Swine Influenza Virus: Zoonotic Potential and Vaccination Strategies for the Control of Avian and Swine Influenzas

Similar documents
Sponsors. Production Assistants Steven Claas Lynn Leary. Layout David Brown

Influenza Viruses A Review

INFLUENZA-2 Avian Influenza

Introduction to Avian Influenza

Agricultural Outlook Forum Presented: February 16, 2006 THE CURRENT STATE OF SCIENCE ON AVIAN INFLUENZA

OIE Situation Report for Highly Pathogenic Avian Influenza

Avian influenza Avian influenza ("bird flu") and the significance of its transmission to humans

VIROLOGY OF INFLUENZA. Subtypes: A - Causes outbreak B - Causes outbreaks C - Does not cause outbreaks

REVIEW Cell-mediated Immunity to Influenza Virus Infections: From the Perspective to the Vaccine Development against Highly Pathogenic Avian Influenza

OIE Situation Report for Highly Pathogenic Avian Influenza

OIE Situation Report for Highly Pathogenic Avian Influenza

Zoonotic potential of non-avian influenza A viruses

OIE Situation Report for Avian Influenza

Influenza and the Poultry Link

Avian Influenza: Outbreak in Spring 2015 and Preparing for Fall

Avian Influenza. Regional Workshops: Veterinary Discussion. Will Garton


SCIENTIFIC DISCUSSION

Pandemic Influenza influenza epidemic: realization of a worst-case scenario

Veterinary Microbiology

Influenza A virus subtype H5N1

HUMAN INFLUENZA VIRUSES AND THE POTENTIAL FOR INTER-SPECIES TRANSMISSION

Vaccines of today and products needed for the short-, intermediate- and longterm. OIE/FAO OFFLU Conference Beijing China December 4-6, 2013

Comparison of three serological assays to determine the cross-reactivity of antibodies from eight genetically diverse U.S. Swine influenza viruses

Research Issues in Animal Surveillance and Pandemic Planning

Current Vaccines: Progress & Challenges. Influenza Vaccine what are the challenges?

Influenza: Ecology and Continuing Evolution

INFLUENZA VIRUS. INFLUENZA VIRUS CDC WEBSITE

Where Health Care Meets Policy. with Dr. Mike Magee

Evolution of influenza

Surviving an HPAI Outbreak

The A(H7N9) influenza outbreak in China

Multiple lineages of antigenically and genetically diverse influenza A virus co-circulate in the United States swine population

Avian Influenza (Bird Flu) Fact Sheet

FluSure XP TM Update with Regards to 2009 H1N1 Swine Influenza Virus

Ralph KY Lee Honorary Secretary HKIOEH

Update: Porcine epidemic diarrhoea and swine influenza. Eric Neumann DVM PhD Epi-Insight Limited Palmerston North, New Zealand

Avian Influenza (AI) National & International Update

For the control of avian influenza

INFLUENZA A VIRUS. Structure of the influenza A virus particle.

Immunogenicity of Avian Influenza H7N9 Virus in Birds

Reassortment of influenza A virus genes linked to PB1 polymerase gene

Impact of Avian Influenza on U.S. Poultry Trade Relations 2002

SEA/CD/154 Distribution : General. Avian Influenza in South-East Asia Region: Priority Areas for Research

Avian influenza - current situation and future trends

High Pathogenic Avian Influenza

Kyoung-Jin Yoon 1, Bruce H. Janke, Rick W. Swalla, Gene Erickson

OIE Situation Report for Avian Influenza

Seasonal Influenza. Zoonotic Influenza. Pandemic Influenza

Influenza: The past, the present, the (future) pandemic

Mexico H7N3 HPAI Summary

Influenza: Seasonal, Avian, and Otherwise

H9N2 avian influenza virus in Korea: evolution and vaccination

Laboratory Diagnosis of Avian Influenza and Newcastle Disease

Min Levine, Ph. D. Influenza Division US Centers for Disease Control and Prevention. June 18, 2015 NIBSC

