhagic Fever
infection in pregnant women is higher than non-pregnant women. Infection also leads to a high
rate of fetal death.
6
S.F. Dept Public Health – Infectious Disease Emergencies VIRAL HEMORRHAGIC FEVER, July 2008 Page 8/18
CLINICAL FEATURES OF NEW WORLD HEMORRHAGIC FEVERS
A
Incubation
Period
Prominent Clinical Features Laboratory Findings
7-12 days
(range 5-19)
Gradual onset of fever, malaise, myalgias (especially lower back),
pharyngitis
Drowsiness, dizziness, tremor, epigastric pain and/or constipation,
photophobia, retro-orbital pain, conjunctivitis, lymphadenopathy,
postural hypotension
Hemorrhagic manifestations (e.g., petechial rash [oral and
dermal], facial flushing, facial edema, capillary leak syndrome,
membrane hemorrhage, narrowing pulse pressure, vasoconstriction)
CNS dysfunction (e.g., hyporeflexia, gait abnormalities,
palmomental reflex, tremors, other cerebellar signs)
Shock, coma, seizures
Leukopenia
Thrombocytopenia
Proteinuria
Rising hematocrit
A
Adapted from
1, 2, 6
Rift Valley Fever
Rift valley fever (RVF) has not been documented to spread from person to person; however, low
titers of virus have been isolated from throat washings. There has been one case where vertical
transmission was suspected.
18
Historically the case-fatality estimate of RVF is less than 1%;
however, a recent outbreak in Saudi Arabia (2000-01) had an overall case-fatality of 14-17% (33%
case fatality in patients admitted to RFV unit because of severe disease). Factors associated with
high mortality include hepatorenal failure, severe anemia, hemorrhagic or neurological
manifestations, jaundice and shock.
3, 19
CLINICAL FEATURES OF RIFT VALLEY FEVER
A
Incubation
Period
Prominent Clinical Features Laboratory Findings
2-6 days Fever, nausea, vomiting
Abdominal pain, diarrhea, jaundice
CNS dysfunction
Hemorrhagic disease (1-17%)
Ocular involvement (photophobia, retro-orbital pain, retinitis, vision loss,
scotoma)
Renal involvement or failure
Shock
Thrombocytopenia
Leukopenia
Severe anemia
Elevated liver enzymes
Elevated LDH and CK
A
Adapted from
1-3, 6, 19
CNS, central nervous system; CK, creatine kinase; LDH, lactate dehydrogenase.
S.F. Dept Public Health – Infectious Disease Emergencies VIRAL HEMORRHAGIC FEVER, July 2008 Page 9/18
Yellow Fever
Yellow fever may resolve after a very mild course or may progress to moderate or severe illness
(15%) after a short remission. Death occurs in 7-10 days after onset of illness.
2, 6
CLINICAL FEATURES OF YELLOW FEVER
A
Incubation
Period
Prominent Clinical Features Laboratory Findings
3-6 days Prodrome:
Acute onset of fever, headache, myalgias,
Facial flushing, conjunctival injection
Illness may resolve, enter remission (lasts hours or days), or progress to:
High fever, headache, severe myalgias (especially back), nausea,
vomiting, abdominal pain, weakness, prostration, bradycardia
Hemorrhagic manifestations
Fulminant infection with severe hepatic involvement
Shock, myocardial failure, renal failure, seizures, coma
Pneumonia, sepsis
Leukopenia (early),
Leukocytosis (late)
Thrombocytopenia
Elevated liver enzymes
and bilirubin
Albuminuria
Azotemia
Alkaline phosphatase
levels only slightly
elevated
A
Adapted from:
1, 2, 6
Kyasanur Forest disease
Kyasanur Forest disease is characterized by biphasic illness: 50% of patients who go on to develop
the second phase with meningoencephalitis. Case-fatality ranges from 3% to 10%.
CLINICAL FEATURES OF KYASANUR FOREST DISEASE
Incubation
Period
Prominent Clinical Features Laboratory Findings
2-9 days Phase I (6-11 days):
Acute onset of fever, myalgias, headache (6-11 days)
Conjunctival involvement, soft palate lesions, GI symptoms
Hyperemia of face and trunk (but no rash)
Lymphadenopathy
Hemorrhagic manifestations (not severe)
Phase II:
Afebrile period of 9-21 days followed by meningoencephalitis (50% of
patients)
Leukopenia
Lymphopenia or
lymphocytosis
Thrombocytopenia
Abnormal liver function
GI, gastrointestinal.
Omsk Hemorrhagic fever
Omsk hemorrhagic fever (OHF) is similar to Kyasanur Forest disease. Some also characterize OHF
as a biphasic illness with the first phase lasting 5-12 days with an estimated 30-50% of patients
S.F. Dept Public Health – Infectious Disease Emergencies VIRAL HEMORRHAGIC FEVER, July 2008 Page 10/18
going on to experience remission of fever, febrile illness, and more severe disease. Case fatality
ranges from 0.5% to 3%. Recovery may take weeks, but sequelae are not common.
1, 7
CLINICAL FEATURES OF OMSK HEMORRHAGIC FEVER
A
Incubation
Period
Prominent Clinical Features Laboratory Findings
3-8 days
(range 1-
10 days)
Acute onset of fever, headache, myalgias
Cough, conjunctivitis, soft pallet lesions, GI symptoms
Hyperemia of face and trunk (but no rash)
Lymphadenopathy, splenomegaly
Hemorrhagic manifestations (not severe)
Pneumonia, CNS dysfunction, meningeal signs, diffuse encephalitis
Leukopenia
Thrombocytopenia
A
Adapted from
2, 6, 7
CNS, central nervous system; GI, gastrointestinal.
DIFFERENTIAL DIAGNOSIS
A high index of suspicion is required to diagnose VHF because there are no readily available rapid
and specific confirmatory tests. In addition, the VHF viruses can have a nonspecific appearance or
resemble a wide range of much more common illnesses.
With a VHF virus used as a biological weapon, patients are less likely to have risk factors for
natural infection such as travel to VHF-endemic countries (Africa, Asia, or South America), contact
with sick animals or people, or arthropod bites within 21 days of symptom onset. The observation
of a severe illness with bleeding manifestations as its primary feature, which develops in several
related cases should be highly suspicious for VHF.
