96-24448. Draft Public Health Service (PHS) Guideline on Infectious Disease Issues in Xenotransplantation (August 1996)  

  • [Federal Register Volume 61, Number 185 (Monday, September 23, 1996)]
    [Notices]
    [Pages 49920-49932]
    From the Federal Register Online via the Government Publishing Office [www.gpo.gov]
    [FR Doc No: 96-24448]
    
    
    
    [[Page 49919]]
    
    
    _______________________________________________________________________
    
    Part VI
    
    
    
    
    
    Department of Health and Human Services
    
    
    
    
    
    _______________________________________________________________________
    
    
    
    Public Health Service
    
    
    
    _______________________________________________________________________
    
    
    
    Draft Public Health Service Guideline on Infectious Disease Issues in 
    Xenotransplantation; Notice
    
    Federal Register / Vol. 61, No. 185 / Monday, September 23, 1996 / 
    Notices
    
    [[Page 49920]]
    
    
    
    DEPARTMENT OF HEALTH AND HUMAN SERVICES
    
    Public Health Service
    [Docket No. 96M-0311]
    
    
    Draft Public Health Service (PHS) Guideline on Infectious Disease 
    Issues in Xenotransplantation (August 1996)
    
    AGENCY: Public Health Service, HHS.
    
    ACTION: Notice.
    
    -----------------------------------------------------------------------
    
    SUMMARY: The Public Health Service (PHS) is publishing a document 
    entitled, ``Draft Public Health Service (PHS) Guideline on Infectious 
    Disease Issues in Xenotransplantation (August 1996).'' The demand for 
    human cells, tissues, and organs for clinical transplantation continues 
    to exceed the supply. Thus, the development of xenotransplantation, an 
    investigational therapeutic approach that uses cells, tissues, and 
    organs of animal origin (xenografts) in human recipients, has become an 
    important area of research. The purpose of this draft guideline is to 
    discuss public health issues related to xenotransplantation and 
    recommend procedures to diminish the risk of transmission of infectious 
    agents to the recipient and the general public.
    
    DATES: Written comments December 23, 1996.
    
    ADDRESSES: Submit written comments on the draft guideline to the 
    Dockets Management Branch (HFA-305), Food and Drug Administration 
    (FDA), 12420 Parklawn Dr., rm. 1-23, Rockville, MD 20857. Requests and 
    comments should be identified with the docket number found in brackets 
    in the heading of this document. A copy of the guideline and received 
    comments are available for public examination in the Documents 
    Management Branch between 9 a.m. and 4 p.m., Monday through Friday. The 
    draft guideline is set forth in this document. Submit written requests 
    for single copies of the draft guideline to the Manufacturers 
    Assistance and Communications Staff (HFM-42), Center for Biologics 
    Evaluation and Research (CBER), Food and Drug Administration, 1401 
    Rockville Pike, Rockville, MD 20852-1448. Send one self-addressed 
    adhesive label to assist that office in processing your request. The 
    document may also be obtained by mail or FAX by calling the CBER FAX 
    Information System at 1-888-CBER-FAX or 301-827-3844.
        Persons with access to the INTERNET may obtain the guidance 
    document using FTP, the World Wide Web (WWW), or bounce-back e-mail. 
    For FTP access, connect to CBER at ``ftp://ftp.fda.gov/ CBER/''. For 
    WWW access, connect to CBER at ``http://www.fda.gov/cber/
    cberftp.html''. For bounce back e-mail send a message to 
    ``Xeno@al.cber.fda.gov''.
    
    FOR FURTHER INFORMATION CONTACT: Timothy W. Beth, Center for Biologics 
    Evaluation and Research (HFM-630), Food and Drug Administration, 1401 
    Rockville Pike, suite 200 North, Rockville, MD 20852-1448, 301-594-
    3074.
    
    SUPPLEMENTARY INFORMATION: For the purposes of this draft guideline, 
    the germ ``xenotransplantation'' refers to any procedure that involves 
    the use of live cells, tissues, and organs from a nonhuman animal 
    source, transplanted or implanted into a human or used for ex vivo 
    perfusion. These live nonhuman cells, tissues, or organs are called 
    xenografts. Xenograft products include those from transgenic or 
    nontransgenic animals, as well as combination products that contain 
    xenografts in combination with drugs or devices. Xenograft products do 
    not include nonliving animal products, many of which are regulated as 
    devices (porcine heart valves), drugs (porcine insulin), and other 
    biologicals (bovine serum albumin).
        As with human transplantation, rejection and failure to engraft 
    remain important medical and scientific challenges in 
    xenotransplantation. In addition, there are concerns about potential 
    infectious disease and public health risks. Diseases of animals can be 
    transmitted to humans through routine exposure to, or consumption of, 
    animals. Because transplantation bypasses most of the patient's usual 
    protective physical and immunological barriers, transmission of known 
    and/or unknown infectious agents to humans through xenografts may be 
    facilitated. Moreover, infectious agents vary considerably from one to 
    another with respect to the nature of the risks they present and the 
    difficulty of managing those risks. For example, some agents, such as 
    retroviruses and prions, may not produce clinically recognizable 
    disease until many years after they enter the host, and some infectious 
    agents are not readily detected or identified in tissue samples by 
    current diagnostic techniques.
        Despite the technical barriers and potential risks, 
    xenotransplantation shows promise both as a treatment for a wide range 
    of diseases including chronic metabolic and neurological disorders and 
    as an alternative source of cells, tissues, and organs for clinical 
    transplantation. For these reasons, academic and commercial sponsors 
    are actively pursuing the development of xenograft products and their 
    clinical application. The Health Resources and Services Administration 
    (HRSA) and the Health Care Financing Administration (HCFA) within the 
    Department of Health and Human Services (DHHS) currently administer 
    programs overseeing human organ transplantation under the authority of 
    the National Organ Transplant Act of 1984 (NOTA) (42 U.S.C. 273 et 
    seq., as amended). In the Federal Register of May 2, 1996 (61 FR 
    19722), DHHS published final rules governing performance standards for 
    organ procurement organizations. FDA currently regulates human somatic 
    cell therapies (see ``Application of Current Statutory Authorities to 
    Human Somatic Cell Therapy Products and Gene Therapy Products,'' (58 FR 
    53248, October 14, 1993)) and human tissue for transplantation (21 CFR 
    part 1270).
        The public health safety issues raised by xenotransplantation 
    differ from those of human transplantation in several significant ways. 
    First, the spectrum of infectious agents transmitted via human organ 
    transplantation has been well established, while the full spectrum of 
    infectious agents potentially transmitted via xenograft transplantation 
    is not well known. Infectious agents that produce minimal symptoms in 
    animals may cause severe morbidity and mortality in humans. Second, 
    HRSA oversight and administration of the human organ donor and 
    recipient matching and tracking creates a system that ensures that high 
    standards are maintained in human organ transplantation. Animals are 
    currently commercially bred and raised as a source of food and other 
    products; animals can also be bred and raised as sources of xenograft 
    products for clinical transplantation. As the commercialization of 
    xenograft production increases throughout the United States and the 
    world, the need for consistent standards of source animal screening and 
    quality control will grow. Third, the potentially unlimited supply of 
    animal cells, tissues, and organs may allow opportunities for 
    developing therapeutic approaches to a wide range of diseases for which 
    treatments have heretofore been limited by the insufficient 
    availability of human organs and tissues.
    
    I. Regulation of Xenotransplantation Clinical Investigations
    
        A number of experimental clinical investigations that use xenograft 
    products are being carried out under FDA oversight using the 
    investigational new drug application (IND). Examples of these clinical 
    trials include using
    
    [[Page 49921]]
    
    fetal porcine neural cells for Parkinson's disease, encapsulated bovine 
    adrenal cells for intractable pain, encapsulated porcine islet cells 
    for diabetes, baboon bone marrow for AIDS and transgenic porcine livers 
    as a temporary bridge to human organ transplantation.
        The clinical investigation of drugs and biological products, 
    including xenograft products (live animal cells, tissues, and whole 
    organs), is subject to investigational new drug regulations in 21 CFR 
    part 312, institutional review board regulations in 21 CFR part 56, and 
    informed consent regulations in 21 CFR part 50. FDA plans to develop 
    further guidance, that will be announced in the Federal Register, to 
    assist sponsors in submitting to FDA the appropriate information to be 
    included in an IND for clinical investigation of xenograft products.
    
    II. Recent Events
    
        In 1994 several Institutional Review Board (IRB) committees 
    contacted the Centers for Disease Control and Prevention (CDC) and FDA 
    regarding proposed solid organ xenotransplants from nontransgenic 
    animals, and expressed concern regarding the source and 
    characterization of donor animal tissues. Contemporaneously, the 
    Assistant Secretary of Health requested that agencies in PHS develop a 
    consensus on the infectious disease risks and safety issues raised by 
    xenotransplantation. Even though there were well documented examples of 
    trans-species infection of humans through routine animal exposure, no 
    guidelines existed regarding the adequate screening of donor animal 
    cells, tissues, and organs intended for human transplant or 
    recommendations for post-transplantation patient monitoring.
        To strike a balance between the public health risks and the 
    potential promise of xenotransplantation, FDA, CDC, and the National 
    Institutes of Health (NIH) have worked together to create a draft PHS 
    guideline that seeks to address the concerns raised by the clinical use 
    of xenograft products in humans. As part of the development of the 
    guideline, FDA held an open public meeting of the Biologics Response 
    Modifiers Advisory Committee (BRMAC) on April 21, 1995, at which 
    elements of the draft xenotransplantation guideline and proposed 
    clinical trials were discussed (see 60 FR 15147, March 22, 1995). 
    Essential elements of the draft PHS guideline and a novel clinical 
    trial to use baboon bone marrow for a patient with AIDS were also 
    discussed at the July 13, 1995 meeting of the BRMAC (see 60 FR 32330, 
    June 21, 1995). The PHS agencies including, FDA, CDC, NIH, and HRSA 
    have discussed the development of the draft PHS guideline on infectious 
    disease issues in xenotransplantation at numerous scientific meetings 
    and public forums, and PHS scientists have authored scientific and lay 
    reports on the subject of xenotransplantation.
        FDA, CDC, NIH, and HRSA also supported a study and public workshop 
    by the Institute of Medicine (IOM) on the scientific, public health, 
    and ethical implications of xenotransplantation which culminated in a 
    report released on July 17, 1996, entitled, ``Xenotransplantation: 
    Science, Ethics, and Public Policy'' (hereinafter referred to as the 
    IOM report). In addition to exploring some of the social, scientific, 
    and ethical concerns associated with xenotransplantation, the IOM 
    report also recommended that national guidelines be established for all 
    experimenters and institutions that undertake xenotransplantation 
    trials in humans. (Copies of the IOM report can be obtained from the 
    National Academy Press, 2101 Constitution Ave. NW., Washington, DC 
    20418, 202-334-3313 or 800-624-6242.)
    
    III. Submission of Comments
    
        It is the intention of PHS to revise the draft guideline based on 
    the comments received and to issue a revised guideline at a later date. 
    The availability of any revised guideline will be announced in the 
    Federal Register, the NIH Guide for Grants and Contracts, and CDC's 
    Morbidity and Mortality Weekly Report. As with other guidelines, PHS 
    does not intend this draft guideline to be all-inclusive and cautions 
    that not all information contained therein may be applicable to all 
    situations. The draft guideline is intended to provide information and 
    does not set forth requirements. The methods and procedures cited in 
    the draft guideline are suggestions.
        PHS recognizes that advances will continue in the area of 
    xenotransplantation and that this document may require revision as 
    those advances occur. This draft guideline does not bind PHS and does 
    not create or confer any rights for or on any person and does not 
    operate to bind PHS or the public. The draft guideline represents PHS's 
    current thinking on infectious disease issues in xenotransplantation. 
    In addition, the issuance of this draft guideline by PHS should not be 
    construed as an endorsement of the readiness of xenotransplantation 
    clinical trials or a commitment to direct funds to support additional 
    basic or preclinical research in this area.
        Interested persons may submit written comments regarding this draft 
    PHS guideline at any time to the Dockets Management Branch (address 
    above). Two copies of any comments are to be submitted, except that 
    individuals may submit one copy. Comments are to be identified with the 
    docket number found in brackets in the heading of this document. 
    Comments received will be considered in any revision to the ``Draft 
    Public Health Service (PHS) Guideline on Infectious Disease Issues in 
    Xenotransplantation (August 1996).''
        The text of the draft guideline follows.
    
