Manual of Laboratory Safety Ramnik Sood, Najat Rashid
INDEX
×
Chapter Notes

Save Clear


BiosafetyChapter One

 
INTRODUCTION
The purpose of this manual is to be a resource for information, guidelines, policies, and procedures that will enable and encourage those working in the laboratory environment to work safely and reduce or eliminate the potential for exposure to biological, chemical and radioactive materials hazard.
The goal of the laboratory safety is to minimize the risk of injury and illness to laboratory workers by ensuring they have the training, information, support and equipment needed.
The manual promotes safe and practical laboratory procedures, included laboratory biosafety, laboratory biosecurity, microbiological risk assessment, laboratory biosafety levels, information on the use of personal protective equipment, laboratory animal facilities, laboratories equipment, laboratory techniques, hazard communication and packing of infectious substances, biosafety and biotechnology, the proper use of disinfection and sterilization, the use and storage of chemicals and radioactive materials, biosafety officer and biosafety committee and the proper methods of waste disposal.
Finally, emphasis must be placed on the practices and procedures used by trained laboratory staff. Since “no biosafety cabinet or other facility or procedure alone guarantees safety unless the users operate safe techniques based on informed understanding.” It is the responsibility of everyone, including managers and laboratory workers, to use the information available in these manual and to perform their work in a safe and secure manner.
 
LABORATORY BIOSAFETY
‘Laboratory biosafety’ is the term used to describe the containment principles, technologies and practices that are implemented to prevent unintentional exposure to pathogens and toxins, or their accidental release.
A laboratory biosafety goal is to ensure that hazardous materials will be handled and disposed of in such a way that people, other living organisms, and the environment are protected from harm. Safety awareness must be a part of everyone's habits, and can only be achieved if all senior and responsible staff has a sincere, visible, and continuing interest in preventing injuries and occupational illnesses.
2
Laboratory facilities are designated as: Basic Biosafety Level 1 (BSL-1), Basic Biosafety Level 2 (BSL-2), Containment Biosafety Level 3 (BSL-3) and Maximum containment Biosafety Level 4 (BSL-4).
 
Biosafety Level 1
Practices, safety equipment, and facility design and construction are appropriate for undergraduate and secondary educational training and teaching laboratories, and for other laboratories in which work is done with defined and characterized strains of viable microorganisms not known to consistently cause disease in healthy adult humans. Bacillus subtilis, Naegleria gruberi, infectious canine hepatitis virus, and exempt organisms.
BSL-1 represents a basic level of containment that relies on standard microbiological practices with no special primary or secondary barriers recommended, other than a sink for handwashing.
 
Biosafety Level 2
Practices, equipment, and facility design and construction are applicable to clinical, diagnostic, teaching, and other laboratories in which work is done with the broad spectrum of indigenous moderate-risk agents that are present in the community and associated with human disease of varying severity. With good microbiological techniques, these agents can be used safely in activities conducted on the open bench, provided the potential for producing splashes or aerosols is low. Hepatitis B virus, HIV, the Salmonella, and Toxoplasma are representative of microorganisms to this containment level. BSL-2 is appropriate when work is done with any human-derived blood, body fluids, tissues, or primary human cell lines where the presence of an infectious agent may be unknown.
  • Primary hazards to personnel working with these agents relate to accidental percutaneous or mucous membrane exposures, or ingestion of infectious materials. Extreme caution should be taken with contaminated needles or sharp instruments. Even though organisms routinely manipulated at BSL-2 are not known to be transmissible by the aerosol route, procedures with aerosol or high splash potential that may increase the risk of such personnel exposure must be conducted in primary containment equipment, or in devices such as a BSC or safety centrifuge cups. Personal protective equipment should be used as appropriate, such as splash shields, face protection, gowns, and gloves.
  • Secondary barriers, such as hand washing sinks and waste decontamination facilities, must be available to reduce potential environmental contamination.
 
Biosafety Level 3
Practices, safety equipment, and facility design and construction are applicable to clinical, diagnostic, teaching, research, or production facilities in which work is done with indigenous or exotic agents with a potential for respiratory transmission, and which may cause serious and potentially lethal infection. Mycobacterium tuberculosis, St. Louis encephalitis virus, and Coxiella burnetii are representative of the microorganisms assigned to this level. Primary hazards to personnel working with these agents relate to autoinoculation, ingestion, and exposure to infectious aerosols. At BSL-3, more emphasis is placed on:
3
  • Primary barriers to protect personnel in contiguous areas, the community, and the environment from exposure to potentially infectious aerosols. For example, all laboratory manipulations should be performed in a BSC or other enclosed equipment, such as a gas-tight aerosol generation chamber.
  • Secondary barriers for this level include controlled access to the laboratory and ventilation requirements that minimize the release of infectious aerosols from the laboratory.
 
Biosafety Level 4
Practices, safety equipment, and facility design and construction are applicable for work with dangerous and exotic agents that pose a high individual risk of life-threatening disease, which may be transmitted via the aerosol route and for which there is no available vaccine or therapy. Agents with a close or identical antigenic relationship to BSL-4 agents also should be handled at this level. When sufficient data are obtained, work with these agents may continue at this level or at a lower level. Viruses such as Marburg or Congo-Crimean hemorrhagic fever are manipulated at BSL-4.
Biosafety level designations are based on a composite of the design features, construction, containment facilities, equipment, practices and operational procedures required for working with agents from the various risk groups. The primary hazards to personnel working with BSL-4 agents are respiratory exposure to infectious aerosols, mucous membrane or broken skin exposure to infectious droplets, and autoinoculation. All manipulations of potentially infectious diagnostic materials, isolates, and naturally or experimentally infected animals, pose a high-risk of exposure and infection to laboratory personnel, the community, and the environment.
The assignment of an agent to a biosafety level for laboratory work must be based on a risk assessment. Such an assessment will take the risk group as well as other factors into consideration in establishing the appropriate biosafety level. For example, an agent that is assigned to Risk Group 2 may generally require Biosafety Level 2 facilities, equipment, practices and procedures for safe conduct of work. However, if particular experiments require the generation of high-concentration aerosols, then Biosafety Level 3 may be more appropriate to provide the necessary degree of safety, since it ensures superior containment of aerosols in the laboratory workplace. The biosafety level assigned for the specific work to be done is therefore driven by professional judgment based on a risk assessment, rather than by automatic assignment of a laboratory biosafety level according to the particular risk group designation of the pathogenic agent to be used.
Classification of infective microorganisms by risk group:
  • Risk Group 1 (no or low individual and community risk): A microorganism that is unlikely to cause human or animal disease.
  • Risk Group 2 (moderate individual risk, low community risk): A pathogen that can cause human or animal disease but is unlikely to be a serious hazard to laboratory workers, the community, livestock or the environment. Laboratory exposures may cause serious infection, but effective treatment and preventive measures are available and the risk of spread of infection is limited.
    4
  • Risk Group 3 (high individual risk, low community risk): A pathogen that usually causes serious human or animal disease but does not ordinarily spread from one infected individual to another. Effective treatment and preventive measures are available.
  • Risk Group 4 (high individual and community risk): A pathogen that usually causes serious human or animal disease and that can be readily transmitted from one individual to another, directly or indirectly. Effective treatment and preventive measures are not usually available.
Effective biosafety practices are the very foundation of “laboratory biosecurity” activities.
In the absence of careful implementation, various aspects of biosafety may conflict with laboratory biosecurity. For example, controls that reduce unauthorized access might also hinder an emergency response by fire or rescue personnel. Mechanisms need to be established that allow entry by emergency responders but ensure uninterrupted and constant laboratory biosecurity, control and accountability.
Signage may also represent a potential conflict between biosafety and laboratory biosecurity. In the past, biohazard signs placed on laboratory doors identified the biological agents present in the laboratory. However, as a laboratory biosecurity measure to better protection it is recommended certain information on biohazard signs to the laboratory biosafety level, the name and telephone number of the responsible investigator, and emergency contact information (Table 1.1).
 
MICROBIOLOGICAL RISK-ASSESSMENT
The backbone of the practice of biosafety is risk-assessment. While there are many tools available to assist in the assessment of risk for a given procedure or experiment, the most important component is professional judgment. Risk assessments should be performed by the individuals most familiar with the specific characteristics of the organisms being considered for use, the equipment and procedures to be employed, animal models that may be used, and the containment equipment and facilities available. The laboratory director or principal investigator is responsible for ensuring that adequate and timely risk assessments are performed, and for working closely with the institution's safety committee and biosafety personnel to ensure that appropriate equipment and facilities are available to support the work being considered. Once performed, risk assessments should be reviewed routinely and revised when necessary, taking into consideration the acquisition of new data having a bearing on the degree of risk and other relevant new information from the scientific literature. One of the most helpful tools available for performing a microbiological risk assessment is the listing of risk groups for microbiological agents. However, simple reference to the risk grouping for a particular agent is insufficient in the conduct of a risk assessment. Other factors that should be considered, as appropriate, include:
  1. Pathogenicity of the agent and infectious dose.
  2. Natural route of infection.
  3. Other routes of infection, resulting from laboratory manipulations (parenteral, airborne, ingestion).
  1. Stability of the agent in the environment.
    5
    Table 1.1   Biosafety level requirements
    Biosafety level
    1
    2
    3
    4
    Isolationa of laboratory
    No
    No
    Yes
    Yes
    Room sealable for decontamination
    No
    No
    Yes
    Yes
    Ventilation:
    — Inward airflow
    No
    Desirable
    Yes
    Yes
    — Controlled ventilating system
    No
    Desirable
    Yes
    Yes
    — HEPA-filtered air exhaust
    No
    No
    Yes/Nob
    Yes
    Double-door entry
    No
    No
    Yes
    Yes
    Airlock
    No
    No
    No
    Yes
    Airlock with shower
    No
    No
    No
    Yes
    Anteroom
    No
    No
    Yes
    Anteroom with shower
    No
    No
    Yes/Noc
    No
    Effluent treatment
    No
    No
    Yes/Noc
    Yes
    Autoclave:
    — On site
    No
    Desirable
    Yes
    Yes
    — In laboratory room
    No
    No
    Desirable
    Yes
    — Double-ended
    No
    No
    Desirable
    Yes
    Biological safety cabinets
    No
    Desirable
    Yes
    Yes
    Personnel safety monitoring capabilityd
    No
    No
    Desirable
    Yes
    a Environmental and functional isolation from general traffic
    b Dependent on location of exhaust
    c Dependent on agent(s) used in the laboratory
    d For example, window, closed-circuit television, two-way communication
  2. Concentration of the agent and volume of concentrated material to be manipulated.
  3. Presence of a suitable host (human or animal).
  4. Information available from animal studies and reports of laboratory-acquired infections or clinical reports.
  5. Laboratory activity planned for example (sonication, aerosolization, centrifugation, etc.).
  6. Any genetic manipulation of the organism that may extend the host range of the agent or alter the agent's sensitivity to known, effective treatment regimens.
  7. Local availability of effective prophylaxis or therapeutic interventions.
On the basis of the information ascertained during the risk assessment, a biosafety level can be assigned to the planned work, appropriate personal protective equipment selected, and standard operating procedures (SOPs) incorporating other safety interventions developed to ensure the safest possible conduct of the work.
6
 
Specimens for which there is Limited Information
The risk assessment procedure described above works well when there is adequate information available. However, there are situations when the information is insufficient to perform an appropriate risk assessment, for example, with clinical specimens or epidemiological samples collected in the field. In these cases, it is prudent to take a cautious approach to specimen manipulation.
  1. Standard precautions should always be followed, and barrier protections applied (gloves, gowns, eye protection), whenever samples are obtained from patients.
  2. Basic containment, Biosafety Level 2 practices and procedures should be the minimum requirement for handling specimens.
  3. Transport of specimens should follow national and/or international rules and regulations.
 
Some Information may be Available to Assist in Determining the Risk of Handling these Specimens
  1. Medical data on the patient.
  2. Epidemiological data (morbidity and mortality data, suspected route of transmission, other outbreak investigation data).
  3. Information on the geographical origin of the specimen.
 
Emergency Procedures in Microbiological Laboratories
 
Puncture Wounds, Cuts and Abrasions
The affected individual should remove protective clothing, wash the hands and any affected area(s), apply an appropriate skin disinfectant, and seek medical attention as necessary. The cause of the wound and the organisms involved should be reported, and appropriate and complete medical records kept.
 
Ingestion of Potentially Infectious Material
Protective clothing should be removed and medical attention sought. Identification of the material ingested and circumstances of the incident should be reported, and appropriate and complete medical records kept.
 
Potentially Infectious Aerosol Release (Outside a Biological Safety Cabinet)
All persons should immediately vacate the affected area and any exposed persons should be referred for medical advice. The laboratory supervisor and the biosafety officer should be informed at once. No one should enter the room for an appropriate amount of time (e.g. 1 h), to allow aerosols to be carried away and heavier particles to settle. If the laboratory does not have a central air exhaust system, entrance should be delayed (e.g. for 24 h).
Signs should be posted indicating that entry is forbidden. After the appropriate time, decontamination should proceed, supervised by the biosafety officer. Appropriate protective clothing and respiratory protection should be worn.7
 
Broken Containers and Spilled Infectious Substances
Broken containers contaminated with infectious substances and spilled infectious substances should be covered with a cloth or paper towels. Disinfectant should then be poured over these and left for the appropriate amount of time. The cloth or paper towels and the broken material can then be cleared away; glass fragments should be handled with forceps. The contaminated area should then be swabbed with disinfectant. If dustpans are used to clear away the broken material, they should be autoclaved or placed in an effective disinfectant. Cloths, paper towels and swabs used for cleaning up should be placed in a contaminated-waste container. Gloves should be worn for all these procedures.
 
Breakage of Tubes Containing Potentially Infectious Material in Centrifuges not having Sealable Buckets
If a breakage occurs or is suspected while the machine is running, the motor should be switched off and the machine left closed (e.g. for 30 minutes) to allow settling. If a breakage is discovered after the machine has stopped, the lid should be replaced immediately and left closed (e.g. for 30 minutes). In both instances, the biosafety officer should be informed.
Strong (e.g. thick rubber) gloves, covered if necessary with suitable disposable gloves, should be worn for all subsequent operations. Forceps, or cotton held in the forceps, should be used to retrieve glass debris.
All broken tubes, glass fragments, buckets, and the rotor should be placed in a noncorrosive disinfectant known to be active against the organisms concerned). Unbroken, capped tubes may be placed in disinfectant in a separate container and recovered.
The centrifuge bowl should be swabbed with the same disinfectant, at the appropriate dilution, and then swabbed again, washed with water and dried. All materials used in the clean-up should be treated as infectious waste.
 
Breakage of Tubes Inside Sealable Buckets (Safety Cups)
All sealed centrifuge buckets should be loaded and unloaded in a biological safety cabinet. If breakage is suspected within the safety cup, the safety cap should be loosened the bucket autoclaved. Alternatively, the safety cup may be chemically disinfected.
 