Current limitations on the use of vaccination against avian influenza

Broadly protective influenza vaccines for pandemic preparedness. Suresh Mittal Department of Comparative Pathobiology Purdue University

Application of Reverse Genetics to Influenza Vaccine Development

Influenza: An Emerging Disease

Avian Influenza: Implications for Agriculture and Public Health. Faculty. Avian Influenza Orthomyxovirus (type A) - 15 (16) Hemagglutinin and 9

Avian Influenza: Armageddon or Hype? Bryan E. Bledsoe, DO, FACEP The George Washington University Medical Center

Avian Influenza Outbreaks. in the USA (12/2014 5/2015)

C E E Z A D. Rational Development of Influenza Vaccines: NDV-based influenza vaccines for poultry and livestock

Optimal Use of Vaccines for Control of Influenza A Virus in Swine

Avian Influenza. Poultry Growers September 2015

AVIAN INFLUENZA & NEWCASTLE ACTIVITIES

Coordinating Global Surveillance for Influenza in Swine. OFFLU Swine Influenza Virus Group

OIE Situation Report for Avian Influenza

Event based and active surveillance for avian influenza

Antigenic categorization of contemporary H3N2 Swine influenza virus isolates using a high-throughput serum neutralization assay

ZOONOTIC DISEASES, HUMAN HEALTH AND

The development of avian influenza vaccines for emergency use

MINI-REVIEW. Protein & Cell. The emergence of pandemic influenza viruses

Frequently Asked Questions on Avian Influenza

Surveillance and control programmes for terrestrial and aquatic animals in Norway

OIE Situation Report for Avian Influenza

Overview of the Influenza Virus

Fraser Menzies Veterinary Epidemiology Unit DARD

Contribution of avian influenza data through OFFLU network

OIE Situation Report for Avian Influenza

Final Report for the Outbreak of Highly Pathogenic Avian Influenza (HPAI) in the United States

Coordinating Global Surveillance for Influenza in Swine. OFFLU Swine Influenza Group


Conflict of Interest and Disclosures. Research funding from GSK, Biofire

Influenza. By Allison Canestaro-Garcia. Disease Etiology:

دکتر بهروز نقیلی استاد بیماریهای عفونی مرکس تحقیقات بیماریهای عفونی و گرمسیری پاییس 88

The surveillance programme for avian influenza (AI) in poultry in Norway 2017

The surveillance programme for avian influenza (AI) in poultry in Norway 2015

Flu, Avian Flu and emerging aspects (H1N1 resistance)

Existence of reassortant A (H1N2) swine influenza viruses in Saitama Prefecture, Japan

Avian Influenza A(H7N9) 6 February 2014 Surveillance Update

Principles for event based and active avian influenza surveillance. Les Sims Asia Pacific Veterinary Information Services

Feasibility of Production of Human AI Vaccine in AI vaccine Manufacturers in China. Gu Hong Ministry of Agriculture of P. R. China

MSU Extension Publication Archive. Scroll down to view the publication.

Highly Pathogenic Avian Influenza Virus Research

NIH Public Access Author Manuscript Clin Infect Dis. Author manuscript; available in PMC 2007 September 7.

Evaluation of a Commercial ELISA Kit (IDEXX) to Differentiate AI Virus-infected Poultry from AI- Vaccinated Poultry (DIVA)

Cover Page. The handle holds various files of this Leiden University dissertation

Avian influenza and pandemic threats

Lecture 19 Evolution and human health

Transcription:

SUPPLEMENT ARTICLE Swine Influenza Virus: Zoonotic Potential and Vaccination Strategies for the Control of Avian and Swine Influenzas Eileen Thacker 1 and Bruce Janke 2 Departments of 1 Veterinary Microbiology and Preventive Medicine and 2 Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames Influenza viruses are able to infect humans, swine, and avian species, and swine have long been considered a potential source of new influenza viruses that can infect humans. Swine have receptors to which both avian and mammalian influenza viruses bind, which increases the potential for viruses to exchange genetic sequences and produce new reassortant viruses in swine. A number of genetically diverse viruses are circulating in swine herds throughout the world and are a major cause of concern to the swine industry. Control of swine influenza is primarily through the vaccination of sows, to protect young pigs through maternally derived antibodies. However, influenza viruses continue to circulate in pigs after the decay of maternal antibodies, providing a continuing source of virus on a herd basis. Measures to control avian influenza in commercial poultry operations are dictated by the virulence of the virus. Detection of a highly pathogenic avian influenza (HPAI) virus results in immediate elimination of the flock. Low-pathogenic avian influenza viruses are controlled through vaccination, which is done primarily in turkey flocks. Maintenance of the current HPAI virus free status of poultry in the United States is through constant surveillance of poultry flocks. Although current influenza vaccines for poultry and swine are inactivated and adjuvanted, ongoing research into the development of newer vaccines, such as DNA, live-virus, or vectored vaccines, is being done. Control of influenza virus infection in poultry and swine is critical to the reduction of potential cross-species adaptation and spread of influenza viruses, which will minimize the risk of animals being the source of the next pandemic. INFLUENZA VIRUSES AND PIGS The influenza viruses that infect pigs are influenza A viruses in the Orthomyxoviridae family. The presence of influenza in pigs was first recognized clinically during the summer of 1918 in the United States, at about the time of the Spanish influenza pandemic [1]. Influenza virus was first isolated from pigs in North America in Potential conflicts of interest: none reported. Presented in part: Harvard University Asian Flus and Avian Influenza Workshop, Cambridge, Massachusetts, 8 10 December 2006. Financial support: supplement sponsorship is detailed in the Acknowledgments. Reprints or correspondence: Dr. Eileen Thacker, Dept. of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, 2118 Vet Med Bldg., Iowa State University, Ames, IA 50011 (ethacker@iastate.edu). The Journal of Infectious Diseases 2008; 197:S19 24 2008 by the Infectious Diseases Society of America. All rights reserved. 0022-1899/2008/19704S1-0005$15.00 DOI: 10.1086/524988 1930 [2]. Isolation of a swine influenza virus from humans in 1974 confirmed that swine influenza viruses are zoonotic in nature [3]. The genetic makeup of swine influenza viruses is identical to that of other influenza A viruses and consists of a negative-sense, segmented RNA genome. This segmented makeup allows for reassortment between viruses, resulting in the generation of new, antigenically novel viruses. The 2 major surface glycoproteins, hemagglutinin (H) and neuraminidase (N), are important for the determination of host range, antigenicity, and potential pathogenicity and serve as targets for diagnostic assays. It is important to recognize that the genetic makeup of swine influenza viruses in swine populations throughout the world can differ significantly. The swine influenza viruses in Europe differ significantly, in their antigenic and genetic makeup, from the viruses currently circulating in North America, even Swine and Avian Influenzas JID 2008:197 (Suppl 1) S19