    Draft Public Health Service (PHS) Guideline on Infectious Disease 
    Issues in Xenotransplantation (August 1996)
    
    Table of Contents
    
    1. Introduction
        1.1. Background
        1.2. Scope of the Document
        1.3. Objectives
    2. Xenotransplantation Protocol Issues
        2.1. Xenotransplant Team
        2.2. Clinical Xenotransplantation Site
        2.3. Clinical Protocol Review
        2.4. Health Surveillance Plans
        2.5. Written Informed Consent and Recipient Education
    3. Animal Sources for Xenotransplants
        3.1. Animal Procurement Sources
        3.2. Biomedical Research Animal Facilities
        3.3. Preclinical Screening for Know Infectious Agents
        3.4. Herd/Colony Health Maintenance and Surveillance
        3.5. Individual Source Animal Screening and Qualification
        3.6. Procurement and Screening of Xenografts
        3.7. Archives of Source Animal Medical Records and Specimens
    4. Clinical Issues
        4.1. Xenotransplant Recipient
        4.2. Contacts of Recipient
        4.3. Hospital Infection Control
        4.4. Health Care Records
    5. Public Health Needs
        5.1. National Registry
        5.2. Serum and Tissue Archives
    6. Bibliography
    
    1. Introduction
    
    1.1. Background
    
        The demand for human cells, tissues, and organs for clinical 
    transplantation continues to exceed the supply. The resultant limited 
    availability of human allografts, coupled with recent scientific and 
    biotechnical advances, has prompted the development of new 
    investigational therapeutic approaches that use cells, tissues, and 
    organs of animal origin (xenografts) in human recipients. Transmission 
    of infections (HIV/AIDS, Creutzfeldt-Jakob Disease, rabies, hepatitis 
    B, hepatitis C, etc.) via transplanted human allografts has been well 
    documented. The use of live
    
    [[Page 49922]]
    
    animal cells, tissues, and organs for transplantation or hemoperfusion 
    of humans raised unique public health concerns about potential 
    infection of the patient with both recognized and/or unknown infectious 
    agents. Additionally, subsequent introduction of these xenogeneic 
    infectious agents into and propagation through the general human 
    population is a risk that must be addressed.
        Zoonoses are defined as diseases of animals transmitted to humans 
    via routine exposure to or consumption of the source animal. Many 
    agents responsible for zoonoses are well characterized and identifiable 
    through available diagnostic tests, e.g., Toxoplasma species, 
    Salmonella species, or Herpes B virus of monkeys. However, public 
    health concerns exist regarding the potential transmission of 
    xenogeneic infectious agents not recognized as classical zoonoses from 
    xenografts to recipients, and then from the recipient to other persons. 
    The intimate contact between the recipient and the xenograft, the 
    associated disruption of anatomical barriers, and immunosuppression of 
    the recipient are more likely to facilitate interspecies transmission 
    of xenogeneic infectious agents than normal contact between humans and 
    animals.
        Emerging infectious agents may not be readily identifiable with 
    current techniques, as exemplified by the delay of several years in 
    identifying HIV-1 as the pathogenic agent for AIDS. Improvement in 
    diagnostic techniques facilitated investigation of exogenous and 
    endogenous retroviruses in all species. Retroviruses and other 
    persistent viral infections may be associated with acute disease with 
    varying incubation periods, followed by periods of clinical latency 
    prior to the onset of clinically evident malignancies or other chronic 
    diseases. As the HIV/AIDS pandemic demonstrates, persistent viral 
    infections may result in person to person transmission for many years 
    before clinical disease develops in the index case, thereby allowing an 
    emerging infectious agent to become established in the susceptible 
    population before it is recognized.
    
    1.2. Scope of the Document
    
        The draft guideline discusses public health issues related to 
    xenotransplantation and recommends procedures for diminishing the risk 
    of transmission of infectious agents to the recipient, health care 
    workers, and the general public. This draft guideline applies to all 
    xenotransplantation procedures performed in the United States. For the 
    purposes of this draft guideline, the term ``xenotransplantation'' 
    refers to any procedure that involves the use of live cells, tissues 
    and organs from a non-human animal source, transplanted or implanted 
    into a human or used for ex vivo perfusion. This draft guideline 
    reflects the status of the field of xenotransplantation and knowledge 
    of the risk of xenogeneic infections at the time of publication. This 
    draft guidelines will require periodic review and may require 
    modification when justified by advances in scientific knowledge and 
    clinical experience.
    
    1.3. Objectives
    
        The objective of this draft Public Health Service (PHS) guideline 
    is to present measures that can be used to minimize the risk to the 
    public of human disease due to known zoonoses and emerging xenogeneic 
    infectious agents arising from xenotransplantation. In order to achieve 
    this goal, this document:
        1.3.1. Outlines the composition and function of the xenotransplant 
    team in order that appropriate technical expertise can be applied and 
    that adequate data management, tissue storage, and surveillance 
    procedures can be established.
        1.3.2. Discusses aspects of the clinical protocol, clinical center 
    and the informed consent relevant to public health concerns regarding 
    infections associated with xenotransplantation.
        1.3.3. Provides a framework for pretransplantation animal source 
    screening to minimize the potential for cross-species transmission of 
    known and unknown zoonotic agents.
        1.3.4. Recommends approaches for postxenotransplantation 
    surveillance to monitor for the potential transmission to the recipient 
    and health care workers of infectious agents, including unlikely or 
    previously unrecognized agents.
        1.3.5. Recommends hospital infection control practices to reduce 
    the risk of nosocomial transmission of xenogeneic infectious agents.
        1.3.6. Recommends the archiving of biologic samples, (including 
    sera, plasma, leukocytes, and tissues), from the source animal and the 
    transplant recipient for the potential investigation of infectious 
    diseases arising from xenotransplantation which could impact upon the 
    public health.
        1.3.7. Recommends the creation of a centralized database. This 
    database will address the need for long term safety data required for 
    public health investigations.
    
    2. Xenotransplantation Protocol Issues
    
    2.1. Xenotransplant Team
    
        The transplantation of animal cells, tissues, and organs requires 
    expertise in the evaluation of infectious agents in the source animal 
    and in the recipient. Consequently, in addition to transplant surgeons, 
    the xenotransplantation team should include as active participants such 
    individuals as: (1) Infectious disease physician with expertise in 
    zoonoses, transplantation, and microbiology; (2) veterinarian with 
    specific expertise in the animal husbandry issues and infectious 
    diseases (particularly zoonoses) of the animal species serving as the 
    source of transplanted cells, tissues or organs (animal source); (3) 
    transplant immunologist; (4) hospital epidemiologist/infection control 
    specialist; and (5) director of the clinical microbiology laboratory.
    
    2.2. Clinical Xenotransplantation Site
    
        All clinical centers involved with xenotransplantation should have 
    active participation with accredited virology and microbiology 
    laboratories that have the documented expertise and capability to 
    isolate and identify unusual and unknown pathogens of both human and 
    veterinary origin. Centers where solid organ xenotransplantation 
    procedures are performed should be members of the Organ Procurement and 
    Transplantation Network and abide by its policies in accordance with 
    Section 1138 of the Social Security Act (42 U.S.C. 13206-13208).
    
    2.3. Clinical Protocol Review
    
        After completion of internal review by all members of the 
    xenotransplant team, clinical protocols should be reviewed by the 
    clinical center Biosafety Committee, Institutional Animal Care and Use 
    Committee (IACUC), and Institutional Review Board (IRB). The Biosafety 
    Committee should have the expertise to assess the potential risks of 
    infection for contact population (including health care providers, 
    family, friends, and the community at large) and the recipient. The 
    IACUC should have the expertise to evaluate epidemiological concerns 
    related to conditions of source animal husbandry (e.g., frequency of 
    screening, animal quarantine, etc.). The IRB should have expertise in 
    human and veterinary infectious diseases, including virology and 
    laboratory diagnostics, epidemiology, and risk assessment. The review 
    committees should discuss their comments and suggestions with the 
    members of the health care team and the informed consent document 
    should
    
    [[Page 49923]]
    
    incorporate and reflect these comments, as needed. In addition, live 
    animal cells, tissues, and organs intended for use in humans are 
    subject to regulation by FDA under the Public Health Service Act and 
    the Federal Food, Drug, and Cosmetic Act (42 U.S.C. 262, 264 and 21 
    U.S.C. 301 et seq.).
    
    2.4. Health Surveillance Plans
    
        The clinical protocols for xenotransplantation should describe the 
    methodologies for screening for known infectious agents before 
    transplantation (including the herd, the individual animal and the 
    xenograft) and surveillance after transplantation (including the 
    recipient(s), their contacts, and the health care workers (section 4)). 
    The agents and screening methods may vary with the different types of 
    procedures, the cells, tissues, and organs used, and the animal source. 
    The clinical protocol should include a summary of the relevant aspects 
    of the health maintenance and surveillance program of the herd and the 
    medical history of the source animal(s) (section 3).
    
    2.5. Written Informed Consent and Recipient Education
    
        In the process of obtaining and documenting informed consent, the 
    investigator should comply with the applicable regulatory 
    requirement(s) (e.g., 45 CFR part 46; 21 CFR part 50), and should 
    adhere to good clinical practices and to the ethical principles derived 
    from the Belmont Report of the National Commission for the Protection 
    of Human Subjects of Biomedical and Behavioral Research. The informed 
    consent discussion, the written informed consent form, and the written 
    information provided to subjects should address the following points 
    relating to the risk of xenotransplantation:
        2.5.1. The potential for infection from zoonotic agents known to be 
    associated with the donor species.
        2.5.2. The potential for transmission of unknown xenogeneic 
    infectious agents to the recipient. The patient should be informed of 
    the uncertainty regarding these risks, the possibility that infections 
    with these agents may not be recognized for some time, and that the 
    nature of clinical diseases that these agents may cause are unknown.
        2.5.3. The potential risk for transmission of xenogeneic infectious 
    agents to the recipient's family or close contacts, especially sexual 
    contacts. Close contacts are defined as household members and others 
    with whom the recipient participates in activities that could result in 
    exchanges of body fluids. The recipient should be informed that 
    transmission of these agents may be minimized by the use of barriers 
    during sexual intercourse and that infants, pregnant women, elderly, 
    and chronically ill or immunosuppressed persons may be at increased 
    risk for infection from zoonotic or opportunistic agents (section 4.2).
        2.5.4. Any need for isolation procedures during hospitalization 
    (including the estimated duration of such confinement), and any 
    specialized precautions (e.g., dietary, travel) following hospital 
    discharge.
        2.5.5. The need to comply with long-term or potentially life-long 
    surveillance necessitating routine physical evaluations with archiving 
    of tissue and/or serum specimens. The schedule for clinical and 
    laboratory monitoring should be provided to the extent possible. The 
    patient should be informed that any serious or unexplained illness in 
    themselves or their contacts should be reported to their physician 
    immediately.
        2.5.6. The need for the subject to inform the investigator or his/
    her designee of any change in address or telephone number in order to 
    maintain accurate data for long-term health surveillance.
        2.5.7. Discussion with the patient regarding performance of a 
    complete autopsy. Joint discussion with the recipient and his/her 
    family concerning the need to conduct an autopsy is also encouraged in 
    order to communicate the recipient's intent.
        2.5.8. Access by the appropriate public health agencies to all 
    medical records. To the extent permitted by applicable laws and/or 
    regulations, the confidentiality of medical records will be maintained.
        2.5.9. Consent forms should state clearly that xenograft recipients 
    should never, subsequent to receiving the transplant, donate Whole 
    Blood, blood components, Source Plasma, Source Leukocytes, tissues, 
    breast milk, ova, sperm, or any other body parts for use in humans.
    