LABORATORIES BIOSAFETY LEVELS
For the purposes of this manual, the guidance and recommendations given as minimum requirements pertaining to laboratories of all biosafety levels are directed at microorganisms in Risk Groups 1–4. Although some of the precautions may appear to be unnecessary for some organisms in Risk Group 1, they are desirable for training purposes to promote good (i.e. safe) microbiological techniques GMT. Diagnostic and health-care laboratories (public health, clinical or hospital-based) must all be designed for Biosafety Level 2 or above. As no laboratory has complete control over the specimens it receives, laboratory workers may be exposed to organisms in higher risk groups than anticipated. This possibility must be recognized in the 8development of safety plans and policies. In some countries, accreditation of clinical laboratories is required. Globally, standard precautions should always be adopted and practiced.
The guidelines for basic laboratories, Biosafety Levels 1 and 2 presented here are comprehensive and detailed, as they are fundamental to laboratories of all biosafety levels. The guidelines for containment laboratories, Biosafety Level 3 and maximum containment laboratories, Biosafety Level 4 are modifications of and additions to these guidelines, designed for work with the more dangerous (hazardous) pathogens.
 
Code of Practice
This code is a listing of the most essential laboratory practices and procedures that are basic to GMT. In many laboratories and national laboratory programs, this code may be used to develop written practices and procedures for safe laboratory operations. Each laboratory should adopt a safety or operations manual that identifies known and potential hazards, and specifies practices and procedures to eliminate or minimize such hazards. GMT is fundamental to laboratory safety. Specialized laboratory equipment is a supplement to but can never replace appropriate procedures. The most important concepts are listed below:
 
Access
  1. The international biohazard warning symbol and sign must be displayed on the doors of the rooms where microorganisms of Risk Group 2 or higher risk groups are handled (Figure 1.1).
  2. Only authorized persons should be allowed to enter the laboratory working areas.
  3. Laboratory doors should be kept closed.
  4. Children should not be authorized or allowed to enter laboratory working areas.
  5. Access to animal houses should be specially authorized.
  6. No animals should be admitted other than those involved in the work of the laboratory.
 
Personal Protection
  1. Laboratory coveralls, gowns or uniforms must be worn at all times for work in the laboratory.
  2. Appropriate gloves must be worn for all procedures that may involve direct or accidental contact with blood, body fluids and other potentially infectious materials or infected animals. After use, gloves should be removed aseptically and hands must then be washed.
  3. Personnel must wash their hands after handling infectious materials and animals, and before they leave the laboratory working areas.
  4. Safety glasses, face shields (visors) or other protective devices must be worn when it is necessary to protect the eyes and face from splashes, impacting objects and sources of artificial ultraviolet radiation.
  5. It is prohibited to wear protective laboratory clothing outside the laboratory, e.g. in canteens, coffee rooms, offices, libraries, staff rooms and toilets.
    9
    zoom view
    Figure 1.1: Biohazard warning sign for laboratory doors
  6. Open-toed footwear must not be worn in laboratories.
  7. Eating, drinking, smoking, applying cosmetics and handling contact lenses is prohibited in the laboratory working areas.
  8. Storing human foods or drinks anywhere in the laboratory working areas is prohibited.
  9. Protective laboratory clothing that has been used in the laboratory must not be stored in the same lockers or cupboards as street clothing.
 
Procedures
  1. Pipetting by mouth must be strictly forbidden.
  2. Materials must not be placed in the mouth. Labels must not be licked.
  3. All technical procedures should be performed in a way that minimizes the formation of aerosols and droplets.
  4. The use of hypodermic needles and syringes should be limited. They must not be used as substitutes for pipetting devices or for any purpose other than parenteral injection or aspiration of fluids from laboratory animals.
  5. All spills, accidents and overt or potential exposures to infectious materials must be reported to the laboratory supervisor. A written record of such accidents and incidents should be maintained.
  6. A written procedure for the clean-up of all spills must be developed and followed.
    10
  7. Contaminated liquids must be decontaminated (chemically or physically) before discharge to the sanitary sewer. An effluent treatment system may be required, depending on the risk assessment for the agent(s) being handled.
  8. Written documents that are expected to be removed from the laboratory need to be protected from contamination while in the laboratory.
 
Laboratory Working Areas
  1. The laboratory should be kept neat, clean and free of materials that are not pertinent to the work.
  2. Work surfaces must be decontaminated after any spill of potentially dangerous material and at the end of the working day.
  3. All contaminated materials, specimens and cultures must be decontaminated before disposal or cleaning for reuse.
  4. Packing and transportation must follow applicable national and/or international regulations.
  5. When windows can be opened, they should be fitted with arthropod-proof screens.
 
Biosafety Management
  1. It is the responsibility of the laboratory director (the person who has immediate responsibility for the laboratory) to ensure the development and adoption of a biosafety management plan and a safety or operations manual.
  2. The laboratory supervisor (reporting to the laboratory director) should ensure that regular training in laboratory safety is provided.
  3. Personnel should be advised of special hazards, and required to read the safety or operations manual and follow standard practices and procedures. The laboratory supervisor should make sure that all personnel understand these. A copy of the safety or operations manual should be available in the laboratory.
  4. There should be an arthropod and rodent control program.
  5. Appropriate medical evaluation, surveillance and treatment should be provided for all personnel in case of need, and adequate medical records should be maintained.
 
Laboratory Design and Facilities
In designing a laboratory and assigning certain types of work to it, special attention should be paid to conditions that are known to pose safety problems. These include:
  1. Formation of aerosols.
  2. Work with large volumes and/or high concentrations of microorganisms.
  3. Overcrowding and too much equipment.
  4. Infestation with rodents and arthropods.
  5. Unauthorized entrance.
  6. Workflow: Use of specific samples and reagents.
 
Laboratory Equipment
Together with good procedures and practices, the use of safety equipment will help to reduce risks when dealing with biosafety hazards. This section deals with 11basic principles related to equipment suitable for laboratories of all biosafety levels.
The laboratory director should, after consultation with the biosafety officer and safety committee (if designated), ensure that adequate equipment is provided and that it is used properly. Equipment should be selected to take account of certain general principles, i.e. it should be:
  1. Designed to prevent or limit contact between the operator and the infectious material.
  2. Constructed of materials that are impermeable to liquids, resistant to corrosion and meet structural requirements.
  3. Fabricated to be free of burrs, sharp edges and unguarded moving parts.
  4. Designed, constructed and installed to facilitate simple operation and provide for ease of maintenance, cleaning, decontamination and certification testing. Glassware and other breakable materials should be avoided, whenever possible.
 
Essential Biosafety Equipment
  1. Pipetting aids, to avoid mouth pipetting. Many different designs are available.
  2. Biological safety cabinets, to be used whenever:
    • Infectious materials are handled. Such materials may be centrifuged in the open laboratory if sealed centrifuge safety cups are used and if they are loaded and unloaded in a biological safety cabinet there is an increased risk of airborne infection.
    • Procedures with a high potential for producing aerosols are used. These may include centrifugation, grinding, blending, vigorous shaking or mixing, sonic disruption, opening of containers of infectious materials (whose internal pressure may be different from the ambient pressure), intranasal inoculation of animals, and harvesting of infectious tissues from animals and eggs.
  3. Plastic disposable transfer loops. Alternatively, electric transfer loop incinerators may be used inside the biological safety cabinet to reduce aerosol production.
  4. Screw-capped tubes and bottles.
  5. Autoclaves or other appropriate means to decontaminate infectious materials.
  6. Plastic disposable Pasteur pipettes, whenever available, to avoid glass.
  7. Equipment such as autoclaves and biological safety cabinets must be validated with appropriate methods before being taken into use.
 
Guidelines for the Surveillance of Laboratory Workers Handling Microorganisms at Biosafety Level 1
Historical evidence indicates that the microorganisms handled at this level are unlikely to cause human disease or animal disease of veterinary importance. Ideally, however, all laboratory workers should undergo a pre-employment health check at which their medical history is recorded. Prompt reporting of illnesses or laboratory accidents is desirable and all staff members should be made aware of the importance of maintaining GMT.12
 
Guidelines for the Surveillance of Laboratory Workers Handling Microorganisms at Biosafety Level 2
  1. A pre-employment or preplacement health check is necessary. The person's medical history should be recorded and a targeted occupational health assessment performed.
  2. Records of illness and absence should be kept by the laboratory management.
  3. Women of childbearing age should be made aware of the risk to an unborn child of occupational exposure to certain microorganisms, e.g. rubella virus. The precise steps taken to protect the fetus will vary, depending on the microorganisms to which the women may be exposed.
 
Training
Human error and poor technique can compromise the best of safeguards to protect the laboratory worker. Thus, a safety conscious staff, well-informed about the recognition and control of laboratory hazards, is key to the prevention of laboratory acquired infections, incidents and accidents. For this reason, continuous in-service training in safety measures is essential. An effective safety program begins with the laboratory managers, who should ensure that safe laboratory practices and procedures are integrated into the basic training of employees. Training in safety measures should be an integral part of new employees' introduction to the laboratory. Employees should be introduced to the code of practice and to local guidelines, including the safety or operations manual. Measures to assure that employees have read and understood the guidelines, such as signature pages, should be adopted. Laboratory supervisors play the key role in training their immediate staff in good laboratory techniques. The biosafety officer can assist in training and with the development of training aids and documentation.
Staff training should always include information on safe methods for highly hazardous procedures that are commonly encountered by all laboratory personnel and which involve:
  1. Inhalation risks (i.e. aerosol production) when using loops, streaking agar plates, pipetting, making smears, opening cultures, taking blood/serum samples, centrifuging, etc.
  2. Ingestion risks when handling specimens, smears and cultures.
  3. Risks of percutaneous exposures when using syringes and needles.
  4. Bites and scratches when handling animals.
  5. Handling of blood and other potentially hazardous pathological materials.
  6. Decontamination and disposal of infectious material.
 
Waste Handling
Waste is anything that is to be discarded. In laboratories, decontamination of wastes and their ultimate disposal are closely interrelated. In terms of daily use, few if any contaminated materials will require actual removal from the laboratory or destruction. Most glassware, instruments and laboratory or clothing will be reused or recycled.
The overriding principle is that all infectious materials should be decontaminated, autoclaved or incinerated within the laboratory.
13
The principal questions to be asked before discharge of any objects or materials from laboratories that deal with potentially infectious microorganisms or animal tissues are:
  1. Have the objects or materials been effectively decontaminated or disinfected by an approved procedure?
  2. If not, have they been packaged in an approved manner for immediate on-site incineration or transfer to another facility with incineration capacity?
  3. Does the disposal of the decontaminated objects or materials involve any additional potential hazards, biological or otherwise, to those who carry out the immediate disposal procedures or who might come into contact with discarded items outside the facility?
 
Decontamination
Steam autoclaving is the preferred method for all decontamination processes. Materials for decontamination and disposal should be placed in containers, e.g. autoclavable plastic bags, that are color-coded according to whether the contents are to be autoclaved and/or incinerated. Alternative methods may be envisaged only if they remove and/or kill microorganisms.
 
Handling and Disposal Procedures for Contaminated Materials and Wastes
Identification and separation system for infectious materials and their containers should be adopted. National and international regulations must be followed. Categories should include:
  1. Non-contaminated (non-infectious) waste that can be reused or recycled or disposed of as general, “household” waste.
  2. Contaminated (infectious) “sharps”—hypodermic needles, scalpels, knives and broken glass; these should always be collected in puncture proof containers fitted with covers and treated as infectious.
  3. Contaminated material for decontamination by autoclaving and thereafter washing and reuse or recycling.
  4. Contaminated material for autoclaving and disposal.
  5. Contaminated material for direct incineration.
 
Sharps
After use, hypodermic needles should not be recapped, clipped or removed from disposable syringes. The complete assembly should be placed in a sharps disposal container.
Disposable syringes, used alone or with needles, should be placed in sharps disposal containers and incinerated, with prior autoclaving if required.
Sharps disposal containers must be puncture-proof/resistant and must not be filled to capacity. When they are three-quarters full they should be placed in “infectious waste” containers and incinerated, with prior autoclaving if laboratory practice requires it. Sharps disposal containers must not be discarded in landfills.14
 
Contaminated Materials for Autoclaving and Reuse
No precleaning should be attempted of any contaminated materials to be autoclaved and reused. Any necessary cleaning or repair must be done only after autoclaving or disinfection.
 
Contaminated Materials for Disposal
Apart from sharps, which are dealt with above, all contaminated materials should be autoclaved in leak proof containers, e.g. autoclavable, color-coded plastic bags, before disposal.
After autoclaving, the material may be placed in transfer containers for transport to the incinerator. If possible, materials deriving from health care activities should not be discarded in landfills even after decontamination. If an incinerator is available on the laboratory site, autoclaving may be omitted. The contaminated waste should be placed in designated containers (e.g. color coded bags) and transported directly to the incinerator. Reusable transfer containers should be leak proof and have tight-fitting covers. They should be disinfected and cleaned before they are returned to the laboratory for further use.
Discard containers, pans or jars, preferably unbreakable (e.g. plastic), should be placed at every work station. When disinfectants are used, waste materials should remain in intimate contact with the disinfectant (i.e. not protected by air bubbles) for the appropriate time, according to the disinfectant used. The discard containers should be decontaminated and washed before reuse. Incineration of contaminated waste must meet with the approval of the public health and air pollution authorities, as well as that of the laboratory biosafety officer.
 
Biosafety Level 3 (The Containment Laboratory)
The containment laboratory, Biosafety Level 3 is designed and provided for work with Risk Group 3 microorganisms and with large volumes or high concentrations of Risk Group 2 microorganisms that pose an increased risk of aerosol spread. Biosafety Level 3 containment requires the strengthening of the operational and safety program over and above those for basic laboratories, Biosafety Levels 1 and 2.
The guidelines given in this chapter are presented in the form of additions to those for basic laboratories, Biosafety Levels 1 and 2, which must therefore be applied before those specific for the containment laboratory, Biosafety Level 3.
 
The Major Additions and Changes
  1. Code of practice
  2. Laboratory design and facilities
  3. Health and medical surveillance.
Laboratories in this category should be registered or listed with the national or other appropriate health authorities.
 
Code of Practice
The code of practice for basic laboratories, Biosafety Levels 1 and 2 applies except where modified as follows:15
  1. The international biohazard warning symbol and sign displayed on laboratory access doors must identify the biosafety level and the name of the laboratory supervisor who controls access, and indicate any special conditions for entry into the area, e.g. immunization.
  2. Laboratory protective clothing must be of the type with solid-front or wrap-around gowns, scrub suits, coveralls, head covering and, where appropriate, shoe covers or dedicated shoes. Front-buttoned standard laboratory coats are unsuitable, as are sleeves that do not fully cover the forearms. Laboratory protective clothing must not be worn outside the laboratory, and it must be decontaminated before it is laundered. The removal of street clothing and change into dedicated laboratory clothing may be warranted when working with certain agents (e.g. agricultural or zoonotic agents).
  3. Open manipulations of all potentially infectious material must be conducted within a biological safety cabinet or other primary containment device.
  4. Respiratory protective equipment may be necessary for some laboratory Procedures or working with animals infected with certain pathogens.
 