though they consist of the same H and N subtypes [4]. Less is known about the genetic makeup of viruses currently circulating among pigs throughout Asia. Historically, the genetic makeup of swine influenza viruses was relatively stable in North America for nearly 80 years and consisted of 1 predominant subtype, known as classic swine influenza A(H1N1) virus (ch1n1 virus) [1]. ch1n1 virus had a genetic makeup composed primarily of swine influenza virus genes and was genetically and antigenically similar to the first swine influenza virus isolated in 1930. In 1998, H3N2 virus emerged in the US swine population and was composed of avian, human, and swine influenza virus genes [5 7]. This H3N2 virus consisted of H, N, and polymerase B1 genes that originated from human influenza virus; matrix and nonstructural protein and nucleoprotein (NP) genes from ch1n1 virus; and polymerase A and polymerase B2 genes that originated from avian influenza virus. Genetically, the human influenza virus genes in this H3N2 virus were similar to those in an influenza virus contained in a human vaccine used in the mid- 1990s. The entrance of this H3N2 virus into the US swine population has resulted in the emergence of multiple reassortant influenza viruses [8]. Since then, influenza viruses that have circulated in US swine-production systems have consisted of H1 viruses with H and N from the ch1n1 virus and internal genes from the H3N2 virus (i.e., rh1n1 virus) or with H from the ch1n1 virus and N and internal genes from the H3N2 virus (i.e., H1N2 virus) [6, 9]. An increased rate of genetic change has occurred among both H1 and H3 subtypes, with multiple genetically and antigenically diverse viruses of both major subtypes (H1 and H3) circulating in swine herds. Recently, there have been reports of H3N1 viruses circulating in Asia and the United States [10 12]. Even more recently, an H1N1 virus composed of only human influenza virus genes has entered the US swine population [13]. In addition to the influenza viruses described above, which are isolated fairly commonly from US swine herds, H3 and H1N1 avian influenza virus subtypes have been isolated from pigs in Canada [14]. As a result of the constantly changing genetic makeup of influenza viruses in pigs, the US swine industry is continually scrambling to respond to the influenza viruses circulating within individual production systems. The relationship between swine and human influenza disease has long been recognized as important. Pigs often are thought to be mixing vessels or to act as transmission agents in the adaptation of avian influenza viruses to mammals, including humans. Infection by influenza viruses is initiated by a binding event between viral H and sialic acid containing moieties on target cells. Different host species display differing virus-binding receptors with preferences for either Neu5Aca2-3Gal or Neu5Aca2-6Gal epitopes [15, 16]. H1, H2, and H3 subtypes of human influenza virus bind to the NeuAca2-6Gal linked sialic acid found in the human respiratory tract. In contrast, avian influenza viruses bind to the NeuAca2-3Gal receptors found primarily in the enteric tract of waterfowl [17]. The receptor specificity of avian influenza viruses is due to conserved amino acids at positions 190, 194, 225, 226, and 223 [16]. Swine influenza viruses have been reported to bind to both Neu5Aca2-3Gal and NeuAca2-6Gal receptors in the swine respiratory tract [16, 18, 19]. In addition, the N-glycolyl analog (Neu5Gc), a receptor that binds to swine influenza viruses with increased affinity, has been shown to be expressed abundantly in pigs [20]. More recently, both swine and avian influenza viruses have been shown to bind, with high affinity, to sulfated forms of sialyloligosaccharide receptors [21, 22]. Much of the research regarding influenza viruses has used viruses propagated in embryonated hen eggs, which has been shown to alter virus receptor specificity, increasing the affinity of human viruses for a2-3 linked sialic acid [17, 23, 24]. Recent research has demonstrated that the binding abilities of influenza viruses propagated by use of Madin-Darby canine kidney cells differ according to their H subtype [18]. Thus, either H3 subtypes of human influenza virus or ch1n1 preferentially bind to Neu5Aca2-6Gal, a pattern typical of human influenza viruses. In contrast, avian-like H1 and H3 subtypes of swine influenza virus bind to Neu5Aca2-6Gal and Neu5Aca2-3Gal. Transmission of influenza viruses between host species is of obvious concern, and transmission of human and swine influenza viruses between the 2 species has been well documented. The fear that such transmission can generate was demonstrated by the concern for a swine influenza pandemic in 1974 that was raised when an influenza outbreak caused by ch1n1 virus was documented in Fort Dix, New Jersey. During this outbreak, 1 soldier died, and 12 additional soldiers were hospitalized owing to respiratory disease [25 28]. Interestingly, no known exposure to pigs was ever documented. A subsequent investigation found that up to 230 soldiers had been infected with or exposed to the virus [25, 26]. Since then, a number of studies have found that people in contact with swine have higher antibody levels due to exposure to swine influenza viruses, although cases of clinical disease associated with swine influenza viruses are sporadic [29, 30]. In addition to transmission between humans and pigs, swine influenza viruses also have been isolated from turkeys on a fairly regular basis, indicating transmission between pigs and avian species [31 34]. It has been documented that pigs can be infected with the current highly pathogenic avian influenza (HPAI) H5N1 virus that has been responsible for disease in poultry in Asia [35]. These findings suggest that pigs would be the ideal mixing vessel for the creation of new avian/mammalian influenza viruses capable of causing novel diseases with the potential for producing pandemics in the human population. Whether this will happen S20 JID 2008:197 (Suppl 1) Thacker and Janke