    3. Animal Sources For Xenotransplants
    
        Recognized zoonotic infectious agents and other organisms present 
    in animals, such as normal flora or commensals, may cause disease in 
    humans when introduced by transplantation of cells, tissues, or organs, 
    especially in immunocompromised patients. The ability to screen 
    extensively the cells, tissues, or organs intended for clinical use may 
    be limited by the need to ensure graft viability. The risk of 
    transmitting infectious agents can be minimized by procurement of 
    source animals from herds or colonies that are screened and qualified 
    as pathogen free for specific agents appropriate for the clinical 
    application, and are maintained in an environment that minimizes 
    exposure to vectors of infectious agents.
    
    3.1. Animal Procurement Sources
    
        3.1.1. Cells, tissues, and organs intended for use in 
    xenotransplantation should be procured only from animals with 
    documented lineages and that have been bred and reared in captivity.
        3.1.2. Animals should be obtained from closed herds or colonies 
    that are serologically well-characterized and as free as possible of 
    infectious agents of concern for the animal species and the patient.
        3.1.3. The use of animals from controlled environments such as 
    closed corrals (captive free-ranging animals) should be used only when 
    they are the only suitable source for a given xenotransplant procedure. 
    Such animals require more intensive screening because of the higher 
    likelihood that they harbor adventitious infectious agents from 
    uncontrolled contact with arthropods and/or other animals.
        3.1.4. Wild-caught animals should not be used as sources for cells, 
    tissues, or organs intended for transplantation.
        3.1.5. Imported animals or the first generation of offspring of 
    imported animals should not be used as a source of cells, tissues, or 
    organs unless the animals belong to a species or strain not available 
    for use in the United States. In this case, their use should be 
    considered only if the source characteristics for the imported animals 
    can be documented, validated, and audited.
        3.1.6. Source animals from species in which prion-mediated diseases 
    (e.g., transmissible spongiform encephalopathies) have been reported 
    should be obtained from closed herds with documented absence of 
    dementing illnesses and controlled food sources (section 3.2.1.3). 
    Bovine transplant tissue should not be obtained from countries 
    designated by the United States Department of Agriculture (USDA) as 
    those where bovine spongiform encephalopathy (BSE) exists (59 FR 44591, 
    August 29, 1994, and 60 FR 44036, August 24, 1995).
        3.1.7. Animals or live animal cells, tissues, or organs obtained 
    through abattoirs should not be used as a source of xenografts. These 
    animals are obtained from geographically divergent farms or markets and 
    are more likely to carry infectious agents due to increased exposure to 
    other animals, and increased activation and shedding of infectious 
    agents during the stress of slaughter. In addition, health histories
    
    [[Page 49924]]
    
    of slaughterhouse animals are usually not available.
    
    3.2. Biomedical Research Animal Facilities
    
        For the purposes of xenotransplantation, animals should be housed 
    in facilities built and operated in accordance with standards outlined 
    in this section. As a minimum, these facilities should meet the 
    recommendations of the Guide for the Care and Use of Laboratory Animals 
    (the criteria for accreditation by the American Association for the 
    Accreditation of Laboratory Animal Care (AAALAC)) and be subject to 
    inspection by appropriate members of the transplant teams and public 
    health agencies. Animal facilities should have a routine well-
    documented herd health and surveillance system. Animal facilities 
    should have on staff veterinarians with expertise in the infectious 
    diseases prevalent in the animal species and should maintain active 
    collaboration with accredited microbiology laboratories.
        3.2.1. The biomedical animal facility standard operating procedures 
    should be thoroughly described regarding the following: (1) Criteria 
    for animal admission; (2) description of the disease monitoring 
    program; (3) criteria for the isolation or elimination of diseased 
    animals; (4) criteria for the health screening and surveillance of 
    humans entering the facility; (5) facility cleaning arrangements; (6) 
    the source and delivery of feed, water, and supplies; (7) measures to 
    exclude arthropods and other animals; (8) animal transportation; and 
    (9) dead animal disposition. Entry and exit of animals, animal care 
    staff, and other humans should be controlled to minimize environmental 
    exposures/inadvertent exposure to transmissible infectious agents.
        3.2.1.1. Animal movement through the secured facility should be 
    described in the standard operating procedures of the facility. All 
    animals introduced into the source colony other than by birth should go 
    through a well-defined quarantine and testing period (section 3.5). 
    With regard to the reproduction and raising of suitable animals, the 
    use of methods such as artificial insemination (AI), embryo transfer, 
    medical early weaning (MEW), cloning, or hysterotomy/hysterectomy and 
    fostering may minimize further colonization with infectious agents.
        3.2.1.2. During final screening and qualification of individual 
    source animals and xenograft procurement, the potential for 
    transmission of an infectious agent is minimized by utilizing a step-
    wise ``batch'' or ``all-in/all-out'' method of source animal movement 
    through the facility rather than continuous replacement movement. With 
    the ``all-in/all-out'' or ``batch'' method, one or more individual 
    source animals are selected from the closed herd or colony and 
    quarantined while undergoing final screening qualification and graft 
    procurement. After the entire batch of source animals is removed, the 
    quarantine and graft processing areas of the animal facility are then 
    washed and disinfected prior to the introduction of the next batch of 
    source animals.
        3.2.1.3. The feed components, including any medicinals or other 
    additives, should be documented for a minimum of one generation prior 
    to the source animal. The absence of recycled or rendered animal 
    materials in feed should be specifically documented. The absence of 
    such materials is important for the prevention of prion-associated 
    diseases and slow viral infections, as well as for the prevention of 
    transmission of other infectious agents. Potentially extended periods 
    of clinical latency, severity of consequent disease, and the difficulty 
    in current detection methods highlight the importance of eliminating 
    risk factors associated with prion transmission.
        3.2.1.4. Facilities supplying research animals for use in 
    xenotransplant protocols should maintain a source animal record system 
    that documents every animal, organ, tissue, or type of cells supplied 
    for transplantation, and the transplant centers where these were sent. 
    Facilities should maintain records of the following: the lifelong 
    health history of the source animals (section 3.5), the herd health 
    surveillance (sections 3.3, 3.4), and the standard operating procedures 
    of the animal procurement facility (section 3.2). An animal numbering 
    or other identifier system should be employed to allow easy, accurate, 
    and rapid linkage between the information contained in these different 
    record systems.
        3.2.1.5. In the event that the biomedical animal facility ceases to 
    operate, all animal health records and specimens should be transferred 
    to the respective clinical transplant centers or the centers should be 
    notified of the new archive site.
    
    3.3. Preclinical Screening for Known Infectious Agents
    
        The following points discuss measures for appropriate screening of 
    known infectious agents in the herd, individual source animal, and the 
    xenograft (sections 3.4, 3.5, 3.6).
        3.3.1. Preclinical studies should be performed in conjunction with 
    the development of specific clinical applications for the use of 
    xenografts. These preclinical studies should be species specific in the 
    identification of microbial agents in xenografts. These studies should 
    characterize the potential of identified agents for human 
    pathogenicity. Characterization of the human pathogenicity of 
    xenotropic endogenous retroviruses and persistent viral infections 
    present in source animal cells, tissues, and organs is particularly 
    important.
        3.3.1.1. These preclinical studies should identify appropriate 
    assays for the screening program to qualify xenografts for clinical 
    use.
        3.3.2. Programs for screening and detection of known infectious 
    agents in the herd or colony, the individual source animal, and the 
    xenograft should be tailored for the source animal species and clinical 
    application and be updated periodically to reflect advances in the 
    knowledge of infectious diseases. The xenotransplant team should be 
    responsible for the adequacy of the screening program.
        3.3.3. All assays used for the screening and detection of 
    infectious agents (both commensals and pathogens) in the herd or 
    colony, in the individual source animal, and in the final analysis of 
    the xenograft should have well documented specificity and sensitivity 
    as well as validity in the setting in which they are employed. Assays 
    under development may complement the screening process.
        3.3.4. Samples from xenografts should be tested preclinically with 
    cocultivation assays that include a panel of appropriate indicator 
    cells, including human peripheral blood mononuclear cells (PBMC), to 
    facilitate amplification and detection of xenotropic endogenous 
    retroviruses and other xenogeneic viruses capable of producing 
    infection in humans. The selection of indicator cells on the 
    cocultivation panel should be determined by the xenograft and its 
    clinical applications. For instance, xenotransplantation involving the 
    human central nervous system (CNS) may warrant cocultivation of samples 
    from the xenograft with a human neuronal cell line in the attempt to 
    detect neurotropic viruses. Serial blind passages and observation for 
    cytopathic effect, focus formation, reverse transcriptase assay, and 
    electron microscopy may be appropriate. When cultures suggest the 
    presence of viral agents, immunologic or genetic techniques (enzyme 
    immunoassays for detection of serologic cross-reactivity, 
    immunofluorescence or other immunoassays, Southern blot analysis, 
    polymerase chain reaction (PCR)
    
    [[Page 49925]]
    
    techniques, PCR-based reverse transcriptase assay etc.) or cross-
    species in vivo culturing techniques may be useful. Detection of latent 
    viruses may be facilitated by their activation using chemical and 
    irradiation methods. For detection of possible bacteria, universal PCR 
    probes are available and should be considered for screening of 
    xenografts.
    
    3.4. Herd/Colony Health Maintenance and Surveillance
    
        The principal elements recommended to qualify a herd or colony as a 
    source of animals for use in xenotransplantation include: (1) Closed 
    herd or colony, and (2) adequate surveillance programs for infectious 
    agents. Documentation of the herd or colony health maintenance and 
    surveillance program relevant to the specific application should be 
    available in the standard operating procedure of the animal facility. 
    These procedures should be available to the review committees. 
    Permanent medical records for the herd or colony and the specific 
    individual source animals should be maintained indefinitely at the 
    animal facility.
        3.4.1. Herd or colony health measures that constitute standard 
    veterinary care for the species (e.g., anti-parasitic measures) should 
    be implemented and recorded at the animal facility. For example, 
    aseptic techniques and sterile equipment should be used in all 
    parenteral interventions including vaccinations, phlebotomy, and 
    biopsies. All incidents that may affect herd or colony health should be 
    recorded (e.g., breaks in the environmental barriers of the secured 
    facility, disease outbreaks, or sudden animal deaths). Vaccination and 
    screening schedules should be described in detail. The use of live 
    vaccines is discouraged but may be justified when dead or acellular 
    vaccines are not available. Their use should be documented and taken 
    into account in the risk assessment.
        3.4.2. In addition to standard medical care, the herd/colony should 
    be monitored for the introduction of infectious agents which may not be 
    apparent clinically. The standard operating procedures should describe 
    this monitoring program, including the types and the schedules of 
    physical examinations and laboratory tests used in the detection of 
    infectious agents.
        3.4.3. Routine testing of closed herds or colonies in the United 
    States should concentrate on zoonoses known to exist in captive animals 
    of the relevant species in North America. Because many important 
    pathogens are not endemic to the United States or have been found only 
    in wild-caught animals, testing of breeding stock and maintenance of a 
    closed herd or colony reduces the need for extensive testing of 
    individual source animals. Herd or colony geographic locations are 
    relevant to consideration of presence and likelihood of pathogens in a 
    given herd or colony. Veterinarians familiar with the prevalence of 
    different infectious agents in the geographic area of source animal 
    origin and the location where the source animals are to be maintained 
    should be consulted.
        3.4.3.1. As part of the surveillance program, routine serum samples 
    should be obtained from randomly selected animals representative of the 
    herd or colony population. These samples should be tested for 
    infectious agents relevant to the species and epidemiologic exposures. 
    Additional directed serologic analysis or active culturing of 
    individual animals should be performed in response to clinical 
    indications. Infection in one animal in the herd justifies a larger 
    clinical and epidemiologic evaluation of the rest of the herd or 
    colony. In addition, serum samples should be stored indefinitely at the 
    animal research facility for investigation of unexpected disease either 
    in the herd or colony, individual source animals, or in the xenograft 
    recipient or contacts.
        3.4.3.2. Any animal deaths where the cause is unknown or ambiguous, 
    including all fetal stillbirths or abortions, should lead to full 
    necropsy and evaluation for infectious etiologies with documentation.
        3.4.3.3. Standard operating procedures that maintain a subset of 
    sentinel animals for the duration of their natural life are encouraged. 
    Life-long monitoring of these animals will increase the probability of 
    detection of subclinical, latent or late-onset diseases such as prion-
    mediated disease.
    