Laboratory Design and Facilities
The laboratory design and facilities for basic laboratories—Biosafety Levels 1 and 2 apply except where modified as follows:
  1. The laboratory must be separated from the areas that are open to unrestricted traffic flow within the building. Additional separation may be achieved by placing the laboratory at the blind end of a corridor, or constructing a partition and door or access through an anteroom (e.g. a double-door entry or basic laboratory—Biosafety Level 2), describing a specific area designed to maintain the pressure differential between the laboratory and its adjacent space. The anteroom should have facilities for separating clean and dirty clothing and a shower may also be necessary.
  2. Anteroom doors may be self-closing and interlocking so that only one door is open at a time. A breakthrough panel may be provided for emergency exit use.
  3. Surfaces of walls, floors and ceilings should be water-resistant and easy to clean. Openings through these surfaces (e.g. for service pipes) should be sealed to facilitate decontamination of the room(s).
  4. The laboratory room must be sealable for decontamination. Air-ducting systems must be constructed to permit gaseous decontamination.
  5. Windows must be closed, sealed and break-resistant.
  6. A handwashing station with hands-free controls should be provided near each exit door.
  7. There must be a controlled ventilation system that maintains a directional airflow into the laboratory room. A visual monitoring device with or without alarm(s) should be installed so that staff can at all times ensure that proper directional airflow into the laboratory room is maintained.
  8. The building ventilation system must be so constructed that air from the containment laboratory, Biosafety Level 3 is not recirculated to other areas within the building. Air may be high-efficiency particulate air (HEPA) filtered, reconditioned and recirculated within that laboratory. When 16exhaust air from the laboratory (other than from biological safety cabinets) is discharged to the outside of the building, it must be dispersed away from occupied buildings and air intakes. Depending on the agents in use, this air may be discharged through HEPA-filters. A heating, ventilation and air-conditioning (HVAC) control system may be installed to prevent sustained positive pressurization of the laboratory. Consideration should be given to the installation of audible or clearly visible alarms to notify personnel of HVAC system failure.
  9. All HEPA-filters must be installed in a manner that permits gaseous decontamination and testing.
  10. Biological safety cabinets should be sited away from walking areas and out of cross-currents from doors and ventilation systems.
  11. The exhaust air from Class I or Class II biological safety cabinets, which will have been passed through HEPA-filters, must be discharged in such a way as to avoid interference with the air balance of the cabinet or the building exhaust system.
  12. An autoclave for the decontamination of contaminated waste material should be available in the containment laboratory. If infectious waste has to be removed from the containment laboratory for decontamination and disposal, it must be transported in sealed, unbreakable and leak-proof containers according to national or international regulations, as appropriate.
  13. Backflow-precaution devices must be fitted to the water supply. Vacuum lines should be protected with liquid disinfectant traps and HEPA-filters, or their equivalent. Alternative vacuum pumps should also be properly protected with traps and filters.
  14. The containment laboratory, Biosafety Level 3 facility design and operational procedures should be documented.
 
Laboratory Equipment
The principles for the selection of laboratory equipment, including biological safety cabinets are the same as for the basic laboratory—Biosafety Level 2. However, at Biosafety Level 3, manipulation of all potentially infectious material must be conducted within a biological safety cabinet or other primary containment device. Consideration should b e given to equipment such as centrifuges, which will need additional containment accessories, for example, safety buckets or containment rotors. Some centrifuges and other equipment, such as cell-sorting instruments for use with infected cells, may need additional local exhaust ventilation with HEPA filtration for efficient containment.
 
Biosafety Level 4 (The Maximum Containment Laboratory)
The maximum containment laboratory, Biosafety Level 4 is designed for work with Risk Group 4 microorganisms. Before such a laboratory is constructed and put into operation, intensive consultations should be held with institutions that have had experience of operating a similar facility. Operational maximum containment laboratories, Biosafety Level 4 should be under the control of national or other appropriate health authorities. The following information is intended only as introductory material.17
 
Code of Practice
The code of practice for Biosafety Level 3 applies except where modified as follows:
  1. The two-person rule should apply, whereby no individual ever works alone. This is particularly important if working in a Biosafety Level 4 suit facility.
  2. A complete change of clothing and shoes is required prior to entering and upon exiting the laboratory.
  3. Personnel must be trained in emergency extraction procedures in the event of personnel injury or illness.
  4. A method of communication for routine and emergency contacts must be established between personnel working within the maximum containment laboratory, Biosafety Level 4 and support personnel outside the laboratory.
 
Laboratory Design and Facilities
The features of a containment laboratory, Biosafety Level 3 also apply to a maximum containment laboratory, Biosafety Level 4 with the addition of the following:
  1. Primary containment: An efficient primary containment system must be in place, consisting of one or a combination of the following:
    • Class III cabinet laboratory: Passage through a minimum of two doors prior to entering the rooms containing the Class III biological safety cabinet(s) (cabinet room) is required. In this laboratory configuration the Class III biological safety cabinet provides the primary containment. A personnel shower with inner and outer changing rooms is necessary. Supplies and materials that are not brought into the cabinet room through the changing area are introduced through a double-door autoclave or fumigation chamber. Once the outer door is securely closed, staff inside the laboratory can open the inner door to retrieve the materials. The doors of the autoclave or fumigation chamber are interlocked in such a way that the outer door cannot open unless the autoclave has been operated through a sterilization cycle or the fumigation chamber has been decontaminated.
    • Suit laboratory: A protective suit laboratory with self-contained breathing apparatus differs significantly in design and facility requirements from a Biosafety Level 4 laboratory with Class III biological safety cabinets. The rooms in the protective suit laboratory are arranged so as to direct personnel through the changing and decontamination areas prior to entering areas where infectious materials are manipulated.
    • A suit decontamination shower must be provided and used by personnel leaving the containment laboratory area. A separate personnel shower with inner and outer changing rooms is also provided. Personnel who enter the suit area are required to don a one-piece, positively pressurized, HEPA-filtered, supplied-air suit. Air to the suit must be provided by a system that has a 100% redundant capability with an independent source of air, for use in the event of an emergency. Entry into the suit laboratory is through an airlock fitted with airtight doors. An appropriate warning system for personnel working in the suit laboratory must be provided for use in the event of mechanical system or air failure.
    18
  2. Controlled access: The maximum containment laboratory, Biosafety Level 4 must be located in a separate building or in a clearly delineated zone within a secure building. Entry and exit of personnel and supplies must be through an airlock or pass-through system. On entering, personnel must put on a complete change of clothing; before leaving, they should shower before putting on their street clothing.
  3. Controlled air system: Negative pressure must be maintained in the facility. Both supply and exhaust air must be HEPA-filtered. There are significant differences in the ventilating systems of the Class III cabinet laboratory and suit laboratory:
    • Class III cabinet laboratory: The supply air to the Class III biological safety cabinet(s) may be drawn from within the room through a HEPA-filter mounted on the cabinet or supplied directly through the supply air system. Exhaust air from the Class III biological safety cabinet must pass through two HEPA-filters prior to release outdoors. The cabinet must be operated at negative pressure to the surrounding laborator y at all times. A dedicated non-recirculating ventilating system for the cabinet laboratory is required.
    • Suit laboratory: Dedicated room air supply and exhaust systems are required.
      The supply and exhaust components of the ventilating system are balanced to provide directional airflow within the suit area from the area of least hazard to the area(s) of greatest potential hazard. Redundant exhaust fans are required to ensure that the facility remains under negative pressure at all times. The differential pressures within the suit laboratory and between the suit laboratory and adjacent areas must be monitored. Airflow in the supply and exhaust components of the ventilating system must be monitored, and an appropriate system of controls must be used to prevent pressurization of the suit laboratory. HEPA-filtered supply air must be provided to the suit area, decontamination shower and decontamination airlocks or chambers. Exhaust air from the suit laboratory must be passed through a series of two HEPA-filters prior to release outdoors. Alternatively, after double HEPA filtration, exhaust air may be recirculated, but only within the suit laboratory. Under no circumstances shall the exhaust air from the Biosafety Level 4 suit laboratory be recirculated to other areas. Extreme caution must be exercised if recirculation of air within the suit laboratory is elected. Consideration must be given to the types of research conducted, equipment, chemicals and other materials used in the suit laboratory, as well as animal species that may be involved in the research.
All HEPA-filters need to be tested and certified annually. The HEPA-filter housings are designed to allow for in situ decontamination of the filter prior to removal. Alternatively, the filter can be removed in a sealed, gas-tight primary container for subsequent decontamination and/or destruction by incineration.
  1. Decontamination of effluents: All effluents from the suit area, decontamination chamber, decontamination shower, or Class III biological safety cabinet must be decontaminated before final discharge. Heat treatment is the preferred method. Effluents may also require correction 19to a neutral pH prior to discharge. Water from the personnel shower and toilet may be discharged directly to the sanitary sewer without treatment.
  2. Sterilization of waste and materials: A double-door, pass-through autoclave must be available in the laboratory area. Other methods of decontamination must be available for equipment and items that cannot withstand steam sterilization.
  3. Airlock entry ports: For specimens, materials and animals must be provided.
  4. Emergency power and dedicated power supply line(s) must be provided.
  5. Containment drain(s) must be installed.
 
PERSONAL PROTECTIVE EQUIPMENT AND CLOTHING
Personal protective equipment and clothing may act as a barrier to minimize the risk of exposure to aerosols, splashes and accidental inoculation. The clothing and equipment selected is dependent on the nature of the work performed. Protective clothing should be worn when working in the laboratory. Before leaving the laboratory, protective clothing should be removed, and hands should be washed.
 
Laboratory Coats, Gowns, Coveralls, Aprons
Laboratory coats should preferably be fully-buttoned. However, long-sleeved, back-opening gowns or coveralls give better protection than laboratory coats and are preferred in microbiology laboratories and when working at the biological safety cabinet. Aprons may be worn over laboratory coats or gowns where necessary to give further protection against spillage of chemicals or biological materials such as blood or culture fluids. Laundering services should be provided at/near the facility. Laboratory coats, gowns, coveralls, or aprons should not be worn outside the laboratory areas (Table 1.2).
 
Goggles, Safety Spectacles, Face Shields
The choice of equipment to protect the eyes and face from splashes and impacting objects will depend on the activity performed. Prescription or plain eye glasses can be manufactured with special frames that allow lenses to be placed in frame from the front, using shatterproof material either curved or fitted with side shields (safety glasses). Safety spectacles do not provide for adequate splash protection even when side shields are worn with them. Goggles for splash and impact protection should be worn over normal prescription eye glasses and contact lenses (which do not provide protection against biological or chemical hazards). Faces shields (visors) are made of shatterproof plastic fit over the face and are held in place by head straps or caps. Goggles, safety spectacles, or face shields should not be worn outside the laboratory areas.
 
Respirators
Respiratory protection may be used when carrying out high hazard procedures (e.g. cleaning up a spill of infectious material). The choice of respirator will depend on the type of hazard(s). Respirators are available with interchangeable filters for protection against gases, vapors, particulates and microorganisms.
20
Table 1.2   Personal protective equipment
Equipment
Hazard corrected
Safety features
Laboratory coats, gowns, coveralls
Contamination of clothing
• Back opening
• Cover street clothing
Plastic aprons
Contamination of clothing
• Waterproof
Footwear
Impact and splash
• Closed-toe
Goggles
Impact and splash
• Impact-resistant lenses (must be optically correct or worn over corrective eye glasses)
• Side shields
Safety spectacles Impact
• Impact-resistant lenses (must be optically correct)
• Side shields
Face shields
Impact and splash
• Shield entire face
• Easily removable in case of accident
Respirators
Inhalation of aerosols
• Designs available include singleuse disposable; full-face or half-face air purifying; full-face or hooded powered air purifying (PAPR); and supplied air respirators
Gloves
Direct contact with microorganisms
• Disposable microbiologically approved latex, microorganisms vinyl or nitrile
• Hand protection
Cuts
• Mesh
 
PROCEDURE OF PUTTING ON PERSONAL PROTECTIVE EQUIPMENT
 
Laboratory coats, gowns, coveralls
zoom view
21
 
Respiratory protection
zoom view
 
Goggles or face shield
zoom view
 
Disposable gloves
zoom view
22
 
PROCEDURE OF REMOVAL OF PERSONAL PROTECTIVE EQUIPMENT
 
Gloves
zoom view
 
Laboratory coats, gowns, coveralls
zoom view
 
Goggles or face shield
zoom view
23
 
Respiratory protection
zoom view
 
LABORATORY ANIMAL FACILITIES
Those who use animals for experimental and diagnostic purposes have a moral obligation to take every care to avoid causing them unnecessary pain or suffering. The animals must be provided with comfortable, hygienic housing and adequate wholesome food and water. At the end of the experiment they must be dealt with in a humane manner.
For security reasons, the animal house should be an independent, detached unit. If it adjoins a laboratory, the design should provide for its isolation from the public parts of the laboratory should such need arise, and for its decontamination and disinfestation.
Animal facilities, like laboratories, may be designated according to a risk assessment and the risk group of the microorganisms under investigation, as Animal Facility Biosafety Level 1, 2, 3 or 4 (Table 1.3).
With respect to agents to be used in the animal laboratory, factors for consideration include:
  1. The normal route of transmission.
  2. The volumes and concentrations to be used.
    Table 1.3   Animal facility containment levels: summary of practices and safety equipment
    Risk group
    Containment level
    Laboratory practices and safety equipment
    1
    ABSL-1
    Limited access, protective clothing and gloves
    2
    ABSL-2
    ABSL-1 practices plus: Hazard warning signs. Class I or II BSCs for activities that produce aerosols Decontamination of waste and cages before washing
    3
    ABSL-3
    ABSL-2 practices plus: Controlled access. BSCs and special protective clothing for all activities
    4
    ABSL-4
    ABSL-3 plus: Strictly limited access. Clothing change before entering. Class III BSCs or positive pressure suits. Shower on exit. Decontamination of all wastes before removal from facility
    ABSL, Animal Facility Biosafety Level; BSCs, Biological Safety Cabinets
    24
  3. The route of inoculation.
  4. Whether and by what route these agents may be excreted.
With respect to animals to be used in the animal laboratory, factors for consideration include:
  1. The nature of the animals, i.e. their aggressiveness and tendency to bite and scratch.
  2. Their natural ecto- and endoparasites.
  3. The zoonotic diseases to which they are susceptible.
  4. The possible dissemination of allergens.
As with laboratories, the requirements for design features, equipment and precautions increase in stringency according to the animal Biosafety Level.
These guidelines are additive, so that each higher level incorporates the standards of the lower levels.
 
Animal Facility—Biosafety Level 1
This is suitable for the maintenance of most stock animals after quarantine (except nonhuman primates, regarding which national authorities should be consulted), and for animals that are deliberately inoculated with agents in Risk Group 1. GMT is required. The animal facility director must establish policies, procedures and protocols for all operations, and for access to the vivarium. An appropriate medical surveillance program for the staff must be instituted. A safety or operations manual must be prepared and adopted.
 