easily, however, is less clear, although it is apparent that, in the US swine industry, transmission of influenza viruses between swine and humans is fairly common and is bidirectional. The pandemics of the past appear to have involved the introduction of whole or partial avian influenza viruses into a human population directly, rather than through swine. VACCINATION STRATEGIES FOR THE CONTROL OF SWINE INFLUENZA VIRUS INFECTION Two primary obstacles confound programs for successful influenza vaccination of pigs namely, viral antigenic shift and drift and the effect of maternally derived antibodies on vaccine efficacy. The vaccination of sows is a common practice used by the swine industry to increase and prolong maternally derived antibody levels in young pigs, to protect them against clinical disease. However, the presence of maternal antibodies reduces vaccine efficacy, making it difficult to vaccinate pigs prior to exposure to the virus and resulting in an increased incidence of disease among pigs as their maternal antibodies decay and they become susceptible to virus infection and disease [36]. As a result, influenza viruses potentially can circulate in swine herds on a regular basis, owing to the constant introduction of immunologically naive animals into the herd. Current swine influenza vaccines are adjuvanted, inactivated, whole-virus vaccines prepared typically from virus propagated in embryonated hen eggs. These vaccines stimulate high titers of IgG in serum and lungs, which protect against clinical disease. Antibodies against the H protein appear to be the most protective. Protection against infection is not complete, but virus multiplication and shedding are greatly reduced. An issue currently under discussion is the need to update US Department of Agriculture clearance procedures for new vaccine licensure to allow the addition of newer isolates into vaccines, to keep pace with changes in virus genetics in the field. At present, the requirements for licensure of swine influenza vaccines are much more laborious and expensive than those in place for human influenza vaccines. Approval of animal vaccines requires demonstration of vaccine safety, efficacy, and potency through multiple experimental and field trials. As a result of the extensive trials required, a vaccine often takes up to 5 years to be licensed. Fortunately, cross-protection against antigenic variants of influenza viruses appears to be broader in pigs, compared with that provided by human influenza vaccines [37]. Currently, modified live influenza virus vaccines are not available for swine, although results of recent studies of genedeleted vaccines have been reported [38]. The advantage of modified live-virus vaccines is enhanced stimulation of cellmediated immunity, directed primarily against conserved NP, thus providing more heterosubtypic immunity (i.e., protection across subtypes). One concern about live-virus vaccines would be possible reassortment between field strains and the vaccine virus, producing new reassortant viruses. Vectored vaccines using vaccinia virus, baculovirus, alphavirus, or adenovirus also are being studied [39, 40]. DNA vaccines also have been studied by using chicken, mouse, ferret, and primate models of influenza virus infection. The theoretical advantage of these vaccines is the production of viral protein with normal conformation, without the risks associated with the use of live virus. Such vaccines stimulate long-lasting immunity through both humoral and cell-mediated systems. DNA vaccines appear to be ideal for use with swine, since they would provide heterosubtypic immunity and the internalization of DNA inside host cells would minimize interference by maternal antibodies. Unfortunately, to date, experimental trials of DNA vaccines in pigs have not proved successful. However, DNA vaccines may prove to be excellent primer vaccines when followed by more-conventional inactivated vaccines [41, 42]. AVIAN INFLUENZA VIRUSES AND VACCINE STRATEGIES FOR DOMESTIC POULTRY Birds, especially waterfowl, can be infected with any influenza virus, regardless of H or N subtype. The receptors to which avian influenza viruses bind were described above. In contrast to influenza virus infection in swine or humans, disease in wild waterfowl is rare and observed only when caused by HPAI viruses. No infection-control strategies are currently used on wild birds; because influenza viruses are constantly circulating among wild waterfowl, a primary goal is to minimize the exposure of either domestic poultry or swine to these viruses. Protective immunity against influenza virus infection in birds is similar to that in mammals that is, as in pigs and humans, antibodies against the H protein are important for protection against virus infection, which is subtype specific. However, within-subtype protection appears to be more effective in birds over a wider range of viral antigenic variation, compared with that provided by human influenza vaccines. In addition, antigenic drift among field strains that affects cross-protection has not been a significant problem with avian influenza viruses; however, over the past decade of vaccine use in commercial poultry operations in Mexico, surveillance and genomic/antigenic analysis of influenza viruses have documented that antigenic drift can occur in avian species as well [43]. Inactivated vaccines most commonly are used to vaccinate commercial poultry against influenza viruses and are fairly effective. Subunit vaccines in which selected influenza virus genes have been inserted into a fowlpox vector virus have been developed and tested in experimental trials but have not been used in field situations [44]. In contrast to human and swine influenza viruses, variation in the pathogenicity of avian influenza viruses and, thus, their Swine and Avian Influenzas JID 2008:197 (Suppl 1) S21