The Working Group for Civilian Biodefense suggests considering VHF in any patient with the
following clinical presentation:
Acute onset of fever (<3 weeks duration) in severely ill patient
Hemorrhagic manifestations (at least two of the following: hemorrhagic or purpuric rash,
epistaxis, hematemesis, hemoptysis, blood in stool, or other bleeding)
No conditions predisposing for hemorrhagic illness
No alternative diagnosis
Differential Diagnosis–Infectious Conditions (viral, rickettsial, bacterial and parasitic)
1, 2
Gram-negative bacterial septicemia
toxic shock syndrome (Staphylococcus,
Streptococcus)
meningococcemia
secondary syphilis
septicemic plague
salmonellosis (Salmonella typhi)
influenza
measles
rubella
dengue hemorrhagic fever
hemorrhagic varicella
hemorrhagic smallpox
Viral hepatitis
S.F. Dept Public Health – Infectious Disease Emergencies VIRAL HEMORRHAGIC FEVER, July 2008 Page 11/18
If you are testing or considering testing for
viral hemorrhagic fever, you should:
IMMEDIATELY notify
SFDPH Communicable Disease Control
(24/7 Tel: 415-554-2830).
SFDPH can authorize and facilitate testing, and
will initiate the public health response as needed.
Inform your lab that viral hemorrhagic fever
is under suspicion.
shigellosis
Chlamydia infection
borreliosis
leptospirosis
rickettsiosis
hantavirus pulmonary syndrome
malaria
African trypanosomiasis
Noninfectious Conditions
1, 2
thrombotic or idiopathic thrombocytopenic purpura
acute leukemia
hemolytic-uremic syndrome
collagen-vascular diseases
LABORATORY DIAGNOSIS AND RADIOGRAPHIC FINDINGS
Viral hemorrhagic fevers are a risk to
laboratory personnel. Clinicians should
immediately notify their laboratory, local health
department, and infection control professional
when VHF is suspected. In the event of an
outbreak, public health authorities will provide
recommendations for specimen collection
based on this situation (e.g., identification of
etiologic agent, laboratory capacity).
Diagnosis of VHF requires a high index of
suspicion because the disease initially presents
with nonspecific symptoms and non-specific results of routine lab tests. Routine laboratory
findings for specific HF viruses are listed in the clinical features tables.
A number of test methods can be used to diagnose VHF at specialized laboratories. These include
antigen-capture testing by enzyme-linked immunosorbent assay (ELISA), IgM antibody testing,
paired acute-convalescent serum serologies, reverse transcriptase polymerase chain reaction (RT-
PCR), immunohistochemistry methods, and electron microscopy. Viral identification in cell culture
is the gold standard of viral detection; however, this may only be attempted at a Biosafety Level 4
(BSL-4) facility. Combined ELISA Ag/ IgM has high specificity and sensitivity for early diagnosis of
Lassa fever and provides prognostic information (presence of indirect fluorescent antibody early in
disease associated with death).
20
S.F. Dept Public Health – Infectious Disease Emergencies VIRAL HEMORRHAGIC FEVER, July 2008 Page 12/18
These recommendations are current as of this document date. SFDPH will provide periodic updates as needed
and situational guidance in response to events (www.sfcdcp.org).
TREATMENT AND PROPHYLAXIS
Treatment
Medical management should follow the guidelines below:
MEDICAL MANAGEMENT RECOMMENDATIONS
Categorization Medical Management
Exposed Persons Medical Surveillance
No post-exposure prophylaxis is recommended
A
Suspected VHF Case of Unknown Viral Type
Supportive Care + Ribavirin Therapy
B
Suspected or Confirmed VHF Case
known to be caused by an Flavivirus or Filovirus
Supportive Care Only
Suspected Confirmed VHF Case
known to be caused by an Arenavirus or Bunyavirus
Supportive Care + Ribavirin Therapy
A
Previous CDC recommendations
state that Ribavirin should be given to high-risk contacts of persons with Lassa fever.
The Working Group on Civilian Biodefense recommends medical surveillance only, and notes that the CDC guidelines may
be under review.
B
Ribavirin therapy should be initiated promptly unless another diagnosis is confirmed or the etiologic agent is known to be
a Flavivirus or Filovirus.
Medical Surveillance: Persons should be instructed to record their temperature twice daily and
report any temperature of 38.0ºC or 100.4 ºF or higher (or any other signs or symptoms) to their
clinician and/or the proper public health authorities. Patients should be advised not to share
thermometers between family members and to properly disinfect thermometers after each use.
Supportive Care: Supportive care, including careful maintenance of fluid and electrolyte balance
and circulatory volume is essential for patients with all types of VHF. Mechanical ventilation,
dialysis, and appropriate therapy for secondary infections may be indicated. Treatment of other
suspected causes of disease, such as bacterial sepsis, should not be withheld while awaiting
confirmation or exclusion of the diagnosis of VHF. Anticoagulant therapies, aspirin, nonsteroidal
anti-inflammatory medications, and intramuscular injections are contraindicated.
Ribavirin Therapy: Ribavirin is recommended for: (1) suspect or probable cases of VHF of
unknown viral type or (2) suspect, probable, or confirmed cases caused by an Arenavirus or
Bunyavirus. Ribavirin has shown in vitro and in vivo activity against Arenaviruses (Lassa fever,
New World hemorrhagic fevers) and Bunyaviruses (Rift Valley fever and others). Ribavirin has
shown no activity against, and is not recommended for Filoviruses (Ebola and Marburg hemorrhagic
fever) or Flaviviruses (Yellow fever, Kyasanur Forest disease, Omsk hemorrhagic fever).
Recommendations for intravenous (IV) ribavirin therapy are shown below. Use of oral ribavirin may
be necessary if the number of patients exceeds the medical care capacity for individual medical
management.
S.F. Dept Public Health – Infectious Disease Emergencies VIRAL HEMORRHAGIC FEVER, July 2008 Page 13/18
Passive immunotherapy with convalescent human plasma has been used in the treatment and
prophylaxis of several VHFs with inconclusive results. Some suggest passive immunotherapy for
treatment of New World HFs based on effectiveness in Argentine HF (Junin).
1, 16, 21, 22
Post Exposure Prophylaxis
According to the Working Group on Civilian Biodefense, exposure is defined as proximity to an
initial release of VHF virus, or close or high-risk contact with a patient suspected of having VHF.
High risk contacts are defined as persons who “have had mucous membrane contact with a patient
(such as during kissing or sexual intercourse) or have had percutaneous injury involving contact
with a patient’s secretions, excretions, or blood.” Close contact is defined as, “those who live with,
shake hands with, hug, process laboratory specimens from, or care for a patient with VHF prior to
initiation of appropriate precautions.”
1
Medical surveillance (see above) is recommended for 21
days following the potential exposure or contact with the ill person.