Animal Facility—Biosafety Level 2
This is suitable for work with animals that are deliberately inoculated with micro-organisms in Risk Group 2. The following safety precautions apply:
  1. All the requirements for animal facilities—Biosafety Level 1 must be met.
  2. Biohazard warning signs should be posted on doors and other appropriate places.
  3. The facility must be designed for easy cleaning and housekeeping.
  4. Doors must open inwards and be self-closing.
  5. Heating, ventilation and lighting must be adequate.
  6. If mechanical ventilation is provided, the airflow must be inwards. Exhaust air is discharged to the outside and should not be recirculated to any part of the building.
  7. Access must be restricted to authorized persons.
  8. No animals should be admitted other than those for experimental use.
  9. There should be an arthropod and rodent control program.
  10. Windows, if present, must be secure, resistant to breakage and, if able to be opened, must be fitted with arthropod-proof screens.
  11. After use, work surfaces must be decontaminated with effective disinfectants.
  12. Biological safety cabinets (Classes I or II) or isolator cages with dedicated air supplies and HEPA-filtered exhaust air must be provided for work that may involve the generation of aerosols.
  13. An autoclave must be available on site or in appropriate proximity to the animal facility.
    25
  14. Animal bedding materials must be removed in a manner that minimizes the generation of aerosols and dust.
  15. All waste materials and bedding must be decontaminated before disposal.
  16. Use of sharp instruments should be restricted whenever possible. Sharps should always be collected in puncture-proof/ resistant containers fitted with covers and treated as infectious.
  17. Material for autoclaving or incineration must be transported safely, in closed containers.
  18. Animal cages must be decontaminated after use.
  19. Animal carcasses should be incinerated.
  20. Protective clothing and equipment must be worn in the facility, and removed on leaving.
  21. Hand-washing facilities must be provided. Staff must wash their hands before leaving the animal facility.
  22. All injuries, however minor, must be treated appropriately, reported and recorded.
  23. Eating, drinking, smoking and application of cosmetics must be forbidden in the facility.
  24. All personnel must receive appropriate training.
 
Animal Facility—Biosafety Level 3
This is suitable for work with animals that are deliberately inoculated with agents in Risk Group 3, or when otherwise indicated by a risk-assessment. All systems, practices and procedures need to be reviewed and recertified annually. The following safety precautions apply:
  1. All the requirements for animal facilities—Biosafety Levels 1 and 2 must be met.
  2. Access must be strictly controlled.
  3. The facility must be separated from other laboratory and animal house areas by a room with a double-door entrance forming an anteroom.
  4. Hand-washing facilities must be provided in the anteroom.
  5. Showers should be provided in the anteroom.
  6. There must be mechanical ventilation to ensure a continuous airflow through all the rooms. Exhaust air must pass through HEPA-filters before being discharged to the atmosphere without recirculation. The system must be designed to prevent accidental reverse flow and positive pressurization in any part of the animal house.
  7. An autoclave must be available at a location convenient for the animal house where the biohazard is contained. Infectious waste should be autoclaved before it is moved to other areas of the facility.
  8. An incinerator should be readily available on site or alternative arrangements should be made with the authorities concerned.
  9. Animals infected with Risk Group 3 microorganisms must be housed in cages in isolators or rooms with ventilation exhausts placed behind the cages.
  10. Bedding should be as dust-free as possible.
  11. All protective clothing must be decontaminated before it is laundered.
  12. Windows must be closed and sealed, and resistant to breakage.
  13. Immunization of staff, as appropriate, should be offered.
    26
 
Animal Facility—Biosafety Level 4
Work in this facility will normally be linked with that in the maximum containment laboratory—Biosafety Level 4, and national and local rules and regulations must be harmonized to apply to both. If work is to be done in a suit laboratory, additional practices and procedures must be used over and above those described here:
  1. All the requirements for animal facilities—Biosafety Levels 1, 2 and 3 must be met.
  2. Access must be strictly controlled; only staff designated by the director of the establishment should have authority to enter.
  3. Individuals must not work alone: the two-person rule must apply.
  4. Personnel must have received the highest possible level of training as microbiologists and be familiar with the hazards involved in their work and with the necessary precautions.
  5. Housing areas for animals infected with Risk Group 4 agents must maintain the criteria for containment described and applied for maximum containment laboratories—Biosafety Level 4.
  6. The facility must be entered by an airlock anteroom, the clean side of which must be separated from the restricted side by changing and showering facilities.
  7. Staff must remove street clothing when entering and put on special, protective clothing. After work they must remove the protective clothing for autoclaving, and shower before leaving.
  8. The facility must be ventilated by a HEPA-filtered exhaust system designed to ensure a negative pressure (inward directional airflow).
  9. The ventilation system must be designed to prevent reverse flow and positive-pressurization.
  10. A double-ended autoclave with the clean end in a room outside the containment rooms must be provided for exchange of materials.
  11. A pass-through airlock with the clean end in a room outside the containment rooms must be provided for exchange of non-autoclavable materials.
  12. All manipulations with animals infected with Risk Group 4 agents must take place under maximum containment—Biosafety Level 4 conditions.
  13. All animals must be housed in isolators.
  14. All animal bedding and waste must be autoclaved before removal from the facility.
  15. There must be medical supervision of staff.
 
Invertebrates
As with vertebrates, the animal facility biosafety level will be determined by the risk groups of the agents under investigation or when otherwise indicated by a risk-assessment. The following additional precautions are necessary with certain arthropods, particularly with flying insects:
  1. Separate rooms should be provided for infected and non-infected invertebrates.
  2. The rooms should be capable of being sealed for fumigation.
  3. Insecticide sprays should be readily available.
  4. “Chilling” facilities should be provided to reduce, where necessary, the activity of invertebrates.
    27
  5. Access should be through an anteroom containing insect traps and with arthropod-proof screens on the doors.
  6. All exhaust ventilation ducts and openable windows should be fitted with arthropod-proof screens.
  7. Waste traps on sinks and sluices should not be allowed to dry out.
  8. All waste should be decontaminated by autoclaving, as some invertebrates are not killed by all disinfectants.
  9. A check should be kept on the numbers of larval and adult forms of flying, crawling and jumping arthropods.
  10. Containers for ticks and mites should stand in trays of oil.
  11. Infected or potentially infected flying insects must be contained in double-netted cages.
  12. Infected or potentially infected arthropods must be handled in biological safety cabinets or isolators.
  13. Infected or potentially infected arthropods may be manipulated on cooling trays.
 
LABORATORY EQUIPMENT
 
Biological Safety Cabinets
Biological safety cabinets (BSCs) are designed to protect the operator, the laboratory environment and work materials from exposure to infectious aerosols and splashes that may be generated when manipulating materials containing infectious agents, such as primary cultures, stocks and diagnostic specimens. Aerosol particles are created by any activity that imparts energy into a liquid or semiliquid material, such as shaking, pouring, stirring or dropping liquid onto a surface or into another liquid. Other laboratory activities, such as streaking agar plates, inoculating cell culture flasks with a pipette, using a multi-channel pipette to dispense liquid suspensions of infectious agents into microculture plates, homogenizing and vortexing infectious materials, and centrifugation of infectious liquids, or working with animals, can generate infectious aerosols. Aerosol particles of less than 5 m in diameter and small droplets of 5–100 m in diameter are not visible to the naked eye. The laboratory worker is generally not aware that such particles are being generated and may be inhaled or may cross-contaminate work surface materials. BSCs, when properly used, have been shown to be highly effective in reducing laboratory-acquired infections and cross-contaminations of cultures due to aerosol exposures. BSCs also protect the environment.
Over the years the basic design of BSCs has undergone several modifications. A major change was the addition of a high-efficiency particulate air (HEPA) filter to the exhaust system. The HEPA-filter traps 99.97% of particles of 0.3 in diameter and 99.99% of particles of greater or smaller size. This enables the HEPA-filter to effectively trap all known infectious agents and ensure that only microbe-free exhaust air is discharged from the cabinet. A second design modification was to direct HEPA-filtered air over the work surface, providing protection of work surface materials from contamination. This feature is often referred to as product protection. These basic design concepts have led to the evolution of three classes of BSCs.
28Note: Horizontal and vertical outflow cabinets (“clean-air work stations”) are not biological safety cabinets and should not be used as such.
 
Class I Biological Safety Cabinet
The directional flow of air whisks aerosol particles that may be generated on the work surface away from the laboratory worker and into the exhaust duct. The front opening allows the operator's arms to reach the work surface inside the cabinet while he or she observes the work surface through a glass window. The window can also be fully raised to provide access to the work surface for cleaning or other purposes (Table 1.4).
The air from the cabinet is exhausted through a HEPA-filter: (a) Into the laboratory and then to the outside of the building through the building exhaust; (b) To the outside through the building exhaust; or (c) Directly to the outside. The HEPA-filter may be located in the exhaust plenum of the BSC or in the building exhaust. Some Class I BSCs are equipped with an integral exhaust fan, whereas others rely on the exhaust fan in the building exhaust system.
The Class I BSC is the first recognized BSC and, because of its simple design, is still in wide use throughout the world. It has the advantage of providing personnel and environmental protection and can also be used for work with radionuclides and volatile toxic chemicals. Because unsterilized room air is drawn over the work surface through the front opening, it is not considered to provide consistently reliable product protection.
 
Class II Biological Safety Cabinets
As the use of cell and tissue cultures for the propagation of viruses and other purposes grew, it was no longer considered satisfactory for unsterilized room air to pass over the work surface. The Class II BSC is designed not only to provide personnel protection but also to protect work surface materials from contaminated room air. Class II BSCs, of which there are four types (A1, A2,
Table 1.4   Selection of a biological safety cabinet (BSC), by type of protection needed
Type of protection
BSC selection
Personnel protection, microorganisms in Risk Groups 1–3
Class I, Class II, Class III
Personnel protection, microorganisms in Risk Group 4, glove-box laboratory
Class III
Personnel protection, microorganisms in Risk Group 4, suit laboratory
Class I, Class II
Product protection
Class II, Class III only if laminar flow included
Volatile radionuclide/chemical protection, vented to the outside minute amounts
Class IIB1, Class IIA2
Volatile radionuclide/chemical protection
Class I, Class IIB2, Class III
29B1 and B2), differ from Class I BSCs by allowing only air from a HEPA-filtered (sterile) supply to flow over the work surface. The Class II BSC can be used for working with infectious agents in Risk Groups 2 and 3. Class II BSCs can be used for working with infectious agents in Risk Group 4 when positive-pressure suits are used.
 
Class II Type A1 Biological Safety Cabinet
The Class II type A1 BSC is internal fan draws room air (Supply air) into the cabinet through the front opening and into the front intake grill. The inflow velocity of this air should be at least 0.38 m/s at the face of the front opening. The supply air then passes through a supply HEPA-filter before flowing downwards over the work surface. As the air flows downwards it “splits” about 6–18 cm from the work surface, one half of the downwards flowing air passing through the front exhaust grill, and the other half passing through the rear exhaust grill. Any aerosol particles generated at the work surface are immediately captured in this downward airflow and passed through the front or rear exhaust grills, thereby providing the highest level of product protection. The air is then discharged through the rear plenum into the space between the supply and exhaust filters located at the top of the cabinet. Owing to the relative size of these filters, about 70% of the air recirculates through the supply HEPA-filter back into the work zone; the remaining 30% passes through the exhaust filter into the room or to the outside. Air from the Class IIA1 BSC exhaust can be recirculated to the room or discharged to the outside of the building through a thimble connection to a dedicated duct or through the building exhaust system (Figure 1.2).
Recirculating the exhaust air to the room has the advantage of lowering building fuel costs because heated and/or cooled air is not being passed to the outside environment. A connection to a ducted exhaust system also allows some BSCs to be used for work with volatile radionuclides and volatile toxic chemicals.
zoom view
Figure 1.2: Schematic diagram of a Class I biological safety cabinet. A. Front opening; B. Sash; C. Exhaust HEPA-filter; D. Exhaust plenum
30
 
Class II Type A2 Vented to the Outside, B1 and B2 Biological Safety Cabinets
Class IIA2 vented to the outside, IIB1 and IIB2 BSCs (Figure 1.4) are variations of the type IIA1. Their characteristics, along with those of Class I and Class III BSCs, are indicated (Figure 1.3). Each variation allows the BSC to be used for specialized purpose.
These BSCs differ from one another in several aspects: the air intake velocity through the front opening; the amount of air recirculated over the work surface and exhausted from the cabinet; the exhaust system, which determines whether air from the cabinet is exhausted to the room, or to the outside, through a dedicated exhaust system or through the building exhaust; and the pressure arrangements (whether cabinets have biologically contaminated ducts and plenums under negative pressure, or have biological contaminated ducts and plenums surrounded by negative-pressure ducts and plenums) (Table 1.5).
 
Class III Biological Safety Cabinet
This type provides the highest level of personnel protection and is used for Risk Group 4 agents. All penetrations are sealed “gas tight”. Supply air is HEPA-filtered and exhaust air passes through two HEPA-filters. Airflow is maintained by a dedicated exhaust system exterior to the cabinet, which keeps the cabinet interior under negative pressure (about 124.5 Pa). Access to the work surface is by means of heavy duty rubber gloves, which are attached to ports in the cabinet. The Class III BSC should have an attached pass-through box that can be sterilized and is equipped with a HEPA-filtered exhaust. The Class III cabinet may be connected to a double-door autoclave used to decontaminate all materials entering or exiting the cabinet. Several glove boxes can be joined together to extend the work surface. Class III BSCs are suitable for work in Biosafety Level 3 and 4 laboratories.
zoom view
Figure 1.3: Schematic representation of a Class IIA1 biological safety cabinet. A. Front opening; B. Sash; C. Exhaust HEPA-filter; D. Rear plenum; E. Supply HEPA-filter; F. Blower
31
zoom view
Figure 1.4: Schematic diagram of a Class II B1 biological safety cabinet. A. Front opening; B. Sash; C. Exhaust HEPA-filter; D. Supply HEPA-filter; E. Negative-pressure exhaust plenum; F. Blower; G. HEPA-filter for supply air. Connection of the cabinet exhaust to the building exhaust air system is required
Table 1.5   Differences between Class I, II and III biological safety cabinets (BSCs)
BSC
Face velocity (M/s)
Airflow (%)
Exhaust system
Recirculated
Exhausted
Class Ia
0.36
0
100
Hard duct
Class IIA 1
0.38–0.51
70
30
Exhaust to room or thimble connection
Class IIA 2 vented to the outsidea
0.51
70
30
Exhaust to room or thimble connection
Class IIB 1a
0.51
30
70
Hard duct
Class IIB 2a
0.51
0
100
Hard duct
Class IIIa
NA
0
100
Hard duct
NA, not applicable
a All biologically contaminated ducts are under negative pressure or are surrounded by negative pressure ducts and plenums
 
Biological Safety Cabinet Air Connections
A “thimble” or “canopy hood” is designed for use with Class IIA1 and IIA2 vented to the outside BSCs. The thimble fits over the cabinet exhaust housing, sucking the cabinet exhaust air into the building exhaust ducts. A small opening, usually 2.5 cm in diameter, is maintained between the thimble and the cabinet exhaust housing. This small opening enables room air to be sucked into the building exhaust system as well.
The building exhaust capacity must be sufficient to capture both room air and the cabinet exhaust. The thimble must be removable or be designed to 32allow for operational testing of the cabinet. Generally, the performance of a thimble-connected BSC is not affected much by fluctuations in the airflow of the building Class IIB1 and IIB2 BSCs are hard-ducted, i.e. firmly connected without any openings, to the building exhaust system or, preferably, to a dedicated exhaust duct system. The building exhaust system must be precisely matched to the airflow requirements specified by the manufacturer for both volume and static pressure. Certification of hard-duct connected BSCs is more time-consuming than that for BSCs that recirculate air to the room or which are thimble-connected.
 