designation as HPAI viruses versus low-pathogenic avian influenza (LPAI) viruses is important when vaccine strategies for domestic poultry are considered. In the United States, HPAI viruses are considered to be foreign animal viruses, with detection resulting in complete elimination of a flock. In contrast, infection by LPAI viruses occurs more commonly in US commercial poultry populations, and the management response to infection depends on the influenza virus subtype involved. Vaccination against LPAI viruses is done primarily in turkey breeder flocks. Infection by LPAI viruses does not result in high mortality rates among turkeys, but egg production can be affected significantly. In the 1980s and early 1990s, most turkeys were raised in the open, and frequent infection of these flocks was attributed to migrating waterfowl, which are natural, typically asymptomatic carriers of avian influenza viruses. Vaccination and controlled marketing were used to eradicate influenza virus from affected turkey flocks. After the mid-1990s, turkey production was moved to confinement facilities, thus greatly reducing potential exposure to wild birds. The open buildings used to house turkeys still expose the birds to prevailing winds, and the H1 and H3 influenza virus subtypes that currently are of concern usually originate from neighboring swine operations. Both broiler and egg-laying chickens are housed in more-controlled environments, and infection with any of the influenza viruses, including LPAI isolates, is rare. In addition, a withdrawal period of at least 43 days between vaccination and slaughter must be preserved with broiler chickens; thus, vaccination of broiler chickens is not done routinely. Infection of poultry flocks with any H5 or H7 influenza virus subtype, even LPAI isolates, elicits a response much different from that for infection by other influenza virus subtypes. All HPAI viruses are H5 or H7 subtype strains, although the opposite is not true that is, not all H5 and H7 subtype strains are HPAI viruses. Past experience has shown that LPAI H5 or H7 viruses are capable of mutating into HPAI viruses; thus, flocks with H5 or H7 influenza virus infections usually are eliminated [45]. Although total eradication of flocks has been used more frequently, vaccination in conjunction with quarantine and surveillance also has been used to successfully eliminate H5 or H7 influenza viruses from domestic poultry flocks. Surveillance programs and trade issues are important considerations in decisions about vaccine use in poultry industries. Serologic surveillance is used to monitor against the introduction of HPAI viruses and to certify flocks free of avian influenza virus, especially for marketing purposes. Loss of the US poultry industry s status as free of HPAI viruses would have a severe impact on exports of birds and poultry products to other countries. In the United States, vaccines against avian influenza typically are oil-adjuvanted, inactivated, whole-virus vaccines that induce antibodies to all viral proteins. The serologic tests used routinely in surveillance programs are agar-gel immunodiffusion and commercial ELISA. Both assays use conserved internal viral proteins (matrix or NP) as the test antigen and, thus, give antibody-positive results whether birds are infected or vaccinated. Thus, with current vaccines and serologic assays, it is impossible to determine whether a vaccinated flock is infected with the virus. Because the vaccine does provide protection against clinical disease, HPAI viruses could circulate undetected within a flock. For this reason, vaccination against H5 and H7 influenza viruses is not done routinely, and the use of these vaccines requires federal approval. Surveillance for influenza viruses in poultry flocks often includes the placement of unvaccinated sentinel birds in the flock, and these birds are monitored serologically for the presence of antibodies or virus by use of polymerase chain reaction assays. This allows detection of infection as well as determination of the influenza virus subtype circulating in the flock. A process for differentiating infected from vaccinated animals (called DIVA ) also is used [46]. One of the first such programs in the control of avian influenza used a heterologous N subtype, H7N3, in the vaccine to immunize against H7N2 influenza virus infection, enabling the differential detection of N3 antibodies in vaccinated birds versus infected birds. Similarly, subunit vaccines are used in which the desired gene, typically the H protein gene, is inserted into a fowlpox vector [44]. With this approach, routine serologic assays that detect matrix protein or NP antibodies can be used for surveillance. SUMMARY Influenza virus infections in swine and poultry are potential sources of viruses for the next pandemic among humans. The bidirectional transmission of influenza viruses between pigs and humans has been documented. Although not common, infection of swine by avian influenza viruses does occur and serves as a potential source of new, genetically diverse influenza viruses capable of infecting humans. As a result, the use of control strategies, especially vaccination, is critical for the control of influenza virus infections among domestic animals, to reduce their potential as sources for outbreaks among humans. In addition, the vaccination of people who work with swine is encouraged, to reduce the chance of human influenza viruses being spread to pigs. Current vaccination strategies for the control of disease in both swine and poultry are fairly effective, but ongoing research needs to be done to ensure the control of both swine and avian influenza virus infections. Acknowledgments The Harvard University Asian Flus and Avian Influenza Workshop was hosted by the Harvard University Department of Anthropology, Harvard School of Public Health, and Harvard Asia Center and was supported by S22 JID 2008:197 (Suppl 1) Thacker and Janke