    3.5. Individual Source Animal Screening and Qualification
    
        The qualification of indivudal source animals should include breed 
    and lineage, and documentation of general health, including vaccination 
    history with attention to use of any live attenuated vaccines. The 
    presence of pathogens resulting in acute infections should be 
    controlled for by clinical examination and treatment of individual 
    source animals, by use of appropriate individual quarantine periods 
    that extend beyond the incubation period of pathogens of concern, and 
    by herd surveillance indicating the presence or absence of infection in 
    the herd from which the individual source animal is selected. During 
    quarantine, individual source animals should be screened for infectious 
    agents relevant to the particular clinical application.
        3.5.1. Individual source animals should be quarantined for at least 
    3 weeks prior to xenograft procurement. During this time, acute 
    illnesses due to infectious agents to which the animal may have been 
    exposed shortly before removal from the herd or colony would be 
    expected to become clinically apparent. It may be appropriate to modify 
    this quarantine period depending upon the characterization and 
    surveillance of the source animal herd or colony and the clinical 
    urgency. When the quarantine period is shortened, justification should 
    be documented in the protocol and the potentially increased infectious 
    risk incurred should be addressed in the informed consent document.
        3.5.1.1. During the quarantine period, candidate source animals 
    should be screened for the presence of infectious agents (bacteria, 
    parasites, and viruses) by appropriate serologies and cultures, 
    complete blood count and peripheral blood smear, and fecal exam for 
    parasites. The screening program should be guided by the surveillance 
    and health history of the herd or colony. Evaluation for viral agents 
    which may not be recognized zoonotic agents but which have been 
    documented to infect either human or non-human primate cells in vivo or 
    in vitro should be considered. Particular attention should be given to 
    viruses with demonstrated capacity for recombination, complementation, 
    or pseudotyping. These tests should be performed as closely as possible 
    to the date of transplantation while ensuring availability of results 
    prior to clinical use.
        3.5.1.2. Screening of a candidate source animal should be repeated 
    prior to xenograft procurement if a period greater than 3 months has 
    elapsed since the initial screening and qualification was performed 
    (e.g., if the planned xenograft was not procured or a second xenograft 
    is obtained) or if the animal has been in contact with other 
    nonquarantined animals between the quarantine period and the time of 
    cells, tissue or organ procurement.
        3.5.1.3. Transportation of source animals may compromise the 
    protection ensured by the closed colony. Careful attention to 
    conditions of transport can minimize but not eliminate disease 
    exposures during shipping. A more extensive period of quarantine and 
    screening comparable to that used for entry of new animals into a 
    closed herd or colony should be instituted upon arrival. Xenografts 
    should be procured, when feasible, at the animal facility and
    
    [[Page 49926]]
    
    transported as the cells, tissues, or organ to be transplanted.
        3.5.2. All procured cells, tissues, and organs intended for 
    clinical use should be as free of infectious agents as possible. When 
    feasible, the use of source animals in whom infectious agents, 
    including latent viruses, have been identified should be avoided. The 
    presence of an agent in certain anatomic sites, for example the 
    alimentary tract, may not preclude use of the source animal if the 
    agent is documented to be absent in the xenograft.
        3.5.3. If feasible and when it is unlikely to compromise the 
    xenograft, a biopsy should be studied for infectious agents by 
    appropriate screening assays (section 3.3) and appropriate 
    histopathology prior to transplantation, and then archived (section 
    3.7). The results from all studies should be reviewed by the principal 
    investigator prior to clinical use of the xenograft.
        3.5.4. The sources, relevant husbandry, and health history 
    (including use as experimental subjects) of herds and/or individual 
    source animals should be available to the reviewing committees. All 
    relevant health records for the life of the animal, including both the 
    herd and the individual source animal records and a full history of 
    vaccinations, should be available and reviewed prior to candidate 
    animal selection and procurement of cells, tissues, and organs. These 
    records should be maintained indefinitely for retrospective review. A 
    copy of the individual source animal record should accompany the 
    xenograft and be archived as part of the permanent medical record of 
    the xenograft recipient.
        3.5.5. The biomedical animal facility should notify the clinical 
    center in the event that an infectious agent is identified in the 
    source animal or herd subsequent to xenograft harvest (e.g., 
    identification of delayed onset prion-mediated disease in a sentinel 
    animal).
    
    3.6. Procurement and Screening of Xenografts
    
        3.6.1. Procurement and processing of cells, tissues, and organs 
    should be performed using documented aseptic conditions designed to 
    minimize contamination. These procedures should be conducted in 
    designated facilities which are subject to inspection.
        3.6.2. Procedures that may inactivate or remove pathogens without 
    compromising the integrity and function of the xenograft should be 
    employed.
        3.6.3. Cells, tissues, or organs intended for transplantation that 
    are maintained in culture prior to transplant should be periodically 
    screened for maintenance of sterility, including screening for viruses 
    and mycoplasma (section 3.3.4). The FDA publications entitled ``Points 
    to Consider in Somatic Cell and Gene Therapy (1991),'' ``Points To 
    Consider in the Characterization of Cell Lines Used to Produce 
    Biologicals (1993),'' and ``Points to Consider in the Manufacture and 
    Testing of Therapeutic Products for Human Use Derived from Transgenic 
    Animals (1995)'' should be consulted for guidance.
        3.6.4. To ensure reproducible quality control of the procurement 
    and screening process, all events involved in procurement of the 
    xenograft up to the point of transplanting the tissue into the patient 
    should be rehearsed and documented.
        3.6.5. When the animal is euthanatized during procurement of the 
    cells, tissue, or organ, a full necropsy should be conducted including 
    gross, histopathological, and microbiological evaluation. When 
    xenografts are procured without euthanatizing the source animal, the 
    animal's health should be monitored for life. When these animals die or 
    are euthanatized, a full necropsy should follow, regardless of the time 
    elapsed between graft procurement and death. The results of the 
    necropsy, documented in the animal's permanent medical record, should 
    be archived indefinitely. In the event that the necropsy findings 
    suggest infections pertinent to the health of the xenograft 
    recipient(s) (e.g., evidence of prion-associated disease) the finding 
    should be communicated to all transplant centers that receive cells, 
    tissues, or organs from this source animal (section 3.5.5.).
    
    3.7. Archives or Source Animal Medical Records and Specimens
    
        Systematically archived source animal biologic samples and 
    recordkeeping that allows rapid and accurate linking of xenograft 
    recipients to the individual source animal records and archived 
    biologic specimens are essential for public health investigation and 
    containment of emergent xenogeneic infections.
        3.7.1. Responsibility for the care of, and access to, tissue 
    archiving and recordkeeping should be clearly designated in the 
    research and clinical protocol.
        3.7.2. Animal source herd or colony health records, individual 
    source animal health records, and records of the screening analysis of 
    the xenograft should be maintained indefinitely. A summary of the 
    individual source animal health record and a record of the xenograft 
    screening qualification should be filed at the clinical transplant site 
    as part of the xenotransplant recipient medical record.
        3.7.3. For the purposes of retrospective public health 
    investigations, source animal biologic specimens should be banked at 
    the time of graft procurement and designated for public health. All 
    specimens should remain in archival storage indefinitely to permit 
    retrospective analysis if a public health need arises (section 
    4.1.1.4.). Archived source animal biologic specimens should be readily 
    accessible and linkable to both source animal and recipient(s) health 
    records.
        3.7.4. Ideally, at least five 0.5cc aliquots of each source animal 
    serum and plasma should be banked. At least three aliquots of viable 
    (1 x 107) leukocytes should be cryopreserved. Optimally, DNA and 
    RNA extracted from leukocytes should also be aliquoted and banked. 
    Additionally, paraffin-embedded, formalin fixed, and cryopreserved 
    tissue samples representative of major organ systems (e.g., spleen, 
    liver, bone marrow, central nervous system) should be collected from 
    source animals euthanatized concomitant with procurement of the 
    xenograft.
    
    4. Clinical Issues
    
    4.1. Xenotransplant Recipient
    
        4.1.1. Surveillance of the xenotransplant recipient. Post-
    transplantation clinical and laboratory surveillance of xenograft 
    recipients is critical to monitor for the introduction and propagation 
    of xenogeneic infectious agents in the general population. Performance 
    and documentation of this surveillance should be the responsibility of 
    the clinical center and should continue throughout the life of the 
    recipient. Appropriate surveillance methods include the following:
        4.1.1.1. Adverse clinical events potentially associated with 
    xenogeneic infections should be evaluated during periodic clinic visits 
    following the transplant procedure.
        4.1.1.2. Biological specimens should be collected and archived to 
    allow retrospective investigation of possible xenogeneic infections. 
    These biological specimens should be designated for public health 
    investigative purposes. Specimens to be collected should be appropriate 
    to the specific transplant situation. Serum, plasma, and peripheral 
    blood mononuclear cells (PBMC's) should be collected. Preferably, at 
    least three to five 0.5cc aliquots of citrated or EDTA-anticoagulated 
    plasma should be banked
    