Selection of a Biological Safety Cabinet
A BSC should be selected primarily in accordance with the type of protection needed: product protection, personnel protection against Risk Group 1–4 microorganisms, personnel protection against exposure to radionuclides and volatile toxic chemicals, or a combination of these. Table 8 shows which BSCs are recommended for each type of protection. Volatile or toxic chemicals should not be used in BSCs that recirculate exhaust air to the room, i.e. Class I BSCs that are not ducted to building exhaust systems, or Class IIA1 or Class IIA2 cabinets. Class IIB1 BSCs are acceptable for work with minute amounts of volatile chemicals and radionuclides. A Class IIB2 BSC, also called a total exhaust cabinet, is necessary when significant amounts of radionuclides and volatile chemicals are expected to be used (Figure 1.5).
 
Using Biological Safety Cabinets in the Laboratory
 
Location
The velocity of air flowing through the front opening into a BSC is about 0.45 m/s.
zoom view
Figure 1.5: Schematic representation of a Class III biological safety cabinet (glove box). A. Glove ports for arm-length gloves; B. Sash; C. Double-exhaust HEPA-filters; D. Supply HEPA-filter; E. Double-ended autoclave or pass-through box; F. Chemical dunk tank. Connection of the cabinet exhaust to an independent building exhaust airsystem is required
33
At this velocity the integrity of the directional air inflow is fragile and can be easily disrupted by air currents generated by people walking close to the BSC, open windows, air supply registers, and opening and shutting doors. Ideally, BSCs should be situated in a location remote from traffic and potentially disturbing air currents.
Whenever possible a 30 cm clearance should be provided behind and on each side of the cabinet to allow easy access for maintenance. A clearance of 30–35 cm above the cabinet may be required to provide for accurate air velocity measurement across the exhaust filter and for exhaust filter changes.
 
Material Placement
The front intake grill of Class II BSCs must not be blocked with paper, equipment or other items. Materials to be placed inside the cabinet should be surface decontaminated with 70% alcohol. Work may be performed on disinfectant-soaked absorbent towels to capture splatters and splashes. All materials should be placed as far back in the cabinet, towards the rear edge of the work surface, as practical without blocking the rear grill. Aerosol-generating equipment (e.g. mixers, centrifuges, etc.) should be placed towards the rear of the cabinet. Bulky items, such as biohazard bags, discard pipette trays and suction collection flasks should be placed to one side of the interior of the cabinet. Active work should flow from clean to contaminated areas across the work surface.
The autoclavable biohazard collection bag and pipette collection tray should not be placed outside the cabinet. The frequent in-and-out movement needed to use these containers is disruptive to the integrity of the cabinet's air barrier, and can compromise both personnel and product protection.
 
Ultraviolet Lights
Ultraviolet lights are not required in BSCs. If they are used, they must be cleaned weekly to remove any dust and dirt that may block the germicidal effectiveness of the light. Ultraviolet light intensity should be checked when the cabinet is recertified to ensure that light emission is appropriate. Ultraviolet lights must be turned off while the room is occupied, to protect eyes and skin from inadvertent exposure.
 
Spills
A copy of the laboratory's protocol for handling spills should be posted, read and understood by everyone who uses the laboratory. When a spill of biohazardous material occurs within a BSC, clean-up should begin immediately, while the cabinet continues to operate. An effective disinfectant should be used and applied in a manner that minimizes the generation of aerosols. All materials that come into contact with the spilled agent should be disinfected and/or autoclaved.
 
Cleaning and Disinfection
All items within BSCs, including equipment, should be surface-decontaminated and removed from the cabinet when work is completed, since residual culture media may provide an opportunity for microbial growth.
The interior surfaces of BSCs should be decontaminated before and after each use. The work surfaces and interior walls should be wiped with a disinfectant 34that will kill any microorganisms that might be found inside the cabinet. At the end of the work day, the final surface decontamination should include a wipe-down of the work surface, the sides, back and interior of the glass. A solution of bleach or 70% alcohol should be used where effective for target organisms. A second wiping with sterile water is needed when a corrosive disinfectant, such as bleach, is used.
It is recommended that the cabinet is left running. If not, it should be run for 5 minutes in order to purge the atmosphere inside before it is switched off.
 
Decontamination
BSCs must be decontaminated before filter changes and before being moved. The most common decontamination method is by fumigation with formaldehyde gas. BSC decontamination should be performed by a qualified professional.
 
Negative-Pressure Flexible-Film Isolators
The negative-pressure flexible-film isolator is a self-contained primary containment device that provides maximum protection against hazardous biological materials. It may be mounted on a mobile stand. The workspace is totally enclosed in a transparent polyvinylchloride (PVC) envelope suspended from a steel framework. The isolator is maintained at an internal pressure lower than atmospheric pressure. Inlet air is passed through one HEPA-filter and outlet air is passed through two HEPA-filters, thus obviating the need to duct exhaust air outside the building. The isolator may be fitted with an incubator, microscope and other laboratory equipment, such as centrifuges, animal cages, heat blocks, etc. Material is introduced and removed from the isolator through supply and sample ports without compromising microbiological security. Manipulations are performed using gloved sleeves incorporating disposable gloves. A manometer is installed to monitor envelope pressure.
Flexible-film isolators are used to manipulate high-risk organisms (Risk Groups 3 or 4) in field work where it is not feasible or appropriate to install or maintain conventional biological safety cabinets.
 
Pipetting Aids
A pipetting aid must always be used for pipetting procedures. Mouth pipetting must be strictly forbidden. The importance of pipetting aids cannot be overemphasized. The most common hazards associated with pipetting procedures are the result of mouth suction. Oral aspiration and ingestion of hazardous materials have been responsible for many laboratory-associated infections.
Pathogens can also be transferred to the mouth if a contaminated finger is placed on the suction end of a pipette. A lesser known hazard of mouth pipetting is the inhalation of aerosols caused by suction. The cotton plug is not an efficient microbial filter at negative or positive pressure, and particles may be sucked through it. Violent suction may be applied when the plug is tightly packed, resulting in the aspiration of plug, aerosol and even liquid.
The ingestion of pathogens is prevented by the use of pipetting aids. Aerosols can also be generated when a liquid is dropped from a pipette on to a work surface, when cultures are mixed by alternate sucking and blowing, and when the last drop is blown out of a pipette. The inhalation of aerosols unavoidably 35generated during pipetting operations can be prevented by working in a biological safety cabinet. Pipetting aids should be selected with care. Their design and use should not create an additional infectious hazard and they should be easy to sterilize and clean. Plugged (aerosol-resistant) pipette tips should be used when manipulating microorganisms and cell cultures. Pipettes with cracked or chipped suction ends should not be used as they damage the seating seals of pipetting aids and so create a hazard.
 
LABORATORY BIOSECURITY
Today the nation is facing a new challenge in safeguarding the public health from potential domestic or international terrorism involving the use of dangerous biological agents or toxins. Existing standards and practices may require adaptation to ensure protection from such hostile actions.
Laboratory biosecurity refers to institutional and personal security measures designed to protect workers, environment and population against the loss, theft, use and release in the environment of pathogenic biological agents and toxins. Biosecurity management and practices are designed to prevent the spread of disease by minimizing the movement of biologic organisms approaches enable staff to deal with the unpredicted and unfamiliar in the most prudent and safe manner.
A specific laboratory biosecurity protocol must be prepared and implemented for each facility according to the requirements of the facility, the type of laboratory work conducted, and the local conditions. Consequently, laboratory biosecurity activities should be representative of the institution's various needs and should include input from scientific directors, principal investigators, biosafety officers, laboratory scientific staff, maintenance staff, administrators, information technology staff, and law enforcement agencies and security staff if appropriate.
Laboratory biosecurity measures should be based on a comprehensive program of accountability for pathogens and toxins that includes an updated inventory with storage location, identification of personnel with access, description of use, documentation of internal and external transfers within and between facilities, and any inactivation and/or disposal of the materials. Likewise, an institutional laboratory biosecurity protocol should be established for identifying, reporting, investigating and remediating breaches in laboratory biosecurity, including discrepancies in inventory results. The involvement and roles and responsibilities of public health and security authorities in the event of a security infraction must be clearly defined.
Laboratory biosecurity training, distinct from laboratory biosafety training, should be provided to all personnel. Such training should help personnel understand the need for protection of such materials and the rationale for the specific biosecurity measures, and should include a review of relevant national standards and institution specific procedures.
Procedures describing the security roles and responsibilities of personnel in the event of a security infraction should also be presented during training. The professional and ethical suitability for working with dangerous pathogens of all personnel who have regular authorized access to sensitive materials is also central to effective laboratory biosecurity activities.
36
In summary, security precautions should become a routine part of laboratory work, just as have aseptic techniques and other safe microbiological practices. Laboratory biosecurity measures should not hinder the efficient sharing of reference materials, clinical and epidemiological specimens and related information necessary for clinical or public health investigations. Competent security management should not unduly interfere with the day-to-day activities of scientific personnel or be an impediment to conducting research. Legitimate access to important research and clinical materials must be preserved. Assessment of the suitability of personnel, security-specific training and rigorous adherence to pathogen protection procedures are reasonable means of enhancing laboratory biosecurity. All such efforts must be established and maintained through regular risk and threat assessments, and regular review and updating of procedures.
 
LABORATORY TECHNIQUES
Human error, poor laboratory techniques and misuse of equipment cause the majority of laboratory injuries and work-related infections. This section provides a compendium of technical methods that are designed to avoid or minimize the most commonly reported problems of this nature.
 
Safe Handling of Specimens in the Laboratory
Improper collection, transport and handling of specimens in the laboratory carry a risk of infection to the personnel involved.
 
Specimen Containers
Specimen containers may be of glass or preferably plastic. They should be robust and should not leak when the cap or stopper is correctly applied. No material should remain on the outside of the container. Containers should be correctly labeled to facilitate identification. Specimen request or specification forms should not be wrapped around the containers but placed in separate, preferably waterproof envelopes.
 
Transport of Specimens within the Facility
To avoid accidental leakage or spillage, secondary containers, such as boxes, should be used, fitted with racks so that the specimen containers remain upright. The secondary containers may be of metal or plastic, should be autoclavable or resistant to the action of chemical disinfectants, and the seal should preferably have a gasket. They should be regularly decontaminated.
 
Receipt of Specimens
Laboratories that receive large numbers of specimens should designate a particular room or area for this purpose.
 
Opening Packages
Personnel who receive and unpack specimens should be aware of the potential health hazards involved, and should be trained to adopt standard precautions, 37particularly when dealing with broken or leaking containers. Primary specimen containers should be opened in a biological safety cabinet. Disinfectants should be available.
 
Use of Pipettes and Pipetting Aids
  1. A pipetting aid must always be used. Pipetting by mouth must be prohibited.
  2. All pipettes should have cotton plugs to reduce contamination of pipetting devices.
  3. Air should never be blown through liquid containing infectious agents.
  4. Infectious materials should not be mixed by alternate suction and expulsion through a pipette.
  5. Liquids should not be forcibly expelled from pipettes.
  6. Mark-to-mark pipettes are preferable to other types as they do not require expulsion of the last drop.
  7. Contaminated pipettes should be completely submerged in a suitable disinfectant contained in an unbreakable container. They should be left in the disinfectant for the appropriate length of time before disposal.
  8. A discard container for pipettes should be placed within the biological safety cabinet, not outside it.
  9. Syringes fitted with hypodermic needles must not be used for pipetting.
  10. Devices for opening septum-capped bottles that allow pipettes to be used and avoid the use of hypodermic needles and syringes should be used.
  11. To avoid dispersion of infectious material dropped from a pipette, an absorbent material should be placed on the working surface; this should be disposed of as infectious waste after use.
 
Avoiding the Dispersal of Infectious Materials
  1. In order to avoid the premature shedding of their loads, microbiological transfer loops should have a diameter of 2–3 mm and be completely closed. The shanks should be not more than 6 cm in length to minimize vibration.
  2. The risk of spatter of infectious material in an open Bunsen burner flame should be avoided by using an enclosed electric micro-incinerator to sterilize transfer loops. Disposable transfer loops, which do not need to be re-sterilized, are preferable.
  3. Care should be taken when drying sputum samples, to avoid creating aerosols.
  4. Discarded specimens and cultures for autoclaving and/or disposal should be placed in leak proof containers, e.g. laboratory discard bags. Tops should be secured (e.g. with autoclave tape) prior to disposal into waste containers.
  5. Working areas must be decontaminated with a suitable disinfectant at the end of each work period.
 
Use of Biological Safety Cabinets
  1. The use and limitations of biological safety cabinets should be explained to all potential users, with reference to national standards and relevant literature. Written protocols or safety or operations manuals should be issued to staff. In particular, it must be made clear that the cabinet will not protect the operator from spillage, breakage or poor technique.
    38
  2. The cabinet must not be used unless it is working properly.
  3. The glass viewing panel must not be opened when the cabinet is in use.
  4. Apparatus and materials in the cabinet must be kept to a minimum. Air circulation at the rear plenum must not be blocked.
  5. Bunsen burners must not be used in the cabinet. The heat produced will distort the airflow and may damage the filters. An electric micro-incinerator is permissible but sterile disposable transfer loops are better.
  6. All work must be carried out in the middle or rear part of the working surface and be visible through the viewing panel.
  7. Traffic behind the operator should be minimized.
  8. The operator should not disturb the airflow by repeated removal and reintroduction of his or her arms.
  9. Air grills must not be blocked with notes, pipettes or other materials, as this will disrupt the airflow causing potential contamination of the material and exposure of the operator.
  10. The surface of the biological safety cabinet should be wiped using an appropriate disinfectant after work is completed and at the end of the day.
  11. The cabinet fan should be run for at least 5 minutes before beginning work and after completion of work in the cabinet.
  12. Paperwork should never be placed inside biological safety cabinets.
 