the National Science Foundation, Harvard Asia Center, and the Michael Crichton Fund. Supplement sponsorship. This article was published as part of a supplement entitled Avian and Pandemic Influenza: A Biosocial Approach, sponsored by the National Science Foundation, Harvard Asia Center, and the Michael Crichton Fund. References 1. Easterday BC, Van Reeth K. Swine influenza. In: Straw BE, D Allaire SD, Mengeling WL, Taylor DJ, eds. Diseases of swine. 8th ed. Ames, Iowa: Iowa State University Press, 1999:277 90. 2. Shope RE. Swine influenza. III. Filtration experiments and etiology. J Exp Med 1931; 54:373 85. 3. Smith TF, Burgert EO Jr, Dowdle WR, Noble GR, Campbell RJ, Van Scoy RE. Isolation of swine influenza virus from autopsy lung tissue of man. N Engl J Med 1976; 294:708 10. 4. Kothalawala H, Toussaint MJ, Gruys E. An overview of swine influenza. Vet Q 2006; 28:46 53. 5. Karasin AI, Schutten MM, Cooper LA, et al. Genetic characterization of H3N2 influenza viruses isolated from pigs in North America, 1977 1999: evidence for wholly human and reassortant virus genotypes. Virus Res 2000; 68:71 85. 6. Webby RJ, Rossow K, Erickson G, Sims Y, Webster R. Multiple lineages of antigenically and genetically diverse influenza A virus co-circulate in the United States swine population. Virus Res 2004; 103:67 73. 7. Webby RJ, Swenson SL, Krauss SL, Gerrish PJ, Goyal SM, Webster RG. Evolution of swine H3N2 influenza viruses in the United States. J Virol 2000; 74:8243 51. 8. Olsen CW. The emergence of novel swine influenza viruses in North America. Virus Res 2002; 85:199 210. 9. Karasin AI, Olsen CW, Anderson GA. Genetic characterization of an H1N2 influenza virus isolated from a pig in Indiana. J Clin Microbiol 2000; 38:2453 6. 10. Lekcharoensuk P, Lager KM, Vemulapalli R, Woodruff M, Vincent AL, Richt JA. Novel swine influenza virus subtype H3N1, United States. Emerg Infect Dis 2006; 12:787 94. 11. Ma W, Gramer M, Rossow K, Yoon KJ. Isolation and genetic characterization of new reassortant H3N1 swine influenza virus from pigs in the midwestern United States. J Virol 2006; 80:5092 6. 12. Shin JY, Song MS, Lee EH, et al. Isolation and characterization of novel H3N1 swine influenza viruses from pigs with respiratory diseases in Korea. J Clin Microbiol 2006; 44:3923 7. 13. Gramer M. Reassortant human/swine H1N1 and H1N2 influenza virus infections in US swine. In: Proceedings of the 37th annual meeting of the American Association of Swine Veterinarians (Kansas City, MO). Perry, IA: American Association of Swine Veterinarians, 2006:463 4. 