    [[Page 49927]]
    
    at the predetermined time points outlined below. At least 2 aliquots of 
    viable leukocytes (1 x 107) should be cryopreserved. Additionally, 
    DNA and RNA extracted from leukocytes (1 x 107) and/or sera could 
    be aliquoted and banked. Specimens of any xenograft that is removed 
    (e.g., post-rejection or at time of death) should be banked.
        The following schedule for archiving biological specimens is 
    recommended: (1) Two sets of samples should be archived 1 month apart 
    before the xenotransplant procedure. If this is not feasible then two 
    sets should be archived as temporally separated as possible, (2) a set 
    should be archived in the immediate posttransplant period and at 
    approximately 1 month and 6 months post transplantation, (3) collection 
    should then be obtained annually for the first 2 years after 
    transplant, (4) After that, specimens should be archived every 5 years 
    for the remainder of the recipient's life. More frequent archiving may 
    be indicated by the specific protocol or the recipient's medical 
    course.
        4.1.1.3. In the event of death of the recipient, snap-frozen 
    samples store at -70 deg. C, paraffin embedded tissue, and tissue 
    suitable for electron microscopy should be collected at autopsy from 
    the xenograft and all major organs relevant to either the transplant or 
    the clinical syndrome resulting in death. These specimens should be 
    archived indefinitely for potential public health use.
        4.1.1.4. The clinical center should be responsible for maintaining 
    an ongoing and accurate archive of biologic specimens. In the absence 
    of a central facility (section 5.2) the designated public health 
    biologic specimens should be archived with appropriate safeguards to 
    ensure long-term storage (e.g., a monitored storage freezer alarm 
    system and specimen archiving in split portions in separate freezers) 
    and an efficient system for the prompt retrieval and linkage of data to 
    medical records of recipients and source animals.
        4.1.1.5. In addition to archiving of biologic specimens, active 
    laboratory surveillance program of the xenograft recipient should be 
    instituted when xenogeneic agents are known or suspected to be present 
    in the xenograft. The intent of active screening in this setting is 
    detection of sentinel human infections prior to dissemination in the 
    general population. Serum, PBMC's, or tissue should be assayed at 
    periodic intervals post transplantation for xenogeneic agents known to 
    be present in the transplanted tissue. Active surveillance should 
    include more frequent screening in the immediate posttransplant period 
    (e.g., at 2, 4, and 6 weeks after transplantation) with subsequently 
    decreasing frequency in the absence of clinical indication. Assays 
    intended for the generic detection of unknown agents may also be 
    appropriate. Assays should be used to detect classes of viruses known 
    to establish persistent latent infections in the absence of clinical 
    symptoms (e.g., herpesviruses and retroviruses) (section 3.3.1.1.). 
    When the xenogeneic viruses of concern have similar human counterparts, 
    e.g., simian CMV, assays to distinguish between the two should be 
    employed. Depending upon the degree of immunosuppression in the 
    recipient, serological assays may be or may not be useful. Methods for 
    analysis include cocultivation of cells coupled with appropriate 
    detection assays. The sensitivity, specificity, and validity of the 
    testing methods should be predetermined and documented under conditions 
    simulating those employed in the xenotransplant procedure.
        4.1.1.6. In response to a potential xenogeneic infection related to 
    a clinical episode, posttransplantation testing of archived biologic 
    specimens should be conducted in association with an epidemiologic 
    investigation to assess potential public health significance of the 
    infection. This investigation should proceed under the direction of 
    appropriate health authorities following prompt notification of the 
    State health department, CDC, and FDA.
    
    4.2. Contacts of Recipient
    
        The clinical protocol should outline a procedure to inform the 
    recipient of the responsibility to educate his/her close contacts 
    regarding the possibility of the emergence of xenogeneic infections 
    from the source animal species and to offer the recipient assistance 
    with this education process, if desired. Education of close contacts 
    should address the uncertainty regarding the risks of xenogeneic 
    infections, information about behaviors known to transmit infectious 
    agents from human to human (i.e., unprotected sex, intravenous drug use 
    with shared needles and other activities that involve potential 
    exchange of blood or other body fluids) and methods to minimize the 
    risk of transmission. Recipients should educate their close contacts 
    about the need to inform their physician and the research coordinator 
    at the institution where the xenotransplantation was performed of any 
    significant unexplained illnesses in themselves or their close 
    contacts.
    
    4.3. Hospital Infection Control
    
    4.3.1. Infection Control Practices
        4.3.1.1. Standard precautions should be used for the care of all 
    patients, including appropriate handwashing, use of barrier 
    precautions, and care in the use and disposal of needles and other 
    sharp instruments. Strict adherence to these recommended procedures 
    will reduce the risk of transmission of xenogeneic infections and other 
    blood-borne and nosocomial pathogens.
        4.3.1.2. Additional infection control or isolation precautions 
    (e.g., airborne, droplet, contact) should be employed as indicated in 
    the judgment of the hospital epidemiologist and the xenotransplant team 
    infectious disease specialist. For example, appropriate isolation 
    precautions for each hospitalized transplant recipient will depend upon 
    the xenotransplant, the extent of immunosuppression, and the clinical 
    condition of the recipient. The appropriateness of infection control 
    measures should be considered at the time of transplant and reevaluated 
    during each readmission. Isolation precautions should be continued 
    until a suspected xenogeneic infection has been proven and resolved or 
    has been effectively ruled out in the recipient.
        4.3.1.3. Xenotransplant teams should adhere to recommended 
    procedures for handling and disinfection/sterilization of medical 
    instruments and disposal of infectious waste.
        4.3.2. Acute Infectious Episodes. Most acute viral infectious 
    episodes among the general population are never etiologically 
    identified. Xenograft recipients remain at risk for these infections 
    and other infections common among immunosuppressed allograft 
    recipients. When the source of a significant illness in a recipient 
    remains unidentified despite standard diagnostic procedures, more 
    testing of body fluid and tissue samples may be appropriate. The 
    infectious disease specialist, in consultation with the hospital 
    epidemiologist, the veterinarian, the clinical microbiologist and other 
    members of the xenotransplant team should assess each clinical episode 
    and make a considered judgment regarding the need and type of 
    diagnostic testing and appropriate infection control precautions. 
    Experts on infectious diseases and public health may also need to be 
    consulted.
        4.3.2.1. Immunosuppressed transplant patients may be unable to 
    mount a sufficient immunological response for serological assays to 
    detect infections reliably. In this setting, appropriate validated 
    culture systems, genomic detection methodologies and other
    
    [[Page 49928]]
    
    techniques may detect diseases for which serologic testing is 
    inadequate. Consequently, clinical centers where xenotransplantation is 
    performed should have the capability to culture and to identify viral 
    agents using in vitro and in vivo methodologies. Specimens should be 
    handled to ensure their viability and to maximize the probability of 
    isolation and identification of fastidious agents. Algorithms for 
    evaluation of unknown xenogeneic pathogens should be developed in 
    consultation with appropriate experts, including persons with expertise 
    in both medical and veterinary infectious diseases, laboratory 
    identification of unknown infectious agents and the management of 
    biosafety issues associated with such investigations.
        4.3.2.2. Archiving of acute and convalescent sera obtained in 
    association with acute unexplained illnesses should be performed when 
    appropriate as judged by the infectious disease physician and/or the 
    hospital epidemiologist. This would permit retrospective study and 
    perhaps an etiologic diagnosis of the clinical episode.
        4.3.3. Health Care Workers. A comprehensive occupational health 
    services program should be designed to educate workers regarding the 
    risks associated with xenotransplantation and to monitor for possible 
    infections in workers. Health care workers, including laboratory 
    personnel, who handle the animal tissues/organs prior to 
    transplantation will have a definable risk of infection not exceeding 
    that of animal care, veterinary, or abattoir workers routinely exposed 
    to the source animal species provided equivalent biosafety standards 
    are employed. However, the risk to health care workers who provide 
    direct/indirect post-transplantation care for xenograft recipients is 
    undefined. Decisions regarding work restrictions or assignments for 
    immunocompromised workers should be determined by each institution. The 
    occupational health services program should include the following:
        4.3.3.1. Education of Health Care Workers. All centers where 
    xenotransplantation procedures are performed should develop appropriate 
    educational materials for their staff tailored to each procedure. These 
    materials should describe the xenotransplant procedure(s), and the 
    known and potential risks of xenogeneic infections posed by the 
    procedure(s). Those research or health care activities that are 
    considered to be associated with the greatest risk of infection should 
    be emphasized in order to minimize exposure and transmission of both 
    zoonotic and nosocomial agents between the recipient and the health 
    care workers. The use of Standard Precautions should be reviewed. 
    Education programs should detail the circumstances for use of personal 
    protective equipment (e.g., gloves, gowns, masks, etc.) and the 
    importance of handwashing before and after all patient contacts, even 
    if gloves are worn. The potential for transmission of these agents to 
    the general public should be discussed.
        4.3.3.2. Worker Surveillance. Protocols should be developed for the 
    collection and archiving of baseline sera (i.e., prior to exposure to 
    xenografts or recipients) from health care workers either on the 
    xenotransplant team or caring for xenograft recipients and any 
    laboratory personnel who may handle the animal cells, tissues, and 
    organs or future biologic specimens from transplant recipients. 
    Archived sera serve as a baseline specimen for comparing sera collected 
    following nosocomial exposures. In addition, these protocols should 
    describe methods of recording, storing, and retrieving information 
    related to health care workers and specific nosocomial exposures. The 
    activities of the Occupational Health Service should be coordinated 
    with the Infection Control Program to ensure appropriate surveillance 
    of infections in personnel.
        4.3.3.3. Postexposure Evaluation and Management. Written protocols 
    should be in place for the evaluation of health care workers who 
    experience an exposure where there is a risk of transmission of an 
    infectious agent, e.g., an accidental needlestick. Health care workers, 
    including laboratory personnel, should be instructed to report 
    exposures immediately to the Occupational Health Service. The 
    postexposure protocol should describe the information to be recorded 
    including the date and nature of exposure, the xenotransplantation 
    procedure, recipient information, actions taken as a result of such 
    exposures (e.g., counseling, postexposure management and followup) and 
    the outcome of the event. This information should be archived in a 
    Health Exposure Log (section 4.4) and maintained indefinitely at the 
    xenotransplantation center despite any change in employment of the 
    health care worker or discontinuation of xenotransplantation procedures 
    at that center. Health care and laboratory workers should be counseled 
    to report and seek medical evaluation for unexplained clinical 
    illnesses occurring after the exposure.
    
    4.4. Health Care Records
    
        Each clinical xenotransplantation center should maintain 
    indefinitely the three cross-referenced record systems: (1) An 
    Institutional Xenotransplantation Record which documents for all 
    xenotransplant procedures: The principal investigator, the individual 
    source animal and its procurement facility, the date and type of 
    procedure, the xenograft tissue recipient and a summary of the 
    recipient's clinical course, close contacts, and the health care 
    workers associated with each procedure; (2) a Xenotransplantation 
    Nosocomial Health Exposure Log which documents the dates, involved 
    persons, and nature of all nosocomial exposures which are associated 
    with a xenotranplantation protocol and which potentially pose risk of 
    transmission of xenogeneic infections; (3) individual xenotransplant 
    recipient health records which document comprehensively each patient's 
    clinical course, the results of post-transplant surveillance studies 
    (section 4.1), and contain a summary of both the health status report 
    and the results of the screening assays performed on source animal(s) 
    from which the xenograft was obtained.
        These records should be current and accurately cross-referenced. 
    This systematic data maintenance will facilitate epidemiologic 
    investigation of adverse events. In the future, these data should be 
    linked to any national registry (section 5.1) to facilitate recognition 
    of rates of occurrence and clustering of adverse health events, 
    including events that may represent the outcomes of xenogeneic 
    infections and mortality patterns, and linkage of those events to 
    specific exposures on a national level.
    
    5. Public Health Needs
    
    5.1. National Registry
    
        The public health interest would best be served by the 
    establishment of a national registry. A national registry would enable 
    rapid identification of epidemiologically significant common features 
    among xenograft recipients and provide a data base for the assessment 
    of long-term safety. Such a data base would make possible the rapid 
    recognition of rates of occurrence and clustering of health events that 
    may represent outcomes of xenogeneic infections; allow the accurate 
    linkage of these events to exposures on a national level; facilitate 
    notification of individuals and clinical centers regarding 
    epidemiologically significant adverse events associated with
    
    [[Page 49929]]
    
    xenotransplantation; and enable biological and clinical research 
    assessments. Information derived from the registry should be reasonably 
    available to the public with appropriate confidentiality protection for 
    any patient identifying information and/or proprietary information.
    