Previous recommendations from the Centers for Disease Control and Prevention (CDC)
23
state that
prophylaxis with ribavirin should be given to persons exposed to Lassa virus. However, because
the efficacy of ribavirin prophylaxis for Lassa virus is unknown, the Working Group also
recommends that persons exposed be placed under medical surveillance until 21 days after the last
exposure.
1
The CDC recommendation is under review.
RIBAVIRIN THERAPY FOR PATIENTS WITH VHF OF UNKNOWN CAUSE OR CAUSED BY AN ARENAVIRUS
OR BUNYAVIRUS
A
IV Therapy
in Contained Casualty Situation
B
Therapy in a Mass-Casualty Setting
B
Adult
Ribavirin,
Loading dose 30 mg/kg (max 2 gm) IV,
Followed by:
16 mg/kg (max 1 gm) IV q6 hr
for 4 days,
Followed by:
8 mg/kg (max 500 mg) IV q8 hr
for 6 days
Ribavirin,
Loading dose of 2000 mg PO,
Followed by:
(Weight >75 kg): 1200 mg/day PO in 2
divided doses (600 mg in am and 600
mg in pm) for 10 days
C
(Weight <75 kg): 1000 mg/day PO in
divided doses (400 mg in am and 600
mg in pm) for 10 day
C
Children
D
Same as for adults
Loading dose of 30 mg/kg PO,
Followed by:
15 mg/kg/d PO in 2 divided doses for 10
days
Pregnant women
E
Same as for non-pregnant adults
Same as for non-pregnant adults
A
Ribavirin is not labeled for use in treatment of VHF by the US Food and Drug Administration (FDA) for treatment and must be used under an
Investigational New Drug (IND) protocol.
B
Use of oral vs. parenteral treatment will depend on resource availability
C
The current available formulation of ribavirin is 200-mg capsules, which cannot be broken open.
D
IV and oral ribavirin are not approved for children by the FDA; however, the benefits may outweigh the risk of ribavirin therapy.
E
Ribavirin is contraindicated in pregnant women; however, the benefits may outweigh the fetal risk of ribavirin therapy.
S.F. Dept Public Health – Infectious Disease Emergencies VIRAL HEMORRHAGIC FEVER, July 2008 Page 14/18
Vaccine
A licensed vaccine against yellow fever is effective if given prior to exposure. It is used for
travelers going to endemic areas. This vaccine does not prompt development of antibodies rapidly
enough to be used in the post-exposure setting. A rare, but serious adverse reaction to yellow
fever vaccine, viscerotropic and neurotropic disease, has recently been recognized and reported.
24
There is no licensed vaccine for any of the other VHFs, though research is underway on several
candidates.
Developmental VHF Therapeutics
Additional therapeutic candidates for vaccine, treatment, and prophylaxis of VHFs are currently
under development.
DEVELOPMENTAL VHF THERAPEUTICS
Virus Candidates for vaccine, treatment, and prophylaxis
Ebola/Marburg
A live attenuated recombinant vaccine for Ebola and Marburg HF has produced protective immune
responses in non-human primates
A vaccine used as a PEP produced some protective effect for Ebola in non-human primates when
administered soon after infection (20-30 min)
25
A Phase I clinical trial for an Ebola DNA vaccine was safe and produced an immune response in
humans
Treatment with small interfering RNAs (siRNAs) produced protective immune response in an animal
model (guinea pigs)
26
Lassa Fever
An attenuated recombinant vaccine produced protective immune responses in non-human
primates
27
New World HF
Live-attenuated vaccine available as investigational new drug in Argentine HF
28
Passive immunotherapy with convalescent human serum has been effective in Argentine HF
16
Rift Valley
virus
Vaccine available as investigational new drug
29
Yellow Fever
Licensed vaccine available (see above)
Kyasanur
Forest disease
virus
Formalin inactivated vaccine licensed and used in endemic areas
30
Omsk HF
NA
COMPLICATIONS AND ADMISSION CRITERIA
Patients with filovirus infection (Ebola and Marburg viruses) often experience hemorrhagic and
severe central nervous system (CNS) manifestations along with fever and jaundice during the first
week of illness. In the second week patients defervesce and either improve markedly or die as a
result of multiorgan dysfunction, shock, and disseminated intravascular coagulation. Survivors
may develop one or more complications including arthralgia, orchitis, hepatitis, transverse
myelitis, or uveitis.
Death from Lassa virus infection, when it occurs, is typically during the second week of illness and
is associated with hypotension, edema, and capillary leak syndrome. Up to one-third of Lassa
S.F. Dept Public Health – Infectious Disease Emergencies VIRAL HEMORRHAGIC FEVER, July 2008 Page 15/18
These recommendations are current as of this document date. SFDPH will provide periodic updates as needed
and situational guidance in response to events (www.sfcdcp.org).
fever survivors develop sensorineural deafness. The arenaviruses (Lassa and New World viruses)
share a propensity to cause fetal demise and high mortality rates in pregnant women.
Among the bunyavirus infections (Rift Valley fever and Crimean-Congo hemorrhagic fever), a
fulminant, fatal form of the disease with hemorrhage, hepatitis, and organ failure occurs in a
minority of patients. Rift Valley fever encephalitis is known to occur in a small percentage of
those affected.
Although many infections with yellow fever are clinically inapparent, patients may develop
multisystem illness dominated by an icteric hepatitis and a severe bleeding diathesis. In the latter
stages of illness encephalopathy, shock, and death may ensue. Patients who recover frequently
suffer from secondary bacterial infections.
The need for hospitalization and life support will be apparent in patients with bleeding diatheses,
CNS dysfunction, shock, or severe hepatorenal dysfunction. Patients exhibiting milder
manifestations of VHF or who appear to be in the early stages of disease could benefit from
hospitalization for supportive care and close observation. Treatment with intravenous ribavirin
should be initiated in patients known to have arenavirus or bunyavirus infection and in those with
VHF of unknown etiology pending viral identification.
INFECTION CONTROL
Clinicians should notify local public health authorities, their institution’s infection control
professional, and their laboratory of any suspected VHF cases. Public health authorities may
conduct epidemiologic investigations and implement disease control interventions to protect the
public.
Many VHF viruses are virulent, and some are highly infectious (e.g., filoviruses and arenaviruses)
with person to person transmission from direct contact with infected blood and bodily secretions.
Effective therapies and prophylaxis are extremely limited for VHF; therefore, early detection and
strict adherence with infection control measures are essential. Transmission rarely (if ever) occurs
before the onset of symptoms. Risk of transmission is greatest during the latter stages of illness
when viral loads are highest.