Avoiding Ingestion of Infectious Materials and Contact with Skin and Eyes
  1. Disposable gloves should be worn. Laboratory workers should avoid touching their mouth, eyes and face.
  2. Food and drink must not be consumed or stored in the laboratory.
  3. No articles should be placed in the mouth; pens, pencils, chewing gum in the laboratory.
  4. Cosmetics should not be applied in the laboratory.
  5. The face, eyes and mouth should be shielded or otherwise protected during any operation that may result in the splashing of potentially infectious materials.
 
Avoiding Injection of Infectious Materials
  1. Accidental inoculation resulting from injury with broken or chipped glassware can be avoided through careful practices and procedures. Glassware should be replaced with plasticware whenever possible.
  2. Accidental injection may result from sharps injuries e.g. with hypodermic needles (needle-sticks), glass Pasteur pipettes, or broken glass.
  3. Needle-stick injuries can be reduced by: (a) Minimizing the use of syringes and needles (e.g. simple devices are available for opening septum stoppered bottles so that pipettes can be used instead of syringes and needles; or (b) Using engineered sharp safety devices when syringes and needles are necessary.
  4. Needles should never be recapped. Disposable articles should be discarded into puncture-proof/resistant containers fitted with covers.
  5. Plastic Pasteur pipettes should replace those made of glass.39
 
Separation of Serum
  1. Only properly trained staff should be employed for this work.
  2. Gloves and eye and mucous membrane protection should be worn.
  3. Splashes and aerosols can only be avoided or minimized by good laboratory technique. Blood and serum should be pipetted carefully, not poured. Pipetting by mouth must be forbidden.
  4. After use, pipettes should be completely submerged in suitable disinfectant. They should remain in the disinfectant for the appropriate time before disposal or washing and sterilization for reuse.
  5. Discarded specimen tubes containing blood clots, etc. (with caps replaced) should be placed in suitable leak proof containers for autoclaving and/or incineration.
  6. Suitable disinfectants should be available for clean-up of splashes and spillages.
 
Use of Centrifuges
  1. Satisfactory mechanical performance is a prerequisite of microbiological safety in the use of laboratory centrifuges.
  2. Centrifuges should be operated according to the manufacturer's instructions.
  3. Centrifuges should be placed at such a level that workers can see into the bowl to place buckets correctly.
  4. Centrifuge tubes and specimen containers for use in the centrifuge should be made of thick-walled glass or preferably of plastic and should be inspected for defects before use.
  5. Tubes and specimen containers should always be securely capped.
  6. The buckets must be loaded, equilibrated, sealed and opened in a biological safety cabinet.
  7. Buckets should be paired by weight and, with tubes in place, correctly balanced.
  8. The amount of space that should be left between the level of the fluid and the rim of the centrifuge tube should be given in manufacturer's instructions.
  9. Distilled water or alcohol (propanol, 70%) should be used for balancing empty buckets. Saline or hypochlorite solutions should not be used as they corrode metals.
  10. Sealable centrifuge buckets (safety cups) must be used for microorganisms in Risk Groups 3 and 4.
  11. When using angle-head centrifuge rotors, care must be taken to ensure that the tube is not overloaded as it might leak.
  12. The interior of the centrifuge bowl should be inspected daily for staining or soiling at the level of the rotor. If staining or soiling are evident then the centrifugation protocols should be re-evaluated.
  13. Centrifuge rotors and buckets should be inspected daily for signs of corrosion and for hair-line cracks.
  14. Buckets, rotors and centrifuge bowls should be decontaminated after each use.
    40
  15. After use, buckets should be stored in an inverted position to drain the balancing fluid.
  16. Infectious airborne particles may be ejected when centrifuges are used. These particles travel at speeds too high to be retained by the cabinet airflow if the centrifuge is placed in a traditional open-fronted Class I or Class II biological safety cabinet. Enclosing centrifuges in Class III safety cabinets prevents emitted aerosols from dispersing widely.
However, good centrifuge technique and securely capped tubes offer adequate protection against infectious aerosols and dispersed particles.
 
Use of Homogenizers, Shakers, Blenders and Sonicators
  1. Domestic (kitchen) homogenizers should not be used in laboratories as they may leak or release aerosols. Laboratory blenders and stomachers are safer.
  2. Caps and cups or bottles should be in good condition and free from flaws or distortion. Caps should be well-fitting and gaskets should be in good condition.
  3. Pressure builds up in the vessel during the operation of homogenizers, shakers and sonicators. Aerosols containing infectious materials may escape from between the cap and the vessel. Plastic, in particular, polytetrafluoroethylene (PTFE) vessels are recommended because glass may break, releasing infectious material and possibly wounding the operator.
  4. When in use, homogenizers, shakers and sonicators should be covered by a strong transparent plastic casing. This should be disinfected after use. Where possible, these machines should be operated, under their plastic covers, in a biological safety cabinet.
  5. At the end of the operation the containers should be opened in a biological safety cabinet.
  6. Hearing protection should be provided for people using sonicators.
 
Use of Tissue Grinders
  1. Glass grinders should be held in absorbent material in a gloved hand. Plastic (PTFE) grinders are safer.
  2. Tissue grinders should be operated and opened in a biological safety cabinet.
 
Care and Use of Refrigerators and Freezers
  1. Refrigerators, deep-freezers and solid carbon dioxide (dry-ice) chests should be defrosted and cleaned periodically, and any ampoules, tubes, etc. that have broken during storage removed. Face protection and heavy duty rubber gloves should be worn during cleaning. After cleaning, the inner surfaces of the cabinet should be disinfected.
  2. All containers stored in refrigerators, etc. should be clearly labeled with the scientific name of the contents, the date stored and the name of the individual who stored them. Unlabeled and obsolete materials should be autoclaved and discarded.
  3. An inventory must be maintained of the freezer's contents.
  4. Flammable solutions must not be stored in a refrigerator unless it is explosion-proof. Notices to this effect should be placed on refrigerator doors.
    41
 
Opening of Ampoules Containing Lyophilized Infectious Materials
Care should be taken when ampoules of freeze-dried materials are opened, as the contents may be under reduced pressure and the sudden inrush of air may disperse some of the materials into the atmosphere. Ampoules should always be opened in a biological safety cabinet.
 
Ampoules Opening Procedure
  1. First decontaminate the outer surface of the ampoule.
  2. Make a file mark on the tube near to the middle of the cotton or cellulose plug, if present.
  3. Hold the ampoule in alcohol-soaked cotton to protect hands before breaking it at a file scratch.
  4. Remove the top gently and treat as contaminated material.
  5. If the plug is still above the contents of the ampoule, remove it with sterile forceps.
  6. Add liquid for resuspension slowly to the ampoule to avoid frothing.
 
Storage of Ampoules Containing Infectious Materials
Ampoules containing infectious materials should never be immersed in liquid nitrogen because cracked or imperfectly sealed ampoules may break or explode on removal. If very low temperatures are required, ampoules should be stored only in the gaseous phase above the liquid nitrogen. Otherwise, infectious materials should be stored in mechanical deep-freeze cabinets or on dry ice. Laboratory workers should wear eye and hand protection when removing ampoules from cold storage. The outer surfaces of ampoules stored in these ways should be disinfected when the ampoules are removed from storage.
Standard precautions with blood, another body fluids, tissues and excreta: Standard precautions are designed to reduce the risk of transmission of microorganisms from both recognized and unrecognized sources of infection.
 
Collection, Labeling and Transport of Specimens
  1. Standard precautions should always be followed; gloves should be worn for all procedures.
  2. Blood should be collected from patients and animals by trained staff.
  3. For phlebotomies, conventional needle and syringe systems should be replaced by single-use safety vacuum devices that allow the collection of blood directly into stoppered transport and/or culture tubes, automatically disabling the needle after use.
  4. The tubes should be placed in adequate containers for transport to the laboratory and within the laboratory facility (see section on Transport of specimens within the facility in this chapter). Request forms should be placed in separate waterproof bags or envelopes.
  5. Reception staff should not open these bags.
 
Opening Specimen Tubes and Sampling Contents
  1. Specimen tubes should be opened in a biological safety cabinet.
    42
  2. Gloves must be worn. Eye and mucous membrane protection is also recommended (goggles or face shields).
  3. Protective clothing should be supplemented with a plastic apron.
  4. The stopper should be grasped through a piece of paper or gauze to prevent splashing.
 
Films and Smears for Microscopy
Fixing and staining of blood, sputum and fecal samples for microscopy do not necessarily kill all organisms or viruses on the smears. These items should be handled with forceps, stored appropriately, and decontaminated and/or autoclaved before disposal.
 
Automated Equipment (Sonicators, Vortex Mixers)
  1. Equipment should be of the closed type to avoid dispersion of droplets and aerosols.
  2. Effluents should be collected in closed vessels for further autoclaving and/or disposal.
  3. Equipment should be disinfected at the end of each session, following manufacturers' instructions.
 
Tissues
  1. Formalin fixatives should be used.
  2. Frozen sectioning should be avoided. When necessary, the cryostat should be shielded and the operator should wear a safety face shield. For decontamination, the temperature of the instrument should be raised to at least 20°C.
 
Decontamination
Hypochlorites and high-level disinfectants are recommended for decontamin­ation. Freshly prepared hypochlorite solutions should contain available chlorine at 1 g/L for general use and 5 g/L for blood spillages. Glutaraldehyde may be used for decontaminating surfaces.
 
Precautions with Materials that may Contain Prions
Prions (also referred to as “slow viruses”) are associated with the transmissible spongiform encephalopathies (TSEs), notably Creutzfeldt-Jakob disease (CJD; including the new variant form), Gerstmann-Sträussler-Scheinker syndrome, fatal familial insomnia and kuru in humans; scrapie in sheep and goats; bovine spongiform encephalopathy (BSE) in cattle; and other transmissible encephalopathies of deer, elk and mink. Although CJD has been transmitted to humans, there appear to be no proven cases of laboratory-associated infections with any of these agents. Nevertheless, it is prudent to observe certain precautions in the handling of material from infected or potentially infected humans and animals.
The selection of a biosafety level for work with materials associated with TSEs will depend on the nature of the agent and the samples to be studied, and should be undertaken in consultation with national authorities.
43
The highest concentrations of prions are found in central nervous system tissue. Animal studies suggest that it is likely that high concentrations of prions are also found in the spleen, thymus, lymph nodes and lung. Recent studies indicate that prions in lingual and skeletal muscle tissue may also present a potential infection risk. As complete inactivation of prions is difficult to achieve, it is important to stress the use of disposable instruments whenever possible, and to use a disposable protective covering for the work surface of the biological safety cabinet.
The main precaution to be taken is to avoid ingestion of contaminated materials or puncture of the laboratory worker's skin. The following additional precautions should be taken, as the agents are not killed by the normal processes of laboratory disinfection and sterilization.
  1. The use of dedicated equipment, i.e. equipment not shared with other laboratories, is highly recommended.
  2. Disposable laboratory protective clothing (gowns and aprons) and gloves must be worn (steel mesh gloves between rubber gloves for pathologists).
  3. Use of disposable plastic ware, which can be treated and discarded as dry waste, is highly recommended.
  4. Tissue processors should not be used because of the problems of disinfection. Jars and beakers (plastic) should be used instead.
  5. All manipulations must be conducted in biological safety cabinets.
  6. Great care should be exercised to avoid aerosol production, ingestion, and cuts and punctures of the skin.
  7. Formalin-fixed tissues should be regarded as still infectious, even after prolonged exposure to formalin.
  8. Histological samples containing prions are substantially inactivated after exposure to 96% formic acid for 1 hour
  9. Bench waste, including disposable gloves, gowns and aprons, should be autoclaved using a porous load steam sterilizer at 134–137°C for a single cycle of 18 minutes or six successive cycles of 3 minutes each, followed by incineration.
  10. Non-disposable instruments, including steel mesh gloves, must be collected for decontamination.
  11. Infectious liquid waste contaminated with prions should be treated with sodium hypochlorite containing available chlorine at 20 g/L (2%) (final concentration) for 1 hour.
  12. Paraformaldehyde vaporization procedures do not diminish prion titers and prions are resistant to ultraviolet irradiation. However, the cabinets must continue to be decontaminated by standard methods (i.e. formaldehyde gas) to inactivate other agents that may be present.
  13. Prion-contaminated biological safety cabinets and other surfaces can be decontaminated with sodium hypochlorite containing available chlorine at 20 g/L (2%) for 1 hour.
  14. High-efficiency particulate air (HEPA) filters should be incinerated at a Minimum temperature of 1000°C after removal.
Recommended additional steps prior to incineration include:
  • Spraying of the exposed face of the filter with lacquer hairspray prior to removal
    44
  • “Bagging” of filters during removal.
  • Removal of the HEPA-filters from the working chamber so that the inaccessible plenum of the cabinet is not contaminated.
  1. Instruments should be soaked in sodium hypochlorite containing available chlorine at 20 g/L (2%) for 1 hour and then rinsed well in water before autoclaving.
  2. Instruments that cannot be autoclaved can be cleaned by repeated wetting with sodium hypochlorite containing available chlorine at 20 g/L (2%) over a 1-hour period. Appropriate washing to remove residual sodium hypochlorite is required.
 
PACKING AND TRANSPORTATION OF INFECTIOUS SUBSTANCES
Transport of infectious and potentially infectious materials is subject to strict national and international regulations. These regulations describe the proper use of packaging materials, as well as other shipping requirements. Laboratory personnel must ship infectious substances according to applicable transport regulations. Compliance with the rules will:
  1. Reduce the likelihood that packages will be damaged and leak, and thereby,
  2. Reduce the exposures resulting in possible infections
  3. Improve the efficiency of package delivery. The International Air Transport Association (IATA) issues Infectious Substances Shipping Guidelines every year. IATA guidelines must follow ICAO's Technical Instructions as a minimal standard, but may impose additional restrictions. IATA guidelines must be followed if a shipment is carried by members of IATA.
Since the United Nations Model Regulations on the Transport of Dangerous Goods is a dynamic set of recommendations subject to biennial amendments, the reader is referred to the latest issuances of national and international modal regulations for applicable regulatory texts.
 