    5.2  Serum and Tissue Archives
    
        Samples of sera, plasma, leukocytes, and tissue of the source 
    animal and recipient should be archived for public health investigation 
    purposes as discussed in sections 3.7 and 4.1. Source animal and 
    xenograft recipient specimens should be kept at individual centers 
    under storage conditions outlined in section 4.1.1.4. Information about 
    the location and nature of archived specimens associated with each 
    transplant should be documented in the health care records and 
    delineated in sections 3.7 and 4.4, and ultimately in any national 
    registry that is established.
    
    6. Bibliography
    
        The following references have been placed on display in the Dockets 
    Management Branch (address above) and may be seen by interested persons 
    between 9 a.m. and 4 p.m., Monday through Friday. References 1 through 
    5 may also be obtained from FDA/CBER/Office of Communication, Training 
    and Manufacturers Assistance via FAX by calling 1-800-835-4709 or via 
    mail by calling 301-827-1800. References 21 through 24 may also be 
    obtained from The National Technical Information Service (NTIS), 5285 
    Port Royal Rd., Springfield, VA 22161, 703-487-4650.
    
    A. Federal Laws
    
        1. The Public Health Service Act (42 U.S.C. 262, 264).
        2. The Federal Food, Drug, and Cosmetic Act (21 U.S.C. 301 et 
    seq.).
        3. The Social Security Act (42 U.S.C. 1320b-8).
        4. The National Organ Transplant Act (42 U.S.C. 273 et seq.).
        5. The Animal Welfare Act (7 U.S.C. 2132).
    
    B. Federal Regulations
    
        1. Title 21 of the Code of Federal Regulations (CFR) parts 50, 
    56, 312, and 812.
        2. Title 45 of the CFR part 46.
    
    C. Federal Guidance
    
        1. Points to Consider in Somatic Cell and Gene Therapy, 56 FR 
    61022, November 29, 1991.
        2. Points to Consider in the Characterization of Cell Lines Used 
    to Produce Biologicals, 58 FR 42974, August 12, 1993.
        3. Application of Current Statutory Authorities to Human Somatic 
    Cell Therapy Products and Gene Therapy Products, 58 FR 53248, 
    October 14, 1993.
        4. Bovine Derived Materials; Agency Letters to Manufacturers of 
    FDA Regulated Products, 59 FR 44591, August 29, 1994.
        5. Points to Consider in the Manufacture and Testing of 
    Therapeutic Products for Human Use Derived from Transgenic Animals, 
    60 FR 44036, August 24, 1995.
        6. ``Guidelines for Prevention of Herpesvirus Simiae (B Virus) 
    Infection in Monkey Handlers,'' Mortality and Morbidity Weekly 
    Report, Centers for Disease Control and Prevention (CDC), Department 
    of Health and Human Services (DHHS), Vol. 36, pp. 680-682 and 687-
    689, 1987.
        7. ``Guidelines to Prevent Simian Immunodeficiency Virus 
    Infection in Laboratory Workers and Animal Handlers,'' Mortality and 
    Morbidity Weekly Report, CDC, DHHS, Vol. 37, pp. 693-694 and 699-
    700, 1988.
        8. ``Guidelines for Investigating Clusters of Health Events,'' 
    Mortality and Morbidity Weekly Report, CDC, DHHS, Vol. 39, pp. 39, 
    RR-11, 1990.
        9. Biosafety in Microbiological and Biomedical Laboratories, 
    DHHS, PHS, CDC, the National Institutes of Health (NIH), 3d Ed., HHS 
    Publication No. (CDC) 93-8395, May 1993.
        10. The NIH Guidelines for Recombinant DNA Research, 61 FR 1482, 
    January 19, 1995.
        11. ``Guideline for Isolation Precautions in Hospitals,'' DHHS, 
    CDC, Infection Control and Hospital Epidemiology, Vol. 17, pp. 53-
    80, 1996.
        12. ``Acquired Immune Deficiency Syndrome (AIDS): Precautions 
    For Clinical and Laboratory Staffs,'' Mortality and Morbidity Weekly 
    Report, CDC, DHHS, Vol. 31, pp. 577-580, 1982.
        13. ``Acquired Immunodeficiency Syndrome (AIDS): Precautions for 
    Health-Care Workers and Allied Professionals,'' Mortality and 
    Morbidity Weekly Report, CDC, DHHS, Vol. 32, pp. 450-451, 1983.
        14. ``Update: Acquired Immunodeficiency Syndrome and Human 
    Immunodeficiency Virus Infection Among Health-Care Workers,'' 
    Mortality and Morbidity Weekly Report, CDC, DHHS, Vol. 37, pp. 229-
    234, 1988.
        15. ``Notice to Readers NIOSH Guidelines for Protecting the 
    Safety and Health of Health-Care Workers,'' Mortality and Morbidity 
    Weekly Report, CDC, DHHS, Vol. 39, p. 417, 1990.
        16. ``Occupationally Acquired Human Immunodeficiency Virus 
    Infections in Laboratories Producing Virus Concentrates in Large 
    Quantities: Conclusions and Recommendations of an Expert Team 
    Convened by the Director of the National Institutes of Health,'' 
    Mortality and Morbidity Weekly Report, CDC, DHHS, Vol. 37 (S-4), pp. 
    19-22, 1988.
        17. ``Recommendations for Prevention of HIV Transmission in 
    Health-Care Settings,'' Mortality and Morbidity Weekly Report, CDC, 
    DHHS, Vol. 36 (S002), p. 001, 1987.
        18. ``Update: Universal Precautions for Prevention of 
    Transmission of HIV, Hepatitis B Virus, and Other Blood Borne 
    Pathogens in Health-Care Settings,'' Mortality and Morbidity Weekly 
    Report, CDC, DHHS, Vol. 37, pp. 377-388, 1988.
        19. ``Guidelines for Prevention of Transmission of Human 
    Immunodeficiency Virus and Hepatitis B Virus to Health-Care Workers 
    and Public-Safety Workers,'' Mortality and Morbidity Weekly Report, 
    CDC, DHHS, Vol. 38, No. (S-6), 1989.
        20. ``Rethinking the Role of Isolation Precautions in the 
    Prevention of Nosocomial Infections,'' Annals of Internal Medicine, 
    Vol. 107, pp. 243-246, 1987.
        21. ``Implementing and Evaluating a System of Generic Infection 
    Precautions: Body Substance Isolation, ``American Journal of 
    Infection Control, Vol. 18, pp. 1-12, 1990.
        22. ``The Guideline for Isolation Precautions in Hospital'' 
    NTIS, PB85-923401, 1983.
        23. ``Guideline for Infection Control in Hospital Personnel'' 
    NTIS, PB85-923402, 1983.
        24. ``Guideline for Hand Washing and Hospital Environmental 
    Control,'' NTIS, PB85-923404, 1985.
    
    D. Scientific Articles and Other Reports
    
        1. Allan, J. S., ``Xenograft Transplantation and the Infectious 
    Disease Conundrum,'' Institute of Laboratory Animal Resources 
    Journal, Vol. 37, pp. 37-48, 1985.
        2. Chapman, L. E., T. M. Folks, D. R. Salomon, et al., 
    ``Xenotransplantation and Xenogeneic Infections,'' New England 
    Journal of Medicine, Vol. 333, pp. 1498-1501, 1995.
        3. Chapman L. E., and J. A. Fishman, ``Xenotransplantation and 
    Infectious Diseases,'' In Xenotransplantation, 2d Ed., Cooper, D. K. 
    C. (editor), in press.
        4. Chari, R. S., B. H. Collins, J. C. Magee, et al., ``Brief 
    Report: Treatment of Hepatic Failure With Ex Vivo Pig-Liver 
    Perfusion Followed by Liver Transplantation,'' New England Journal 
    of Medicine, Vol. 331, pp. 234-237, 1994.
        5. Cooper, D. K. C., E. Kemp, K. Reemstma, D. J. G. White 
    (editors), ``Xeno-Transplantation, The Transplantation of Organs and 
    Tissues Between Species,'' Berlin: Springer-Verlag, 1991.
        6. Dunning, J. J., D. J. G. White, and J. Wallwork, ``The 
    Rationale for Xenotransplantation as a Solution to the Donor Organ 
    Shortage,'' Path Biol., Vol. 42, p. 231, 1994.
        7. Eastlund T., ``Infectious Disease Transmission Through Cell, 
    Tissue and Organ Transplantation: Reducing the Risk Through Donor 
    Selection,'' Cell Transplantation, Vol. 4, pp. 455-477, 1995.
        8. Farwell, J. R., G. J. Dohrmann, L. D. Marrett, and J. W. 
    Meigs, ``Effect of SV40 Virus-Contaminated Polio Vaccine on the 
    Incidence and Type of CNS Neoplasms in Children: A Population-Based 
    Study,'' Transactions of the American Neurological Association, Vol. 
    104, pp. 261-264, 1979.
        9. Geissler E., and W. Staneczek, ``SV40 and Human Brain Tumors, 
    Archive for Geschwulstforsch, Vol. 58, pp. 129-134, 1988.
        10. Ho, M., ``Virus Infections After Transplantation in Man,'' 
    Archives of Virology, No. 55, pp. 1-24, 1977.
        11. ``Xenotransplantation: Science, Ethics, and Public Policy,'' 
    Institute of Medicine, Washington, D.C., National Academy Press, 
    1996.
        12. Kalter, S. S., and R. L. Heberling, ``Xenotransplantation 
    and Infectious
    
    [[Page 49930]]
    
    Diseases,'' Institute of Laboratory Animal Resources Journal, Vol. 
    37, pp. 31-37, 1995.
        13. Mortimer, E. A., M. L. Lepow, E. Gold, et al., ``Long-term 
    Follow-up of Persons Inadvertently Inoculated With SV40 as 
    Neonates,'' New England Journal of Medicine, Vol. 305, pp. 1517-
    1518, 1981.
        14. Myers, G., and G. N. Pavlakis, ``Evolutionary Potential of 
    Complex Retroviruses,'' In The Retroviridae,  J. A. Levy (editor), 
    Vol. 1, pp. 57-58, New York: Plenum Press, 1992.
        15. Nalesnik, M. A., and T. Starzl, Epstein-Barr Virus, 
    Infectious Mononucleosis, and Post-Transplant Lymphoproliferative 
    Disorders [Review], Transplantation Science, Vol. 4, pp. 61-79, 
    1994.
        16. ``Animal-to-Human Transplants: The Ethics of 
    Xenotransplantation,'' Nuffield Council on Bioethics: London, 1996.
        17. Shah, K., and N. Nathanson, ``Human Exposure to SV40: Review 
    and Comment, American Journal of Epidemiology, No. 103, pp. 1-12, 
    1976.
        18. Simonds, R. J., HIV Transmission by Organ and Tissue 
    Transplantation [Review], Acquired Immune Deficiency Syndromes, Vol. 
    7, Suppl. 2, pp. S35-38, 1993.
        19. Stevens, J. G., ``Overview of Herpesvirus Latency,'' 
    Seminars in Virology, No. 5, pp. 191-196, 1994.
        20. Weiss, R. A., ``Foamy Retroviruses. A Virus in Search of a 
    Disease,'' Nature, No. 333, pp. 497-498, 1988.
        21. Wick, G., T. Klemens, A. Aguzzi, et al., ``Possible Role of 
    Human Foamy Virus in Grave's Disease,'' Intervirolology, No. 35, pp. 
    101-107, 1993.
    