Among household contacts, secondary transmission for Ebola and Marburg ranges from 10 to 20%.
In the 1995 Ebola outbreak in the Democratic Republic of Congo, transmission did not occur among
household contacts with no direct physical contact with patients. Persons with physical contact
S.F. Dept Public Health – Infectious Disease Emergencies VIRAL HEMORRHAGIC FEVER, July 2008 Page 16/18
with patients were at increased risk of transmission, and those with body fluid contact had the
greatest risk.
31, 32
Guidance on infection control precautions have been published by both the CDC and the Working
Group for Civilian Biodefense in 2005 and 2002 respectively; these contained some
inconsistencies.
1, 33
More recent guidance was provided by the CDC’s Healthcare Infection Control
Practices Advisory Committee.
34
For patients infected or suspected to be infected with VHF healthcare workers and visitors should
use Standard, Contact and Droplet Precautions with eye protection. Single gloves are adequate for
routine patient care; double-gloving is advised during invasive procedures (e.g., surgery) that pose
an increased risk for blood exposure. Routine eye protection (i.e. goggles or face shield) is
particularly important. Fluid-resistant gowns should be worn for all patient contact. Airborne
Precautions are not required for routine patient care; however, use of airborne infection isolation
room (AIIR) is prudent when procedures that could generate infectious aerosols are performed
(e.g., endotracheal intubation, bronchoscopy, suctioning, autopsy procedures involving oscillating
saws). N95 or higher level respirators may provide added protection for individuals in a room
during aerosol-generating procedures. When a patient with a syndrome consistent with
hemorrhagic fever also has a history of travel to an endemic area, precautions are initiated upon
presentation and then modified as more information is obtained. Patients with hemorrhagic fever
syndrome in the setting of a suspected bioweapon attack should be managed using Airborne
Precautions, including AIIRs, since the epidemiology of a potentially weaponized hemorrhagic fever
virus is unpredictable.
34
All persons exposed to VHF should immediately wash the affected skin surfaces with soap and
water. Mucous membranes should be irrigated with copious amounts of water or eyewash solution.
Exposed persons should receive medical evaluation and monitoring.
PEARLS AND PITFALLS
1. Since effective postexposure prophylaxis is unavailable for VHF, strict adherence to infection
control measures is essential for limiting the spread of disease.
2. The risk for person-to-person transmission of VHF is highest during the latter phases of illness,
when viral loads are high and disease manifestations are most severe.
3. VHF viruses are not endemic in the United States, with the rare exception of Whitewater
Arroyo virus which caused three cases of human disease in California in 1999-2000 and may
have been related to wild rodents. Nearly all U.S. cases of VHF have been acquired by
overseas travelers or by scientific research personnel.
S.F. Dept Public Health – Infectious Disease Emergencies VIRAL HEMORRHAGIC FEVER, July 2008 Page 17/18
REFERENCES
1. Borio L, Inglesby T, Peters CJ, et al. Hemorrhagic fever viruses as biological weapons: medical
and public health management. Jama. May 8 2002;287(18):2391-2405.
2. CIDRAP. Viral Hemorrhagic Fever (VHF): Current, comprehensive information on
pathogenesis, microbiology, epidemiology, diagnosis, treatment, and prophylaxis. Center for
Infectious Disease Research and Policy, University of Minnesota. Available at:
http://www.cidrap.umn.edu/cidrap/content/bt/vhf/biofacts/vhffactsheet.html
.
3. Madani TA, Al-Mazrou YY, Al-Jeffri MH, et al. Rift Valley fever epidemic in Saudi Arabia:
epidemiological, clinical, and laboratory characteristics. Clin Infect Dis. Oct 15
2003;37(8):1084-1092.
4. WHO. Yellow Fever. World Health Organization. Available at:
http://www.who.int/csr/disease/yellowfev/en/index.html
.
5. CDC. Factsheet: Kyasanur Forest Disease. Available at:
http://www.cdc.gov/ncidod/dvrd/spb/mnpages/dispages/kyasanur.htm
.
6. Tsai TF, Vaughn DW, Solomon T. Chapter 149 - Flaviviruses (Yellow Fever, Dengue
Hemorrhagic Fever, Japanese Encephaliitis, West Nile Encephalitis, Tick-Borne Encephalitis. In:
Mandell GL, Bennett JE, Dolin R, eds. Principles and practice of infectious diseases, Ed 6. New
York, NY: Churchill Livingstone; 2005.
7. Charrel RN, Attoui H, Butenko AM, et al. Tick-borne virus diseases of human interest in
Europe. Clin Microbiol Infect. Dec 2004;10(12):1040-1055.
8. CDC. Factsheet: Omsk Hemorrhagic Fever. Available at:
http://www.cdc.gov/ncidod/dvrd/spb/mnpages/dispages/omsk.htm
.
9. Peters CJ. Chapter 326 - Bioterrorism: Viral Hemorrhagic Fevers. In: Mandell GL, Bennett JE,
Dolin R, eds. Principles and practice of infectious diseases, Ed 6. New York, NY: Churchill
Livingstone; 2005.
10. Leroy EM, Kumulungui B, Pourrut X, et al. Fruit bats as reservoirs of Ebola virus. Nature. Dec 1
2005;438(7068):575-576.
11. Towner JS, Pourrut X, Albarino CG, et al. Marburg Virus Infection Detected in a Common
African Bat. PLos ONE. August 22 2007;2(8):e764.
12. CDC. Imorted Lasa Fever -- New Jersey, 2004. MMWR. 2004;53(38):894-897.
13. McFarland JM, Baddour LM, Nelson JE, et al. Imported yellow fever in a United States citizen.
Clin Infect Dis. Nov 1997;25(5):1143-1147.
14. CDC. Fatal Yellow Fever in a Traveller Returning from Amazonas, Brazil, 2002. MMWR.
2002;51(15):324-325.
15. Mupapa K, Mukundu W, Bwaka MA, et al. Ebola hemorrhagic fever and pregnancy. J Infect Dis.
Feb 1999;179 Suppl 1:S11-12.
16. Peters CJ. Chapter 164 - Lymphocytic Choriomeningitis virus, Lassa Virus, and the South
American Hemorrhagic Fevers. In: Mandell GL, Bennett JE, Dolin R, eds. Principles and
practice of infectious diseases, Ed 6. New York, NY: Churchill Livingstone; 2005.
17. Price ME, Fisher-Hoch SP, Craven RB, McCormick JB. A prospective study of maternal and fetal
outcome in acute Lassa fever infection during pregnancy. Bmj. Sep 3 1988;297:584-586.