Infectious Substances
Defined as substances which are known or are reasonably expected to contain pathogens. Pathogens are defined as microorganisms (including bacteria, viruses, rickettsiae, parasites, fungi) and other agents such as prions which can cause disease in humans or animals (Table 1.6 and Figure 1.7).
  • Category A (UN 2814 and UN 2900): An infectious substance which is transported in a form that, when exposure to it occurs, is capable of causing permanent disability, life-threatening or fatal disease in otherwise healthy humans or animals.
  • Category B (UN 3373): An infectious substance that does not meet the criteria for Category A (Figure 1.8 for packaging such specimens).
  • Exemptions: The following are exempt from these regulations:
    • Substances containing microorganisms which are non-pathogenic to humans or animals e.g. Hazard Group 1 agents.45
      Table 1.6   Category A infectious substances
      UN number and Proper shipping name
      Microorganism
      UN 2814 infectious substances affecting humans
      Bacillus anthracis (cultures only)
      Brucella abortus (cultures only), Brucella melitensis (cultures only)
      Brucella suis (cultures only), Burkholderia mallei-Pseudomonas mallei—Glanders (cultures only)
      Burkholderia pseudomalleii – Pseudomonas pseudomalleii (cultures only)
      Chlamydia psittaci—avian strains (cultures only)
      Clostridium botulinum (cultures only)
      Coccidioides immitis (cultures only)
      Coxiella burnetii (cultures only)
      Crimean-Congo hemorrhagic fever virus
      Dengue virus (cultures only)
      Eastern equine encephalitis virus (cultures only)
      Escherichia coli, verotoxigenic (cultures only)
      Ebola virus
      Flexal virus
      Francisella tularensis (cultures only)
      Guanarito virus
      Hantaan virus
      Hantaviruses causing hemorrhagic fever with renal syndrome ⁠
      Hendra virus
      Hepatitis B virus (cultures only)
      Herpes B virus (cultures only)
      Human immunodeficiency virus (cultures only)
      Highly pathogenic avian influenza virus (cultures only)
      Japanese encephalitis virus (cultures only)
      Junin virus
      Kyasanur forest disease virus
      Lassa virus
      Machupo virus
      Marburg virus
      Monkeypox virus
      Mycobacterium tuberculosis (cultures only)
      Nipah virus
      Omsk hemorrhagic fever virus
      Poliovirus (cultures only)
      Rabies virus (cultures only) ⁠
      Rickettsia prowazekii (cultures only)
      Rickettsia rickettsii (cultures only)
      Rift Valley fever virus (cultures only) ⁠
      Russian spring-summer encephalitis virus (cultures only)
      Sabia virus
      Shigella dysenteriae type 1 (cultures only)
      Tick-borne encephalitis virus (cultures only)
      Variola virus
      Venezuelan equine encephalitis virus
      West Nile virus (cultures only)
      Yellow fever virus (cultures only)
      Yersinia pestis (cultures only)
      Cont…
      46
      Cont…
      UN 2900 infectious substances affecting animals only
      African swine fever virus (cultures only)
      Avian paramyxovirus Type 1-Velogenic Newcastle disease virus (cultures only) ⁠
      Classical swine fever virus (cultures only) ⁠
      Foot and mouth disease virus (cultures only) ⁠
      Lumpy skin disease virus (cultures only) ⁠
      Mycoplasma mycoides-Contagious bovine pleuropneumonia (cultures only) ⁠
      Peste des petits ruminants virus (cultures only) ⁠
      Rinderpest virus (cultures only) ⁠
      Sheep-pox virus (cultures only) ⁠
      Goat-pox virus (cultures only) ⁠
      Swine vesicular disease virus (cultures only) ⁠
      Vesicular stomatitis virus (cultures only) ⁠
    • Substances transported in a form whereby any pathogens present have been neutralized or inactivated such that they no longer pose a health risk e.g. substances fixed in formaldehyde.
    • Environmental samples (including food and water) which are not considered to pose a significant risk of infection.
 
Marking and Labeling
The package must include the following:
  • The proper shipping name, e.g. ‘Infectious substance, affecting humans'
  • Consignor and consignee addresses
  • UN specification markings
  • UN ID number and proper shipping name of contents
  • The actual name of the infectious agent in brackets after the proper shipping name (no longer required for air transport), the name and telephone number (24 hours) of the person responsible for the shipment.
  • The Class 6.2 (infectious substance) hazard warning diamond.
  • The appropriate UN number (For example, ‘Infectious substances, affecting humans' this would be UN 2814)
  • The appropriate warning label (Figure 1.6).
 
The Triple Packaging System
Transport of infectious substances requires a basic triple packaging system. It consists of three layers as follows:
  • Primary receptacle: A primary watertight leak-proof receptacle containing the specimen. The receptacle is packaged with enough absorbent material to absorb all fluid in case of breakage (Figure 1.7).
  • Secondary packaging: A second durable, watertight, leak-proof packaging to enclose and protect the primary receptacle(s) (Figure 1.8). Several cushioned primary receptacles may be placed in one secondary packaging, but sufficient additional absorbent material shall be used to absorb all fluid in case of breakage.
    47
    zoom view
    Figure 1.6: Packaging warning signs
  • Outer packaging: Secondary packaging is placed in outer shipping packaging with suitable cushioning material. Outer packaging protects contents from outside influences, such as physical damage, while in transit.
Each completed package is normally required to be marked, labeled and accompanied with appropriate shipping which is available from the Department for Transport.
 
Spill Clean-up Procedure
In the event of a spill of infectious or potentially infectious material, the following spill clean-up procedure should be used.
  1. Wear gloves and protective clothing, including face and eye protection if indicated.
  2. Cover the spill with cloth or paper towels to contain it.
  3. Pour an appropriate disinfectant over the paper towels and the immediately surrounding area (generally, 5% bleach solutions are appropriate; but for spills on aircraft, quaternary ammonium disinfectants should be used).
    48
    zoom view
    Figure 1.7: Packing and labeling of category A infectious substances
    zoom view
    Figure 1.8: Packing and labeling of category B infectious substances
  4. Apply disinfectant concentrically beginning at the outer margin of the spill area, working toward the center.
  5. After the appropriate amount of time (e.g. 30 minutes), clear away the materials. If there is broken glass or other sharps involved, use a dustpan or a piece of stiff cardboard to collect the material and deposit it into a puncture-resistant container for disposal.
  6. Clean and disinfect the area of the spillage (if necessary, repeat steps 2–5).
  7. Dispose of contaminated materials into a leak proof, puncture resistant waste disposal container.
    49
    zoom view
    Figure 1.9: Shipper's declaration for dangerous goods example
  8. After successful disinfection, inform the competent authority that the site has now been decontaminated.
Figure 1.9 shows Shippers declaration format.
 
BIOSAFETY AND BIOTECHNOLOGY
Recombinant DNA technology involves combining genetic material from different sources thereby creating genetically modified organisms (GMOs) that may have never existed in nature before. Initially, there was concern among molecular biologists that such organisms might have unpredictable and 50undesirable properties that could represent a biohazard if they escaped from the laboratory.
Recombinant DNA technology or genetic engineering was first used to clone DNA segments in bacterial hosts in order to overexpress specific gene products for further studies. Recombinant DNA molecules have also been used to create GMOs such as transgenic and “knock-out” animals and transgenic plants.
Recombinant DNA technology has already had an enormous impact on biology and medicine, and will probably have an even greater influence now that the nucleotide sequence of the entire human genome is available. Tens of thousands of genes of yet unknown functions will be studied using recombinant DNA technology. Genetherapy may become a routine treatment for certain diseases, and new vectors for gene transfer are likely to be devised using genetic engineering techniques. Also, transgenic plants produced by recombinant DNA technology may play an increasingly important role in modern agriculture.
Experiments involving the construction or use of GMOs should be conducted after performing a biosafety risk-assessment.
The pathogenic properties and any potential hazards associated with such organisms may be novel and not well-characterized. The properties of the donor organism, the nature of the DNA sequences that will be transferred, the properties of the recipient organism, and the properties of the environment should be evaluated. These factors should help determine the biosafety level that is required for the safe handling of the resulting GMO, and identify the biological and physical containment systems that should be used.
 
Biosafety Considerations for Biological Expression Systems
Biological expression systems consist of vectors and host cells. A number of criteria must be satisfied to make them effective and safe to use. An example of such a biological expression system is plasmid pUC18. Frequently used as a cloning vector in combination with Escherichia coli K12 cells, the pUC18 plasmid has been entirely sequenced. All genes required for expression in other bacteria have been deleted from its precursor plasmid pBR322. E. coli K12 is a non-pathogenic strain that cannot permanently colonize the gut of healthy humans or animals. Routine genetic engineering experiments can safely be performed in E. coli K12/pUC18 at Biosafety Level 1 provided the inserted foreign DNA expression products do not require higher biosafety levels.
 
Biosafety Considerations for Expression Vectors
Higher biosafety levels may be required when:
  1. The expression of DNA sequences derived from pathogenic organisms may increase the virulence of the GMO.
  2. Inserted DNA sequences are not well-characterized, e.g. during preparation of genomic DNA libraries from pathogenic microorganisms.
  3. Gene products have potential pharmacological activity.
  4. Gene products code for toxins.
 
Viral Vectors for Gene Transfer
Viral vectors, e.g. adenovirus vectors, are used for the transfer of genes to other cells. Such vectors lack certain virus replication genes and are 51propagated in cell lines that complement the defect. Stocks of such vectors may be contaminated with replication-competent viruses, generated by rare spontaneous recombination events in the propagating cell lines, or may derive from insufficient purification. These vectors should be handled at the same biosafety level as the parent adenovirus from which they are derived.
 
Transgenic and “Knock-Out” Animals
Animals carrying foreign genetic material (transgenic animals) should be handled in containment levels appropriate to the characteristics of the products of the foreign genes. Animals with targeted deletions of specific genes (“knock-out”animals) do not generally present particular biological hazards.
Examples of transgenic animals include animals expressing receptors for viruses normally unable to infect that species. If such animals escaped from the laboratory and transmitted the transgene to the wild animal population, an animal reservoir for that particular virus could theoretically be generated.
This possibility has been discussed for poliovirus and is particularly relevant in the context of poliomyelitis eradication. Transgenic mice expressing the human poliovirus receptor generated in different laboratories were susceptible to poliovirus infection by various inoculation routes and the resulting disease was clinically and histopathologically similar to human poliomyelitis. However, the mouse model differs from humans in that alimentary tract replication of orally administered poliovirus is either inefficient or does not occur. It is, therefore, very unlikely that escape of such transgenic mice to the wild would result in the establishment of a new animal reservoir for poliovirus. Nevertheless, this example indicates that, for each new line of transgenic animal, detailed studies should be conducted to determine the routes by which the animals can be infected, the inoculum size required for infection, and the extent of virus shedding by the infected animals. In addition, all measures should be taken to assure strict containment of receptor transgenic mice.
 
Transgenic Plants
Transgenic plants expressing genes that confer tolerance to herbicides or resistance to insects are currently a matter of considerable controversy in many parts of the world. The discussions focus on the food-safety of such plants, and on the long-term ecological consequences of their cultivation. Transgenic plants expressing genes of animal or human origin are used to develop medicinal and nutritional products. A risk-assessment should determine the appropriate biosafety level for the production of these plants.
 
Risk-Assessments for Genetically Modified Organisms
Risk-assessments for work with GMOs should consider the characteristics of donor and recipient/host organisms. Examples of characteristics for consideration include the following:
 
Hazards Arising Directly from the Inserted Gene (Donor Organism)
Assessment is necessary in situations where the product of the inserted gene has known biologically or pharmacologically active properties that may give rise to harm, for example:52
  1. Toxins
  2. Cytokines
  3. Hormones
  4. Gene expression regulators
  5. Virulence factors or enhancers
  6. Oncogenic gene sequences
  7. Antibiotic resistance
  8. Allergens.
The consideration of such cases should include an estimation of the level of expression required to achieve biological or pharmacological activity.
 
Hazards Associated with the Recipient/Host
  1. Susceptibility of the host.
  2. Pathogenicity of the host strain, including virulence, infectivity and toxin production.
  3. Modification of the host range.
  4. Recipient immune status.
  5. Consequences of exposure.
 
Hazards Arising from the Alteration of Existing Pathogenic Traits
Many modifications do not involve genes whose products are inherently harmful, but adverse effects may arise as the result of alteration of existing non-pathogenic or pathogenic traits. Modification of normal genes may alter pathogenicity. In an attempt to identify these potential hazards, the following points may be considered (the list is not exhaustive).
  1. Is there an increase in infectivity or pathogenicity?
  2. Could any disabling mutation within the recipient be overcome as a result of the insertion of the foreign gene?
  3. Does the foreign gene encode a pathogenicity determinant from another organism?
  4. If the foreign DNA does include a pathogenicity determinant, is it foreseeable that this gene could contribute to the pathogenicity of the GMO?
  5. Is treatment available?
  6. Will the susceptibility of the GMO to antibiotics or other forms of therapy be affected as a consequence of the genetic modification?
  7. Is eradication of the GMO achievable?
 
Further Considerations
The use of whole animals or plants for experimental purposes also requires careful consideration.
Investigators must comply with the regulations, restrictions and requirements for the conduct of work with GMOs in host countries and institutions. Countries may have national authorities that establish guidelines for work with GMOs, and may help scientists classify their work at the appropriate biosafety level. In some cases classification may differ between countries, or countries may decide to classify work at a lower or higher level when new information on a particular vector/ host system becomes available.
53
Risk-assessment is a dynamic process that takes into account new developments and the progress of science. The performance of appropriate risk-assessments will assure that the benefits of recombinant DNA technology remain available to human kind in the years to come.
 
DISINFECTION AND STERILIZATION
A basic knowledge of disinfection and sterilization is crucial for biosafety in the laboratory. Since heavily soiled items cannot promptly be disinfected or sterilized, it is equally important to understand the fundamentals of cleaning prior to disinfection (precleaning). In this regard, the following general principles apply to all known classes of microbial pathogens.
Specific decontamination requirements will depend on the type of experimental work and the nature of the infectious agent(s) being handled. The generic information given here can be used to develop both standardized and more specific procedures to deal with biohazard(s) involved in a particular laboratory.
Contact times for disinfectants are specific for each material and manufacturer. Therefore, all recommendations for use of disinfectants should follow manufacturers' specifications.
 
Definitions
Many different terms are used for disinfection and sterilization. The following are among the more common in biosafety:
 
Antimicrobial
An agent that kills microorganisms or suppresses their growth and multiplication.
 
Antiseptic
A substance that inhibits the growth and development of microorganisms without necessarily killing them. Antiseptics are usually applied to body surfaces.
 
Biocide
A general term for any agent that kills organisms.
 
Chemical Germicide
A chemical or a mixture of chemicals used to kill microorganisms.
 
Decontamination
Any process for removing and/or killing microorganisms. The same term is also used for removing or neutralizing hazardous chemicals and radioactive materials.
 
Disinfectant
A chemical or mixture of chemicals used to kill microorganisms, but not necessarily spores. Disinfectants are usually applied to inanimate surfaces or objects.54
 
Disinfection
A physical or chemical means of killing microorganisms, but not necessarily spores.
 
Microbicide
A chemical or mixture of chemicals that kills microorganisms. The term is often used in place of “biocide”, “chemical germicide” or “antimicrobial”.
 
Sporocide
A chemical or mixture of chemicals used to kill microorganisms and spores.
 
Sterilization
A process that kills and/or removes all classes of microorganisms and spores.
 
Cleaning Laboratory Materials
Cleaning is the removal of dirt, organic matter and stains. Cleaning includes brushing, vacuuming, drydusting, washing or damp mopping with water containing a soap or detergent. Dirt, soil and organic matter can shield microorganisms and can interfere with the killing action of decontaminants (antiseptics, chemical germicides and disinfectants). Precleaning is essential to achieve proper disinfection or sterilization.
Many germicidal products claim activity only on precleaned items. Pre-cleaning must be carried out with care to avoid exposure to infectious agents.
Materials chemically compatible with the germicides to be applied later must be used. It is quite common to use the same chemical germicide for precleaning and disinfection.
 