    E. Animal Sources for Xenotransplants
    
        1. Brack, M., Agents Transmissible to Man, Berlin: Springer-
    Velag, 1987.
        2. Fox, J. G., and N. S. Lipman, Infections Transmitted by Large 
    and Small Laboratory Animals [Review], Infectious Disease Clinics of 
    North America, Vol. 5, No. 1, pp. 131-163, March 1991.
        3. Glaser, C. A., F. J. Angulo, J. A. Rooney, Animal-Associated 
    Opportunistic Infections Among Persons Infected With the Human 
    Immunodeficiency Virus [Reviews], Clinical Infectious Diseases, Vol. 
    18, No. 1, pp. 14-24, January, 1994.
        4. Miller, C.D., J.R. Songer, J.F. Sullivan, ``A Twenty-five 
    Year Review of Laboratory-Acquired Human Infections at the National 
    Animal Disease Center,'' American Industrial Hygiene Association 
    Journal, Vol. 48, No. 3, pp. 271-275, March 1987.
        5. Prusiner, S.B., ``Prion Encephalopathies of Animals and 
    Humans,'' In Transmissible Spongioform Encephalopathies--Impact on 
    Animal and Human Health, F. Brown (editor), Developments in 
    Biologics Standardization Basel, Karger, Vol. 80, pp. 31-44, 1993.
        6. Weinberg, A.N., Ecology and Epidemiology of Zoonotic 
    Pathogens, [Review], Infectious Diseases Clinics of North America, 
    Vol. 5910, pp. 1-6, March [year].
        7. Murphy, G.P., H.D. Brede, E. Cohen, J.T. Grace Jr., ``The 
    Cape Western Baboon in Organ Allotransplantation,'' Trans. Proc. 
    1970, Vol. 2, pp. 546-549, 1970.
        8. Reemstma, K., ``Renal Heterotransplantation from Nonhuman 
    Primates to Man,'' Ann N Y Acad Sci, No. 162, pp. 412-418, 1969.
        9. Starzl, T.H.E., J. Fung, A. Tzakis, et al., ``Baboon-to-Human 
    Liver Transplantation,'' Lancet, Vol. 341, pp. 65-71, 1993.
        10. Starzl, T.H.E., T.L. Marchioro, G.N. Peters, et al., ``Renal 
    Heterotransplantation from Baboon to Man: Experience With 6 Cases,'' 
    Transplantation, Vol. 2, pp. 752-776, 1964.
        11. Benirschke, K., ``Primates: The Road to Self-sustaining 
    Populations,'' New York: Springer-Verlag, 1986.
        12. Goodwin, W.J., and A.M. Coehlo Jr., ``Development of a Large 
    Scale Baboon Breeding Program,'' Laboratory Animal Science, Vol. 32, 
    pp. 672-676, 1982.
        13. Lerche, N.W., J.L. Yee, and M.B. Jennings, ``Establishing 
    Specific Retrovirus-free Breeding Colonies of Macaques: An Approach 
    to Primary Screening and Surveillance,'' Laboratory Animal Science, 
    Vol 44, pp. 217-221, 1994.
        14. Ward, J.A., and J.K. Hilliard, ``B Virus-Specific Pathogen-
    Free (SPF) Breeding Colonies of Macaques: Issues, Surveillance, and 
    Results in 1992,'' Laboratory Animal Science, No. 44, pp. 222-228, 
    1994.
        15. Allan, J.S., Primates and New Viruses, (Letter) Science 
    Letters, No. 265, pp. 1345-1346, 1994.
        16. Barahona, H., L.V. Melendez, and J.L. Melnick, ``A 
    Compendium of Herpesviruses Isolated From Non-human Primates,'' 
    Intervirology, Vol. 3, pp. 175-192, 1974.
        17. Allan, J.S., P. Ray, S. Broussard, E. Whitehead, et al., 
    ``Infection of Baboons with Simian/Human Immunodeficiency Viruses,'' 
    Journal of Acquired Immune Deficiency Syndromes and Human 
    Retrovirology, in press, 1995.
        18. Barin, F., S.M. Boup, F. Denis, et al., ``Serological 
    Evidence for a Virus Related to Simian T-lymphotropic Retrovirus III 
    in Residents of West Africa,'' Lancet, II: 1387-1389, 1985.
        19. Bieniasz, P.D., A. Rethwilm, R. Pitman, M.D. Daniel, I. 
    Chrystie, M.O. McClure, ``A Comparative Study of Higher Primate 
    Foamy Viruses, Including a New Virus from a Gorilla,'' Virology, No. 
    207, pp. 217-228, 1995.
        20. Brede, H.D., and G.P. Murphy, ``Bacteriologic and Virologic 
    Considerations in Primate Transplants,'' Primates in Medicine, Vol. 
    7, pp. 18-28, 1972.
        21. Castro, B.A., M. Nepomuceno, N.W. Lerche, et al., 
    ``Persistent Infection of Baboons and Rhesus Monkeys with Different 
    Strains of HIV-2,'' Virology, Vol. 184, pp. 219-226, 1991.
        22. ``Anonymous Survey for Simian Immunodeficiency Virus 
    Infection in Laboratory Workers and Animal Handlers,'' Mortality and 
    Morbidity Weekly Report, No. 41, pp. 814-815, 1992.
        23. Chakrabarti, L., M. Guyader, M. Alizon, et al., ``Sequence 
    of Simian Immunodeficiency Virus from Macaque and its Relationship 
    to Other Human and Simian Retroviruses,'' Nature, Vol. 328, pp. 543-
    547, 1987.
        24. Courgnaud, V., F. Laure, P.N. Flutz, et al., ``Genetic 
    Differences Accounting for Evolution and Pathogenicity of Simian 
    Immunodeficiency Virus from a Sooty Mangabey Monkey After Cross-
    species Transmission to a Pig-tailed Macaque,'' Journal of Virology, 
    Vol. 66, pp. 414-419, 1992.
        25. Dalgard, D.E., R.J. Hardy, S.L. Pearson, et al., ``Combined 
    Simian Hemorrhagic Fever and Ebola Virus Infection in Cynomolgus 
    Monkeys,'' Laboratory Animal Science, No. 42, pp. 152-157, 1992.
        26. Daniel, M.D., N.L. Letvin, N.W. King, et al., ``Isolation of 
    a T-cell Tropic HTLV-III-like Retrovirus from Macaques,'' Science, 
    Vol. 228, pp. 1201-1204, 1985.
        27. Daniel, M.D., N.L. Letvin, P.K. Sehgal, et al., ``Prevalence 
    of Antibodies to Three Retroviruses in a Captive Colony of Macaque 
    Monkeys,'' International Journal of Cancer, No. 41, pp. 601-608, 
    1988.
        28. Deinhardt, F. ``Biology of Primate Retroviruses,'' In Viral 
    Oncology, G. Klein (editor), New York: Raven Press, pp. 357-398, 
    1980.
        29. Desrosiers, R.C., ``The Simian Immunodeficiency Viruses,'' 
    Annual Reviews of Immunology, No. 8, pp. 557-578, 1990.
        30. Doolittle, R.F., ``The Simian-Human Connection,'' Nature, 
    No. 339, pp. 338-339, 1989.
        31. Fenner, F., Human Monkey Pox: A Newly Discovered Human Virus 
    Disease, In Emerging Viruses, S.S. Morse (editor), New York: Oxford 
    University Press, pp. 176-183, 1993.
        32. Flugel, R.M., ``Spumaviruses: A Group of Complex 
    Retroviruses,'' Journal of Acquired Immune Deficiency Syndromes, No. 
    4, pp. 739-750, 1991.
        33. Franchini, G., and M.L. Bosch, ``Genetic Relatedness of the 
    Human Immunodeficiency Viruses Type 1 and 2 (HIV-1, HIV-2) and the 
    Simian Immunodeficiency Viruses (SIV), Annals New York Academy of 
    Sciences, Vol. 554, pp. 81-87, 1989.
        34. Fultz, P.N., Simian T-lymphotropic Virus Type I. In The 
    Retroviridae, Vol. 3 of The Viruses Series, J.A. Levy (editor), New 
    York: Plenum Press, pp. 111-131, 1994.
        35. Fultz, P.N., H.M. McClure, D.C. Anderson, ``Isolation of a 
    T-lymphotropic Retrovirus From a Naturally Infected Sooty Mangabeys 
    (Cercocebus atys.),'' Proceedings of the National Academy of 
    Science, Vol. 83, pp. 5286-5290, 1986.
        36. Gao, F., Y. Ling, and A.T. White, et al., ``Human Infection 
    by Genetically Diverse SIVsm-related HIV-2 in West Africa,'' Nature, 
    Vol. 358, pp. 495-499, 1992.
        37. Hilliard, J. K., D. Black, R. Eberle, ``Simian Alphaviruses 
    and Their Relation to the Human Herpes Simplex Viruses,'' Archives 
    of Virology, No. 109, pp. 83-102, 1989.
        38. Hillis, W. D. ``An Outbreak of Infectious Hepatitis Among 
    Chimpanzee Handlers at a United States Air Force Base,'' American 
    Journal Hygiene, Vol. 73, pp. 316-328, 1961.
        39. Holmes, G. P., L. E. Chapman, J. A. Stewart, et al., ``and 
    the B Virus Working Groups for the prevention and treatment of B-
    virus infections in exposed persons,'' Clinical Infectious Diseases, 
    Vol. 20, pp. 421-439, 1995.
        40. Homma, T., P. J. Kanki, N. W. Kin, R. D. Hunt, M. J. 
    O'Connell, et al., ``Lymphoma
    
    [[Page 49931]]
    