18. Arishi HM, Aqeel AY, Al Hazmi MM. Vertical transmission of fatal Rift Valley fever in a newborn.
Ann Trop Paediatr. Sep 2006;26(3):251-253.
19. Al-Hazmi M, Ayoola EA, Abdurahman M, et al. Epidemic Rift Valley fever in Saudi Arabia: a
clinical study of severe illness in humans. Clin Infect Dis. Feb 1 2003;36(3):245-252.
20. Bausch DG, Rollin PE, Demby AH, et al. Diagnosis and clinical virology of Lassa fever as
evaluated by enzyme-linked immunosorbent assay, indirect fluorescent-antibody test, and
virus isolation. J Clin Microbiol. Jul 2000;38(7):2670-2677.
21. Ruggiero HA, Perez Isquierdo F, Milani HA, et al. [Treatment of Argentine hemorrhagic fever
with convalescent's plasma. 4433 cases]. Presse Med. Dec 20 1986;15(45):2239-2242.
22. Mupapa K, Massamba M, Kibadi K, et al. Treatment of Ebola hemorrhagic fever with blood
transfusions from convalescent patients. International Scientific and Technical Committee. J
Infect Dis. Feb 1999;179 Suppl 1:S18-23.
23. CDC. Update: Management of patients with suspected viral hemorrhagic fever- US. MMWR.
1995;44:475-479.
S.F. Dept Public Health – Infectious Disease Emergencies VIRAL HEMORRHAGIC FEVER, July 2008 Page 18/18
24. CDC. Adverse Events Associated with 17D-Derived Yellow Fever Vaccination - United States,
2001-2002. MMWR. November 8 2002;51(44):989-993.
25. Feldmann H, Jones SM, Daddario-DiCaprio KM, et al. Effective post-exposure treatment of
Ebola infection. PLoS Pathog. Jan 2007;3(1):e2.
26. Geisbert TW, Hensley LE, Kagan E, et al. Postexposure protection of guinea pigs against a
lethal ebola virus challenge is conferred by RNA interference. J Infect Dis. Jun 15
2006;193(12):1650-1657.
27. Geisbert TW, Jones S, Fritz EA, et al. Development of a new vaccine for the prevention of
Lassa fever. PLoS Med. Jun 2005;2(6):e183.
28. Maiztegui JI, McKee KT, Jr., Barrera Oro JG, et al. Protective efficacy of a live attenuated
vaccine against Argentine hemorrhagic fever. AHF Study Group. J Infect Dis. Feb
1998;177(2):277-283.
29. Pittman PR, Liu CT, Cannon TL, et al. Immunogenicity of an inactivated Rift Valley fever
vaccine in humans: a 12-year experience. Vaccine. Aug 20 1999;18(1-2):181-189.
30. Pattnaik P. Kyasanur forest disease: an epidemiological view in India. Rev Med Virol. May-Jun
2006;16(3):151-165.
31. Borchert M, Mulangu S, Swanepoel R, et al. Serosurvey on household contacts of Marburg
hemorrhagic fever patients. Emerg Infect Dis. Mar 2006;12(3):433-439.
32. Dowell SF, Mukunu R, Ksiazek TG, Khan AS, Rollin PE, Peters CJ. Transmission of Ebola
hemorrhagic fever: a study of risk factors in family members, Kikwit, Democratic Republic of
the Congo, 1995. Commission de Lutte contre les Epidemies a Kikwit. J Infect Dis. Feb
1999;179 Suppl 1:S87-91.
33. CDC. Interim Guidance for Managing Patients with Suspected Viral Hemorrhagic Fever in U.S.
Hospitals. Available at: http://www.cdc.gov/ncidod/dhqp/bp_vhf_interimGuidance.html
.
34. Siegel JD, Rhinehart E, Jackson M, Chiarello L, HICPAC. Guideline for Isolation Precautions:
Preventing Transmission of Infectious Agents in Healthcare Settings, 2007. Centers for
Disease Control and Prevention. Available at:
http://www.cdc.gov/ncidod/dhqp/gl_isolation.html
.
S.F. Dept. Public Health – Infectious Disease Emergencies INFECTION CONTROL, July 2008 Page 1/6
Outline
Standard Precautions
Droplet Precautions
Contact Precautions
Airborne Precautions
References
STANDARD PRECAUTIONS
Use Standard Precautions, or the equivalent, for the care of all patients.
Standard Precautions apply to 1) blood; 2) all body fluids, secretions, and excretions except sweat,
regardless of whether or not they contain visible blood; 3) nonintact skin; and 4) mucous
membranes. Standard Precautions are designed to reduce the risk of transmission of
microorganisms from both recognized and unrecognized sources of infection in hospitals.
Hand Hygiene
When hands are visibly dirty or visibly soiled with blood or other body fluids:
Wash with either antimicrobial or non-antimicrobial soap & water
If hands are not visibly soiled:
Use an alcohol-based hand rub or wash with an antimicrobial soap & water to
decontaminate hands
Decontaminate hands before:
Having direct contact with patients
Donning sterile gloves before sterile procedures
Moving from a contaminated-body site to a clean-body site during patient care
Eating
Decontaminate hands after:
Contact with a patient's intact skin
Contact with body fluids or excretions, mucous membranes, nonintact skin, and wound
dressings, inanimate objects (including medical equipment) in the immediate vicinity of the
patient
Removing gloves
Using a restroom
Before eating and after using a restroom:
Wash with either antimicrobial or non-antimicrobial soap & water
INFECTION CONTROL PRECAUTIONS
S.F. Dept. Public Health – Infectious Disease Emergencies INFECTION CONTROL, July 2008 Page 2/6
If exposure to Bacillus anthracis is suspected or confirmed:
Wash with either antimicrobial or non-antimicrobial soap & water. The physical action of
washing and rinsing hands under such circumstances is recommended because alcohols,
chlorhexidine, iodophors, and other antiseptic agents have poor activity against spores.
Gloves
Wear gloves (clean, nonsterile gloves are adequate) when touching blood, body fluids, secretions,
excretions, and contaminated items. Put on clean gloves just before touching mucous membranes
and nonintact skin.
Change gloves between tasks and procedures on the same patient after contact with material that
may contain a high concentration of microorganisms. Remove gloves promptly after use, before
touching noncontaminated items and surfaces, and before going to another patient, and wash
hands immediately to avoid transfer of microorganisms to other patients or environments.