Local Environmental Decontamination
Decontamination of the laboratory space, its furniture and its equipment requires a combination of liquid and gaseous disinfectants. Surfaces can be decontaminated using a solution of sodium hypochlorite (NaOCl); a solution containing 1 g/L available chlorine may be suitable for general environmental sanitation, but stronger solutions (5 g/L) are recommended when dealing with high-risk situations. For environmental decontamination, formulated solutions containing 3% hydrogen peroxide (H2O2) make suitable substitutes for bleach solutions.
Rooms and equipment can be decontaminated by fumigation with formaldehyde gas generated by heating paraformaldehyde or boiling formalin.
This is a highly dangerous process that requires specially trained personnel. All openings in the room (i.e. windows, doors, etc.) should be sealed with masking tape or similar before the gas is generated. Fumigation should be conducted at an ambient temperature of at least 21°C and a relative humidity of 70%.
After fumigation the area must be ventilated thoroughly before personnel are allowed to enter. Appropriate respirators must be worn by anyone entering the room before it has been ventilated. Gaseous ammonium bicarbonate can be used to neutralize the formaldehyde. Fumigation of smaller spaces with 55hydrogen peroxide vapor is also effective but requires specialized equipment to generate the vapor.
 
Decontamination of Biological Safety Cabinets
To decontaminate Class I and Class II cabinets, equipment that independently generates, circulates and neutralizes formaldehyde gas is available. Alternatively, the appropriate amount of paraformaldehyde (final concentration of 0.8% paraformaldehyde in air) should be placed in a frying pan on an electric hot plate. Another frying pan, containing 10% more ammonium bicarbonate than paraformaldehyde, on a second hot plate is also placed inside the cabinet. The hot plate leads are plugged in outside the cabinet, so that operation of the pans can be controlled from the outside by plugging and unplugging the hot plates as necessary. If the relative humidity is below 70%, an open container of hot water should also be placed inside the cabinet before the front closure is sealed in place with strong tape (e.g. duct tape).
Heavy gauge plastic sheeting is taped over the front opening and exhaust port to make sure that the gas cannot seep into the room. Penetration of the electric leads passing through the front closure must also be sealed with duct tape.
The plate for the paraformaldehyde pan is plugged in. It is unplugged when all the paraformaldehyde has vaporized. The cabinet is left undisturbed for at least 6 hours.
The plate for the second pan is then plugged in and the ammonium bicarbonate is allowed to vaporize. This plate is then unplugged and the cabinet blower is switched on for two intervals of approximately 2 seconds each to allow the ammonium bicarbonate gas to circulate. The cabinet should be left undisturbed for 30 minutes before the front closure (or plastic sheeting) and the exhaust port sheeting are removed. The cabinet surfaces should be wiped down to remove residues before use.
 
Handwashing/Hand Decontamination
Whenever possible, suitable gloves should be worn when handling biohazardous materials. However, this does not replace the need for regular and proper hand-washing by laboratory personnel. Hands must be washed after handling bio-hazardous materials and animals, and before leaving the laboratory.
In most situations, thorough washing of hands with ordinary soap and water is sufficient to decontaminate them, but the use of germicidal soaps is recommended in high-risk situations. Hands should be thoroughly lathered with soap, using friction, for at least 10 seconds, rinsed in clean water and dried using a clean paper or towel. Foot or elbow-operated faucets are recommended. Where not fitted, a paper/cloth towel should be used to turn-off the faucet handles to avoid recontaminating washed hands. As mentioned above, alcohol-based hand-rubs may be used to decontaminate lightly soiled hands when proper hand-washing is not available.
 
How to Perform Handwashing
  1. Remove jewelry (rings, bracelets) and watches.
  2. Rinse hands and wrists under water.
    56
  3. Apply soap.
  4. Using friction, wash hands for at least 10–15 seconds cleaning between fingers, nail beds, palms, back of hands, wrists and forearms. If hands are visibly soiled, more time may be requires.
  5. Routine hand washing may be performed with any soap. Plain soap with water can physically remove a certain level of microbes, reduce and maintain minimal counts of colonizing flora as well as to mechanically removing the contaminating flora.
  6. If there is no assistant to close the tap, cover the tap with fresh tissue paper and gently close. Taking care to see that the hand does not come in contact with unsterile tap.
  7. Handwashing technique charts are displayed near sinks and can be followed (Figure 1.10).
Additional alcohol-based solutions (HICC approved hand rub) are recommended for use in setting where handwashing facilities are inadequate or inaccessible and hands are not visibly soiled. If these solutions are used as a substitute for handwashing, handwashing with soap and water should be performed as soon as possible after procedure.
 
Improper Handwashing
It is not always possible to protect the skin against various contaminants in the workplace. Therefore, cleaning and taking care of the hands is an important part of developing a proactive, holistic stance against work related skin disorders.
However, for any organization, implementing and maintaining appropriate hand hygiene practices is a daily challenge as there are inconsistent hand hygiene habits across the population:
  • Separate washroom studies from around the world show that only 70% of people wash their hands and only 30% of people actually use soap when washing their hands.
  • People do not wash their hands frequently or adequately enough.
  • The average person washes their hands for around 10 seconds. This will remove around 90% of germs from their hands.
  • Bacteria grow and double in number in less than 20 minutes (Figure 1.11).
 
Heat Disinfection and Sterilization
Heat is the most common among the physical agents used for the decontamination of pathogens. “Dry” heat, which is totally non-corrosive, is used to process many items of laboratory ware which can withstand temperatures of 160° or higher for 2–4 hours. Burning or incineration (Figures 1.12 and 1.13) is also a form of dry heat. “Moist” heat is most effective when used in the form of autoclaving.
Boiling does not necessarily kill all microorganisms and/or pathogens, but it may be used as the minimum processing for disinfection where other methods (chemical disinfection or decontamination, autoclaving) are not applicable or available. Sterilized items must be handled and stored such that they remain uncontaminated until used.
57
zoom view
Figure 1.10: Handwash method
58
zoom view
Figure 1.11: Handwash—areas usually not washed properly
zoom view
Figure 1.12: Proper autoclave use
59
zoom view
Figure 1.13: Incinerator
 
Autoclaving
Saturated steam under pressure (autoclaving) is the most effective and reliable means of sterilizing laboratory materials. For most purposes, the following cycles will ensure sterilization of correctly loaded autoclaves:
  1. 3 minutes holding time at 134°C.
  2. 10 minutes holding time at 126°C.
  3. 15 minutes holding time at 121°C.
  4. 25 minutes holding time at 115°C.
 
Gravity Displacement Autoclave
  • Prevacuum autoclaves: These machines allow the removal of air from the chamber before steam is admitted. The exhaust air is evacuated through a valve fitted with a HEPA-filter. At the end of the cycle, the steam is automatically exhausted.
    These autoclaves can operate at 134°C and the sterilization cycle can therefore be reduced to 3 minutes. They are ideal for porous loads, but cannot be used to process liquids because of the vacuum.
  • Fuel-heated pressure cooker autoclaves: These should be used only if a gravity displacement autoclave is not available. They are loaded from the top and heated by gas, electricity or other types of fuels. Steam is generated by heating water in the base of the vessel, and air is displaced upwards through a relief vent. When all the air has been removed, the valve on the relief vent is closed and the heat reduced. The pressure and temperature rise until the safety valve operates at a preset level. This is the start of the holding time. At the end of the cycle the heat is turned-off and the temperature allowed to fall to 80°C or below before the lid is opened.
    60
 
Loading Autoclaves
Materials should be loosely packed in the chamber for easy steam penetration and air removal. Bags should allow the steam to reach their contents.
 
Precautions in the Use of Autoclaves
The following rules can minimize the hazards inherent in operating pressurized vessels:
  1. Responsibility for operation and routine care should be assigned to trained individuals.
  2. A preventive maintenance program should include regular inspection of the chamber, door seals and all gauges and controls by qualified personnel.
  3. The steam should be saturated and free from chemicals (e.g. corrosion inhibitors) that could contaminate the items being sterilized.
  4. All materials to be autoclaved should be in containers that allow ready removal of air and permit good heat penetration; the chamber should be loosely packed so that steam will reach the load evenly.
  5. For autoclaves without an interlocking safety device that prevents the door being opened when the chamber is pressurized, the main steam valve should be closed and the temperature allowed falling below 80°C before the door is opened.
  6. Slow exhaust settings should be used when autoclaving liquids, as they may boil over when removed due to superheating.
  7. Operators should wear suitable gloves and visors for protection when opening the autoclave, even when the temperature has fallen below 80°C.
  8. In any routine monitoring of autoclave performance, biological indicators or thermocouples should be placed at the center of each load. Regular monitoring with thermocouples and recording devices in a “worst case” load is highly desirable to determine proper operating cycles.
  9. The drain screen filter of the chamber (if available) should be removed and cleaned daily.
  10. Care should be taken to ensure that the relief valves of pressure cooker autoclaves do not become blocked by paper, etc. in the load (Figure 1.12).
 
Incineration
Incineration is useful for disposing of animal carcasses as well as anatomical and other laboratory waste, with or without prior decontamination. Incineration of infectious materials is an alternative to autoclaving only if the incinerator is under laboratory control.
Proper incineration requires an efficient means of temperature control and a secondary burning chamber. Many incinerators, especially those with a single combustion chamber, are unsatisfactory for dealing with infectious materials, animal carcasses and plastics. Such materials may not be completely destroyed and the effluent from the chimney may pollute the atmosphere with microorganisms, toxic chemicals and smoke. However, there are many satisfactory configuration s for combustion chambers. Ideally, the temperature in the primary chamber should be at least 800°C and that in the secondary chamber at least 1000°C. Materials for incineration, even with prior decontamination, should be transported to the incinerator in bags, preferably 61plastic. Incinerator attendants should receive proper instructions about loading and temperature control. It should also be noted that the efficient operation of an incinerator depends heavily on the right mix of materials in the waste being treated (Figure 1.13).
There are ongoing concerns regarding the possible negative environmental effects of existing or proposed incinerators, and efforts continue to make incinerators more environmentally friendly and energy efficient.
 
Disposal
The disposal of laboratory and medical waste is subject to various regional, national and international regulations, and the latest versions of such relevant documents must be consulted before designing and implementing a program for handling, transportation and disposal of biohazardous waste. In general, ash from incinerators may be handled as normal domestic waste and removed by local authorities. Autoclaved waste may be disposed of by off-site incineration or in licensed land fill sites.
 
THE BIOSAFETY OFFICER AND BIOSAFETY COMMITTEE
It is essential that each laboratory organization has a comprehensive safety policy, a safety manual, and supporting programs for their implementation. The responsibility for this normally rests with the director or head of the institute or laboratory, who may delegate certain duties to a biosafety officer or other appropriate personnel.
Laboratory safety is also the responsibility of all supervisors and laboratory employees, and individual workers are responsible for their own safety and that of their colleagues. Employees are expected to perform their work safely and should report any unsafe acts, conditions or incidents to their supervisor. Periodic safety audits by internal or external personnel are desirable.
 
Biosafety Officer
Wherever possible a biosafety officer should be appointed to ensure that biosafety policies and programs are followed consistently throughout the laboratory. The biosafety officer executes these duties on behalf of the head of the institute or laboratory. In small units, the biosafety officer may be a microbiologist or a member of the technical staff, who may perform these duties on a defined part-time basis.
Whatever the degree of involvement in biosafety, the person designated should possess the professional competence necessary to suggest, review and approve specific activities that follow appropriate biocontainment and biosafety procedures. The biosafety officer should apply relevant national and international rules, regulations and guidelines, as well as assist the laboratory in developing standard operating procedures. The person appointed must have a technical background in microbiology, biochemistry and basic physical and biological sciences. Knowledge of laboratory and clinical practices and safety, including containment equipment, and engineering principles relevant to the design, operation and maintenance of facilities is highly desirable. The biosafety officer 62should also be able to communicate effectively with administrative, technical and support personnel.
The activities of the biosafety officer should include the following:
  1. Biosafety, biosecurity and technical compliance consultations.
  2. Periodic internal biosafety audits on technical methods, procedures and protocols, biological agents, materials and equipment.
  3. Discussions of violation of biosafety protocols or procedures with the appropriate persons.
  4. Verification that all staff has received appropriate biosafety training.
  5. Provision of continuing education in biosafety.
  6. Investigation of incidents involving the possible escape of potentially infectious or toxic material, and reporting of findings and recommendations to the laboratory director and biosafety.
  7. Coordination with medical staff regarding possible laboratory-acquired infections.
  8. Ensuring appropriate decontamination following spills or other incidents involving infectious material(s).
  9. Ensuring proper waste management.
  10. Ensuring appropriate decontamination of any apparatus prior to repair or servicing.
  11. Maintaining awareness of community attitudes regarding health and environmental considerations.
  12. Establishment of appropriate procedures for import/export of pathogenic material to/from the laboratory, according to national regulations.
  13. Reviewing the biosafety aspects of all plans, protocols and operating procedures for research work involving infectious agents prior to the implementation of these activities.
  14. Institution of a system to deal with emergencies.
 
Biosafety Committee
A biosafety committee should be constituted to develop institutional biosafety policies and codes of practice. The biosafety committee should also review research protocols for work involving infectious agents, animal use, recombinant DNA and genetically modified materials. Other functions of the committee may include risk assessments, formulation of new safety policies and arbitration in disputes over safety matters.
The membership of the biosafety committee should reflect the diverse occupational areas of the organization as well as its scientific expertise. The composition of a basic biosafety committee may include:
  1. Biosafety officer(s)
  2. Scientists
  3. Medical personnel
  4. Veterinarian(s) (if work with animals is conducted)
  5. Representatives of technical staff
  6. Representatives of laboratory management.
The biosafety committee should seek advice from different departmental and specialist safety officers (e.g. with expertise in radiation protection, industrial safety, fire prevention, etc.) and may at times require assistance from independent experts in various associated fields, local authorities and national regulatory bodies. 63Community members may also be helpful if there is a particularly contentious or sensitive protocol under discussion.
 
Immunization of Staff
The risks of working with particular agents should be fully discussed with individual researchers. The local availability, licensing state and utility of possible vaccines and/ or therapeutic drugs (e.g. antibiotic treatments) in case of exposure should be evaluated before work with such agents is started. Some workers may have acquired immunity from prior vaccination or infection.
If a particular vaccine or toxoid is locally licensed and available, it should be offered after a risk assessment of possible exposure and a clinical health assessment of the individual have been carried out.
Facilities for specific clinical case management following accidental infections should also be available.
 
EMERGENCY SERVICES: WHOM TO CONTACT
The telephone numbers and addresses of the following should be prominently displayed in the facility:
  • Director of the institution or laboratory
  • Laboratory supervisor
  • Biosafety officer
  • Hospitals/ambulance
  • Medical officer.
64