    in Macques: Association With Virus of Human T-Lymphotropic Family,'' 
    Science, Vol. 225, pp. 716-718, 1984.
        41. Hooks, J. J., and B. Detrick-Hooks, ``Simian Foamy Virus-
    induced Immunosuppression in Rabbits,'' Journal of General Virology, 
    Vol. 44, pp. 383-390, 1979.
        42. Huang, S. A., J. Silberman, H. Rothschild, and J. G. Cohen, 
    ``Replication of Baboon Endogenous Virus in Human Cells,'' Journal 
    of Biological Chemistry, Vol. 264, pp. 8811-8814, 1989.
        43. Hubbard, G. B., J. P. Mone, ``Spontaneously Generated Non-
    Hodgkin's Lymphoma in Twenty-seven Simian T-cell Leukemia Virus Type 
    I Antibody-positive Baboons (Papio species),'' Laboratory Animal 
    Science, Vol. 43, pp. 301-309, 1993.
        44. Hubbard, G. B., K. F. Soike, T. M. Butler, et al., ``An 
    Encenphalomyocarditis Virus Epizootic in a Baboon Colony, Laboratory 
    Animal Science, Vol. 42, pp. 233-239, 1992.
        45. Hull, R. N., ``The Simian Viruses,'' Virology Monographs, 
    Vol. 2, pp. 1-66, 1968.
        46. Human, P., F. van der Riet de St. J., D. K. Cooper, S. S. 
    Kalter, J. F. Fincham, et al., ``The Virological Evaluation of 
    Nonhuman Primates for Xenotransplantation,'' Transplantation, No. 
    19, pp. 146-150, 1987.
         47. Hunt, R. D., L. V. Melendez, ``Herpes Virus Infections in 
    Nonhuman Primates: A Review,'' Laboratory Animal Care, Vol. 19, pp. 
    221-234, 1969.
        48. Jin, M. J., J. Rogers, J. E. Phllips-Conroy, et al., 
    ``Infection of a Yellow Baboon with Simian Immunodeficiency Virus 
    from African Green Monkeys: Evidence for Cross-species Transmission 
    in the Wild,'' Journal of Virology, Vol. 68, pp. 8454-8460, 1994.
        49. Johnson, D. R., G. Klein, L. Falk, ``Interaction of 
    Herpesvirus Ateles and Herpesvirus Saimiri With Primate 
    Lymphocytes,'' Intervirology, Vol, 13, pp. 21-27, 1980.
        50. Kalter, S. S., ``Overview of Simian Viruses and Recognized 
    Virus Diseases and Laboratory Support for the Diagnosis of Viral 
    Infections'' In ``Primates: The Road to Self-Sustaining 
    Populations,'' K. Benirschke (editor), pp. 681-709, New York: 
    Springer-Verlag, 1986.
        51. Kalter, S. S., ``The Nonhuman Primate as Potential Organ 
    Donor for Man: Virological Considerations,'' In 
    ``Xenotransplantation, the Transplantation of Organs and Tissues 
    Between Species,'' D. K. C. Cooper, E. Kemp, and D. J. G. White 
    (editors), pp. 457-479, Berlin: Springer-Verlag, 1991.
        52. Kalter, S. S., and R. L. Heberling, ``Primate Viral Diseases 
    in Perspective,'' Journal of Medical Primatology, Vol. 19, pp. 519-
    535, 1990.
        53. Kalter, S. S., J. Ratner, G. V. Kalter, et al., ``A Survey 
    of Primate Sera for Antibodies of Human and Simian Origin,'' Amer J 
    Epidem, No. 86, pp. 552-568, 1967.
        54. Kanki, P. J., M. F. McLane, N. W. King, N. L. Letvin, R. D. 
    Hunt, et al., ``Serologic Identification and Characterization of a 
    Macaque T-lymphotropic Retrovirus Closely Related to HTLV-III,'' 
    Science, No. 228, pp. 1199-1201, 1985.
        55. Khabbaz, R. F., W. Heneine, J. R. George, et al., ``Brief 
    Report: Infection of a Laboratory Worker with Simian 
    Immunodeficiency Virus,'' New England Journal of Medicine, Vol. 330, 
    pp. 172-177, 1994.
        56. Lecatsas, G., F. A. Neethling, W. A. De Klerk, B. Bridelli, 
    ``Filovirus Seropositivity in Prospective Organ Donor Baboons,'' 
    Transplanation Proceedings, No. 24, pp. 617-618, 1992.
        57. Levin, J. L., J. K. Hilliard, S. L. Lipper, T. M. Butler, W. 
    J. Goodwin, ``A Naturally Occurring Epizootic of Simian Agent 8 in 
    the Baboon,'' Laboratory Animal Science, Vol. 38, pp. 394-397, 1988.
        58. Martini, G. A., Marburg Agent Disease: in Man, Transactions 
    of the Royal Society of Tropical Medicine and Hygiene, Vol. 63, pp. 
    295-302, 1969.
        59. McClure, M. O., P. D. Bieniasz, T. F. Schulz, I. L. 
    Chrystie, G. Simpson, A. Aguzzi, J. G. Hoad, et al., ``Isolation of 
    a New Foamy Retrovirus from Orangutans,'' Journal of Virology, No. 
    68, pp. 7124-7130, 1994.
        60. McClure, H.M., A. R. Brodie, D. C. Anderson, and R. B. 
    Swenson, ``Bacterial Infections of Non-human Primates,'' In 
    ``Primates: The Road to Self-Sustaining Populations,'' K. Benirschke 
    (editor), New York: Springer-Verlag, pp. 531-556, 1986.
        61. Michaels, M. G., J. P. McMichael, K. Brasky, S. Kalter, et 
    al., ``Screening Donors for Xenotransplantation: The Potential for 
    Xenozoonoses,'' Transplantation, No. 57, pp. 1462-1465, 1994.
        62. Michaels, M. G., R. L. Simmons, ``Xenotransplant Associated 
    Zoonoses: Strategies for Prevention,'' Transplantation, No. 57, pp. 
    1-7, 1994.
        63. Migaki, G., ``Mycotic Infections in Nonhuman Primates,'' In 
    ``Primates: The Road to Self-sustaining Populations,'' K. Benirschke 
    (editor), New York: Springer-Verlag, pp. 557-570, 1986.
        64. Mone, J. P., E. W. Whitehead, M. M. Leland, et al., ``Simian 
    T-cell Leukemia Virus Type I Infection in Captive Baboons,'' 
    Acquired Immune Deficiency Syndromes and Research in Human 
    Retrovirology, Vol. 8, pp. 1653-1661, 1992.
        65. Murphey-Corb, M., L. N. Martin, S. R. Rangan, G. Baskin, B. 
    J. Gormus, R. H. Wolf, W. A. Andes, ``Isolation of an HTLV-III-
    related Retrovirus from Macaques with Simian Aids and its Possible 
    Origin in Asymptomatic Mangabeys,'' Nature, Vol. 321, pp. 435-437, 
    1986.
        66. Ohta, Y., T. Masuda, H. Tsujimoto, et al., ``Isolation of 
    Simian Immunodeficiency Virus from African Green Monkeys and 
    Seroepidemiologic Survey of the Virus in Various Nonhuman 
    Primates,'' International Journal of Cancer, Vol. 41, pp. 115-122, 
    1988.
        67. Peeters, M., K. Fransen, E. Delaporte, M. Van den 
    Haesevelde, et al., ``Isolation and Characterization of a New 
    Chimpanzee Lentivirus (Simian Immunodeficiency Virus Isolate Cpz-
    ant) from a Wild-Caught Chimpanzee,'' Acquired Immune Deficiency 
    Syndromes, Vol. 6, pp. 447-451, 1992.
        68. Peters, C. J., A. Sanchez, H. Feldman, P. E. Rollin, 
    ``Filoviruses as Emerging Pathogens,'' Seminars in Virology, Vol. 5, 
    pp. 147-154, 1994.
        69. Ricordi, C., A. G. Rzakis, W. B. Rybka, P. Fontes, E. D. 
    Ball, et al., ``Xenotransplantation of Hematopoietic Cells Resistant 
    to HIV as a Potential Treatment for Patients with AIDS,'' 
    Transplantation Proceedings, Vol. 26, No. 3, pp. 1302-1303, 1994.
        70. Smetana, H. F., A. D. Felsenfeld, and A. J. Riopelle, 
    ``Human Viral Hepatitis and Chimpanzees,'' The Chimpanzee, Vol. 3, 
    pp. 26-55, Karger, Basel, 1970.
        71. Toft II, J. D., ``The Pathoparasitology of Nonhuman 
    Primates: A Review,'' In ``Primates: The Road to Self-sustaining 
    Populations,'' K. Benirschke (editor), New York: Springer-Verlag, 
    pp. 571-679, 1986.
        72. Van der Riet, F., J. de St. J, P. A. Human, D. K. C. Cooper, 
    et al., ``Virological Implications of the Use of Primates in 
    Xenotransplantation,'' Transplantation Proceedings, Vol. XIX, pp. 
    4068-4069, 1987.
        73. Vanin, E. F., M. Kaloss, C. Broscius, and A. W. Neinhuis, 
    ``Characterization of Replication-Competent Retroviruses From 
    Nonhuman Primates With Virus-induced T-cell Lymphomas and 
    Observations Regarding the Mechanism of Oncogenesis,'' Journal of 
    Virology, No. 68, pp. 4241-4250, 1994.
        74. Weigler, B. T., ``Biology of B Virus in Macaque and Human 
    Hosts: A Review,'' Clinical Infectious Diseases, Vol. 14, pp. 555-
    567, 1992.
        75. ``Ebola Haemorrhagic Fever in Zaire, 1976: Report of an 
    International Commission,'' Bulletin of the World Health 
    Organization, No. 56, pp. 271-293, 1978.
        76. ``Basic Consideration in Assessing and Preventing 
    Occupational Infections in Personnel Working with Nonhuman 
    Primates,'' Journal of Medical Primatology, Vol. 16, No. 20, pp. 51-
    138, 1987.
        77. Anonymous, ``Biohazards Associated With Natural and 
    Experimental Diseases of Nonhuman Primates,'' Papers Presented at a 
    Seminar, Baltimore, MD, November 5, 1985, Journal of Medical 
    Primatology, Vol. 16, No. 20, pp. 51-138, 1987.
        78. Chari, R. S., B. H. Collins, J. C. Magee, J. M. DiMaio, et 
    al., Brief Report: Treatment of Hepatic Failure With Ex Vivo Pig-
    Liver Perfusion Followed by Liver Transplantation, New England 
    Journal of Medicine, Vol. 331, pp. 234-237, 1994.
        79. Cooper, D. K. C., Y. Ye, L. L. Rolf, and N. Zuhdi, ``The Pig 
    as Potential Organ Donor for Man,'' In ``Xenotransplantation: The 
    Transplantation of Organ and Tissues Between Species,'' D. K. C. 
    Cooper, E. Kemp, K. Reemstma, and D. J. G. White (editors), Berlin: 
    Springer-Verlag, pp. 481-500, 1991.
        80. Sachs, D. H., ``MHC-homozygous Miniature Swine, ``Swine as 
    Models in Biomedical Research,'' M. M. Swindle (editor) Ames, Iowa: 
    Iowa State University Press, pp. 3-15, 1992.
        81. Moore, C., Biosecurity and Minimal Disease Herds, Veterinary 
    Clinics of North America: Food Animal Practice, pp. 461-474, 199.
        82. Bjoersdorff, A., O. Korsgren, A. Andersson, J. Tollemar, et 
    al., ``Microbiologic Screening as a Preparatory Step for Clinical 
    Xenografting of Procine Fetal Islet-like Cell Clusters,'' 
    Transplantation Proceedings, No. 24, pp. 674-676, 1992.
        83. Bouillant, A.M.P., A.S. Greig, M.M. Lieber, and G.J. Todaro, 
    ``Type C Virus Production by a Continuous Line of Pig Oviduct Cells 
    (PFT),'' Journal of General Virology, Vol. 27, pp. 173-180, 1975.
    
    [[Page 49932]]
    
        84. Fishman, J.A., ``Miniature Swine as Organ Donors for Man: 
    Strategies for Prevention of Xenotransplant-associated Infections,'' 
    Xenotransplantation, Vol. 1, pp. 47-57, 1994.
        85. Frazier, M.E., ``Evidence for Retrovirus in Miniature Swine 
    With Radiation-induced Leukemia or Metaplasia, Archives of Virology, 
    Vol. 83, pp. 83-97, 1985.
        86. Smith, D.M., ``Endogenous Retroviruses in Xenografts,'' New 
    England Journal of Medicine, Vol. 328, pp. 142-143, 1983.
        87. Ye, Y., M. Niekrasz, S. Kosanke, et al., ``The Pig as a 
    Potential Organ Donor for Man, A Study of Potentially Transferrable 
    Disease From Donor Pig to Recipient Man,'' Transplantation, Vol. 57, 
    No. 5, pp. 694-703, March 15, 1994.
        88. Wells, G.A.H., A.C. Scott, C.T. Johnson, R.F. Gunning, et 
    al., ``A Novel Progressive Spongiform Encephalopathy in Cattle,'' 
    Veterinary Record, No. 121, pp. 419-420, 1987.
    
    F. Clinical Issues
    
        1. Nathanson, N., ``Epidemiology,'' Chapter 12, In Virology, 2d 
    Ed., B.N. Fields, D.M. Knipe, et al. (editors), New York: Raven 
    Press, Led., pp. 267-291, 1990.
        2. Declich, S., and A.O. Carter, ``Public Health Surveillance: 
    Historical Origins, Methods and Evaluation,'' Bulletin of the World 
    Health Organization, No. 72, pp. 285-304, 1994.
    
        Dated: September 13, 1996.
    Donna E. Shalala,
    Secretary.
    [FR Doc. 96-24448 Filed 9-20-96; 8:45 am]
    BILLING CODE 4150-04-M
    
    
    

Document Information

Published:
09/23/1996
Department:
Public Health Service
Entry Type:
Notice
Action:
Notice.
Document Number:
96-24448
Dates:
Written comments December 23, 1996.
Pages:
49920-49932 (13 pages)
Docket Numbers:
Docket No. 96M-0311
PDF File:
96-24448.pdf