Mask, Eye Protection, Face Shield
Wear a mask and eye protection or a face shield to protect mucous membranes of the eyes, nose,
and mouth during procedures and patient-care activities that are likely to generate splashes or
sprays of blood, body fluids, secretions, and excretions.
Gown
Wear a gown (a clean, nonsterile gown is adequate) to protect skin and to prevent soiling of
clothing during procedures and patient-care activities that are likely to generate splashes or sprays
of blood, body fluids, secretions, or excretions. Select a gown that is appropriate for the activity
and amount of fluid likely to be encountered. Remove a soiled gown as promptly as possible and
wash hands to avoid transfer of microorganisms to other patients or environments.
Patient-Care Equipment
Handle used patient-care equipment soiled with blood, body fluids, secretions, and excretions in a
manner that prevents skin and mucous membrane exposures, contamination of clothing, and
transfer of microorganisms to other patients and environments. Ensure that reusable equipment is
not used for the care of another patient until it has been cleaned and reprocessed appropriately.
Ensure that single-use items are discarded properly.
Environmental Control
Ensure that the hospital has adequate procedures for the routine care, cleaning, and disinfection of
environmental surfaces, beds, bedrails, bedside equipment, and other frequently touched surfaces,
and ensure that these procedures are being followed.
S.F. Dept. Public Health – Infectious Disease Emergencies INFECTION CONTROL, July 2008 Page 3/6
Linen
Handle, transport, and process used linen soiled with blood, body fluids, secretions, and excretions
in a manner that prevents skin and mucous membrane exposures and contamination of clothing,
and that avoids transfer of microorganisms to other patients and environments.
Occupational Health and Bloodborne Pathogens
Take care to prevent injuries when using, cleaning, and disposing of sharp instruments.
Never recap used needles, manipulate them using both hands, or use any other technique that
involves directing the point of a needle toward any part of the body; rather, use either a one-
handed "scoop" technique or a mechanical device designed for holding the needle sheath.
Do not remove used needles from disposable syringes by hand, and do not bend, break, or
otherwise manipulate used needles by hand. Place used sharp items in appropriate puncture-
resistant containers.
Use mouthpieces, resuscitation bags, or other ventilation devices as an alternative to mouth-to-
mouth resuscitation methods in areas where the need for resuscitation is predictable.
Patient Placement
Place a patient who contaminates the environment or who does not (or cannot be expected to)
assist in maintaining appropriate hygiene or environmental control in a private room.
DROPLET PRECAUTIONS
Droplet transmission involves contact of the conjunctivae or the mucous membranes of the nose or
mouth of a susceptible person with large-particle droplets (larger than 5 µm in size) containing
microorganisms generated from the respiratory tract of a person who has a clinical disease or who
is a carrier of the microorganism. Droplets are generated from the source person primarily during
coughing, sneezing, or talking and during the performance of certain procedures such as suctioning
and bronchoscopy.
Transmission via large-particle droplets requires close contact between source and recipient
persons, because droplets do not generally travel long distances through the air, thus special air
handling and ventilation are not required to prevent droplet transmission.
Mask
In addition to wearing a mask as outlined under Standard Precautions, wear a mask when working
within 6-10 ft of the patient. (Logistically, some hospitals may want to implement the wearing of a
mask to enter the room.)
S.F. Dept. Public Health – Infectious Disease Emergencies INFECTION CONTROL, July 2008 Page 4/6
Patient Placement
Place the patient in a private room. If a private room is not available, place the patient in a room
with a patient(s) who has active infection with the same microorganism but with no other infection
(cohorting). When a private room is not available and cohorting is not achievable, maintain spatial
separation of at least 3 ft between the infected patient and other patients and visitors and draw the
curtain between patient beds. Special air handling and ventilation are not necessary, and the door
may remain open.
Patient Transport
Limit the movement and transport of the patient from the room to essential purposes only. If
transport or movement is necessary, minimize patient dispersal of droplets by masking the patient.
CONTACT PRECAUTIONS
Direct-contact transmission involves skin-to-skin contact and physical transfer of microorganisms
to a susceptible host from an infected or colonized person, such as occurs during patient-care
activities that require physical contact. Direct-contact transmission also can occur between two
patients (e.g., skin-to-skin contact), with one serving as the source of infectious microorganisms
and the other as a susceptible host. Indirect-contact transmission involves contact of a susceptible
host with a contaminated intermediate object, an inanimate object or a person, in the patient's
environment.
Patient Placement
Place the patient in a private room. If a private room is not available, place the patient in a room
with a patient(s) who has active infection with the same microorganism but with no other infection
(cohorting).
Gloves and Handwashing
In addition to wearing gloves as outlined under Standard Precautions, wear gloves (clean,
nonsterile gloves are adequate) when entering the room.
Change gloves between tasks and procedures on the same patient after contact with material that
may contain a high concentration of microorganisms. Remove gloves promptly after use, before
touching noncontaminated items and surfaces, and before going to another patient or leaving the
room, and wash hands immediately.
After glove removal and handwashing, ensure that hands do not touch potentially contaminated
environmental surfaces or items in the patient’s room to avoid transfer of microorganisms to other
patients or environments.
S.F. Dept. Public Health – Infectious Disease Emergencies INFECTION CONTROL, July 2008 Page 5/6
Gown
In addition to wearing a gown as outlined under Standard Precautions, wear a gown (a clean,
nonsterile gown is adequate) when entering the room if you anticipate that your clothing will have
substantial contact with the patient, environmental surfaces, or items in the patient's room.
Remove the gown before leaving the patient's environment. After gown removal, ensure that
clothing does not contact potentially contaminated environmental surfaces to avoid transfer of
microorganisms to other patients or environments.
Patient Transport
Limit the movement and transport of the patient from the room to essential purposes only. If the
patient is transported out of the room, ensure that precautions are maintained to minimize the risk
of transmission of microorganisms to other patients and contamination of environmental surfaces
or equipment.
Patient-Care Equipment
When possible, dedicate the use of noncritical patient-care equipment to a single patient (or cohort
of patients infected or colonized with the pathogen requiring precautions) to avoid sharing between
patients. If use of common equipment or items is unavoidable, then adequately clean and disinfect
them before use for another patient.
AIRBORNE PRECAUTIONS
Airborne transmission occurs by dissemination of either airborne droplet nuclei (small-particle
residue [5 µm or smaller in size] of evaporated droplets that may remain suspended in the air for
long periods of time) or dust particles [5 µm or smaller in size] containing the infectious agent.
Microorganisms carried in this manner can be dispersed widely by air currents and may become
inhaled by or deposited on a susceptible host within the same room or over a longer distance from
the source patient, depending on environmental factors; therefore, special air handling and
ventilation are required to prevent airborne transmission.
Patient Placement
Place the patient in a private room that has: 1) monitored negative air pressure in relation to the
surrounding areas; 2) 6 to 12 air changes per hour; and 3) appropriate discharge of air outdoors or
monitored high-efficiency filtration of room air before the air is circulated to other areas in the
hospital. Keep the room door closed and the patient in the room. If a private room is not
available, place the patient in a room with a patient who has active infection with the same
microorganism but with no other infection (unless otherwise recommended).
S.F. Dept. Public Health – Infectious Disease Emergencies INFECTION CONTROL, July 2008 Page 6/6
Respiratory Protection
Wear respiratory protection (N95 respirator) when entering the room of a patient with known or
suspected infection.
Patient Transport
Limit the movement and transport of the patient from the room to essential purposes only. If
transport or movement is necessary, minimize patient dispersal of droplet nuclei by placing a
surgical mask on the patient, if possible.
REFERENCES
Siegel JD, Rhinehart E, Jackson M, Chiarello L, and the Hospital Infection Control Practices Advisory
Committee, 2007 Guideline for Isolation Precautions: Preventing Transmission of Infectious Agents
in Healthcare Settings, June 2007. http://www.cdc.gov/ncidod/dhqp/pdf/isolation2007.pdf
CDC/HICPAC. Hand Hygeine in Health Care Settings. MMWR October 25, 2002 / 51(RR16);1-44
S.F. Dept. Public Health – Infectious Disease Emergencies July 2008 Page 1/1
Covered entities may disclose protected health information (PHI), without individual authorization, to a
public health authority legally authorized to collect or receive the information for the purpose of
preventing or controlling disease, injury or disability 45 CFR 164.512(b). Further, the Privacy Rule
permits covered entities to make disclosures for public health purposes.
Without individual authorization, a covered entity may disclose PHI to a public health authority (or an
entity working under a grant of authority) that is legally authorized to collect or receive the information
for the purposes of preventing or controlling disease, injury, or disability including, but not limited to:
Reporting of disease, injury, and vital events (e.g., birth or death)
Conducting public health surveillance, investigations, and interventions
PHI may also be disclosed without individual authority to:
Report child abuse or neglect to a public health or other government authority legally authorized
to receive such reports
A person subject to jurisdiction of the Food and Drug Administration (FDA) concerning the
quality, safety, or effectiveness of an FDA-related product or activity for which that person has
responsibility
A person who may have been exposed to a communicable disease or may be at risk for
contracting or spreading a disease or condition, when legally authorized to notify the person as
necessary to conduct a public health intervention or investigation
An individual’s employer, under certain circumstances and conditions, as needed for the
employer to meet the requirements of the Occupational Safety and Health Administration, Mine
Safety, and Health Administration or similar state law.
DISCLOSURES TO PUBLIC HEALTH AGENCIES
UNDER THE HIPAA PRIVACY POLICY
S.F. Dept. Public Health – Infectious Disease Emergencies July 2008 Page 1/1
§2500 REPORTING TO THE LOCAL HEALTH AUTHORITY
§2500 (b)
It shall be the duty of every health care provider, knowing of or in attendance on a case or suspected
case of any of the diseases or conditions listed, to report to the local health officer for the jurisdiction
where the patient resides. Where no health care provider is in attendance, any individual having
knowledge of a person who is suspected to be suffering from one of the diseases or conditions listed
may make such a report to the local health officer for the jurisdiction where the patient resides.
§2500 (c)
The administrator of each health facility, clinic or other setting where more than one health care
provider may know of a case, a suspected case or an outbreak of disease within the facility shall
establish and be responsible for administrative procedures to assure that reports are made to the local
health officer.
§2500 (a)(14)
‘Health care provider’ means a physician and surgeon, a veterinarian, a podiatrist, a nurse
practitioner, a physician assistant, a registered nurse, a nurse midwife, a school nurse, an infection
control practitioner, a medical examiner, a coroner, or a dentist.
Excerpted from the California Code of Regulations
Available at: ccr.oal.ca.gov
TITLE 17. CALIFORNIA CODE OF REGULATIONS, §2500
REPORTABLE DISEASE & CONDITIONS
S.F. Dept. Public Health – Infectious Disease Emergencies Page 1/1
San Francisco Department of Public Health
Health Alert Notification Database
The San Francisco Department of Public Health (SFDPH) periodically sends Health Alerts,
Advisories, and Updates to San Francisco clinicians. Health Alerts provide important, timely
information on the recognition, diagnosis, management, and reporting of communicable disease
threats. Recent Health Alert topics have included measles, botulism, MRSA and influenza.
SIGN UP TO RECEIVE HEALTH ALERTS
KEEP YOUR CONTACT INFORMATION UP-TO-DATE
Fax contact information to: (415) 554-2848
OR
Mail contact information to: Health Alert Notification Database Coordinator
San Francisco Department of Public Health
101 Grove Street, Room 408
San Francisco, CA 94102
OR
Complete our online form at: www.sfcdcp.org/registerforalert
Name: ______________________________________________ Degree: __________
Title: _____________ Specialty:_________________________________________
Company/Organization: ________________________________________________
Department: _________________________________________________________
Address: ____________________________________________________________
City: _________________________________________________ Zip: __________
Business Fax: ____________________ Business Phone: _____________________
Pager: __________________________ Mobile: _____________________________
Email: _______________________________________________________________
Note: All contact information provided to SFDPH is kept confidential.
Communicable Disease Control & Prevention
101 Grove Street, Room 408 y San Francisco, CA 94102
Phone: (415) 554-2830 y Fax: (415) 554-2848 y http://www.sfcdcp.org
2/16/2005
CITY AND COUNTY OF SAN FRANCISCO For Laboratory Use Only
PUBLIC HEALTH LABORATORY
101 Grove Street, Room 419
San Francisco, CA 94102 ______________________ __________________
Sally Liska, Dr. P.H., Lab Director Laboratory Number Date/Time Received
Tel: (415) 554-2800 Fax: (415) 431-0651
CLIA ID # 05D0643643
PLEASE ATTACH PRE-PRINTED LABEL or PRINT CLEARLY
Patient’s Name: ___________________________________________Gender: ____DOB: ____________Race/Ethnicity:____________
Last, First
Address: __________________________________________________________________ Phone: ______________________
City / State:________________________________________________________________ Zip Code: _____________________
Submitted By: _________________________________________ Requesting Clinician: ____________________________________
(Clinic)
Medical Record #: _____________________________Medi-Cal/HAP #: _____________________S-Code #: ___________________
Bill To: Submitter Medi-Cal Family Planning Private Pay
CHECK BOTH SOURCE AND TEST REQUESTED; INDICATE DATE COLLECTED
SPECIMEN SOURCE: DATE SPECIMEN TAKEN: _______________________________
Blood Urine Rectal Throat Rash/Lesion Culture
Serum Cervix Feces Sputum CSF Slide
Plasma Urethral Genital Nasopharyngeal Oral Fluid Other: _______________
BACTERIOLOGY MYCOBACTERI
A
CHLAMYDIA AND GONORRHE
A
Gonorrhea Screen Quantiferon (TB infection blood test)*
Chlamydia
Enteric Screen
A reason for Quantiferon testing MUST
be checked:
Gonorrhea
Special Bacteriology Culture
Immunocompromised
TB suspect Foreign-born
Contact to TB Homeless
Diabetes IVDU
Renal failure Program clearance
School clearance (US born)
Other _______________________
A reason for CT/GC testing MUST be
checked:
Clearance for: ______________
Age 25 MSM screen
Prior CT/GC Infection Pregnant
Cervicitis/Urethritis IUD
Contact to STD
Other__________________
Other: ____________________
Acid Fast Smear
PARASITOLOGY
Specimen for Isolation
SEROLOGY
Ova and Parasites
Culture for Identification
Syphilis - VDRL Syphilis - TP-PA
Clearance for: ______________
TB Susceptibility
Rubella IgG Hepatitis C Antibody
Blood Smear (e.g. Malaria)**
Direct Amplification Test
Herpes Simplex-2 Other:
___________
Cryptosporidia
V
IROLOGY
Cyclospora
HIV VIRAL LOAD (bDNA)*
Herpes Culture
Other: _____________________ Time Collected: ______________ Other: _____________________
Comment: _______________________________________________________________________________________
_________________________________________________________________________________________________
* Specimens have time limitations for submission. Contact laboratory for details.
**Travel History Required
State of California—Health and Human Services Agency Department of Public Health
CONFIDENTIAL MORBIDITY REPORT
NOTE: For STD, Hepatitis, or TB, complete appropriate section below. Special reporting requirements and reportable diseases on back.
PM 110 (SF 12/07)
DISEASE BEING REPORTED:__________________________________________________________________________________
Patient’s Last Name Social Security Number
Birth Date
First Name/Middle Name (or initial) Month Day Year
Age
Address: Number, Street Apt./Unit Number
City/Town State ZIP Code Country of Birth
Estimated Delivery Date
Area Code Home Telephone Gender Pregnant? Month Day Year
Area Code Work Telephone Patient’s Occupation/Setting
r Food service r Day care r Correctional facility
r Health care r School r Other _________________________
DATE OF ONSET
Reporting Health Care Provider
REPORT TO
Month Day Year
Reporting Health Care Facility
DATE DIAGNOSED Address
Month Day Year
City State ZIP Code
DATE OF DEATH Telephone Number Fax
Month Day Year ( ) ( )
Submitted by Date Submitted
(Month/Day/Year) (Obtain additional forms from your local health department.)
SEXUALLY TRANSMITTED DISEASES (STD) VIRAL HEPATITIS Not
Syphilis Syphilis Test Results
Pos Neg Pend Done
r Primary (lesion present) r Late latent > 1 year r RPR Titer:__________
r
Hep A anti-HAV IgM
rrrr
Gender of Sex Partners last 12 months:
r Male
r Transgender (M to F)
r Female r Transgender (F to M)
r
Other: ________
r
Rectal
r
Hep D (Delta) anti-Delta
r r r r
STD TREATMENT INFORMATION r Untreated r Other: ______________ rrrr
r Treated (Drugs, Dosage, Route ): Date Treatment Initiated r Will treat
Suspected Exposure Type
Month Day Year r Unable to contact patient
r
Blood
r
Other needle
r
Sexual
r
Household
_________________________
r Refused treatment
transfusion exposure contact contact
_________________________
r Referred to: _________________
r
Child care
r
Other: ________________________________
TUBERCULOSIS (TB) TB TREATMENT INFORMATION
Status Mantoux TB Skin Test Bacteriology r Current Treatment
r Active Disease Month Day Year Month Day Year r INH r RIF r PZA
r Confirmed r EMB r Other:____________
r Suspected Date Performed Date Specimen Collected Month Day Year
r Infected, No Disease r Pending Date Treatment
r Convertor Results:______________ mm r Not Done Source _______________________________________ Initiated
r Reactor Smear: r Pos r Neg r Pending r Not done
Chest X-Ray Month Day Year Culture: r Pos r Neg r Pending r Not done r Untreated
Site(s) r Will treat
r Pulmonary
Date Performed
Other test(s) ___________________________________ r Unable to contact patient
r Extra-Pulmonary r Normal r Pending r Not done r Refused treatment
r Both r Cavitary r Abnormal/Noncavitary _______________________________________ r Referred to: _____________________
REMARKS
M F
Y
Ethnicity (3 one)
r Hispanic/Latino
r Non-Hispanic/Non-Latino
Race (3 one)
r African-American/Black
r Asian/Pacific Islander (3 one)
r Asian-Indian r Japanese
r Cambodian r Korean
r Chinese r Laotian
r Filipino r Samoan
r Guamanian r Vietnamese
r Hawaiian r Other_________
_____________
r Native American/Alaskan Native
r White: __________________________
r Other: __________________________
Unk
N
r Secondary r Late (tertiary) r VDRL Titer:__________ r Hep B HBsAg rrrr
r Early latent < 1 year r Congenital r FTA/MHA: r Pos r Neg r Acute anti-HBc
rrrr
r Latent (unknown duration) r CSF-VDRL: r Pos r Neg r Chronic anti -HBc IgM
rrrr
r Neurosyphilis r Other:_________________ anti -HBs
rrrr
Chlamydia Site:
r
r Hep C anti -HCV rrrr
r
Gonorrhea
r
Pharyngeal Urethral/Cervical
r
r Acute
PCR-HCV
rrrr
r
Chancroid
r
Chronic
r
Unknown
r
Refused
r
Urine
r
PID
(PH): (415) 554-9050
Communicable Disease Control Unit
San Francisco Dept of Public Health
101 Grove Street, Room 408
San Francisco, CA 94102 
STD (Fax): (415) 431-4628
TB (Fax): (415) 648-8369
AIDS/HIV
PHONE: (415) 554-2830
FAX: (415) 554-2848