14. Karasin AI, West K, Carman S, Olsen CW. Characterization of avian H3N3 and H1N1 influenza A viruses isolated from pigs in Canada. J Clin Microbiol 2004; 42:4349 54. 15. Matrosovich M, Tuzikov A, Bovin N, et al. Early alterations of the receptor-binding properties of H1, H2, and H3 avian influenza virus hemagglutinins after their introduction into mammals. J Virol 2000; 74:8502 12. 16. Matrosovich MN, Gambaryan AS, Teneberg S, et al. Avian influenza A viruses differ from human viruses by recognition of sialyloligosaccharides and gangliosides and by a higher conservation of the HA receptor-binding site. Virology 1997; 233:224 34. 17. Ito T. Interspecies transmission and receptor recognition of influenza A viruses. Microbiol Immunol 2000; 44:423 30. 18. Gambaryan AS, Karasin AI, Tuzikov AB, et al. Receptor-binding properties of swine influenza viruses isolated and propagated in MDCK cells. Virus Res 2005; 114:15 22. 19. Rogers GN, D Souza BL. Receptor binding properties of human and animal H1 influenza virus isolates. Virology 1989; 173:317 22. 20. Suzuki T, Horiike G, Yamazaki Y, et al. Swine influenza virus strains recognize sialylsugar chains containing the molecular species of sialic acid predominantly present in the swine tracheal epithelium. FEBS Lett 1997; 404:192 6. 21. Gambaryan AS, Tuzikov AB, Pazynina GV, Webster RG, Matrosovich MN, Bovin NV. H5N1 chicken influenza viruses display a high binding affinity for Neu5Acalpha2-3Galbeta1-4(6-HSO3)GlcNAc containing receptors. Virology 2004; 326:310 6. 22. Marinina VP, Gambarian AS, Tuzikov AB, et al. Evolution of the receptor specificity of influenza viruses hemagglutinin in its transfer from duck to pig and man [in Russian]. Vopr Virusol 2004; 49:25 30. 23. Gambaryan AS, Robertson JS, Matrosovich MN. Effects of egg-adaptation on the receptor-binding properties of human influenza A and B viruses. Virology 1999; 258:232 9. 24. Gambaryan AS, Tuzikov AB, Piskarev VE, et al. Specification of receptor-binding phenotypes of influenza virus isolates from different hosts using synthetic sialylglycopolymers: non egg-adapted human H1 and H3 influenza A and influenza B viruses share a common high binding affinity for 6 -sialyl(n-acetyllactosamine). Virology 1997; 232: 345 50. 25. Gaydos JC, Hodder RA, Top FH Jr, et al. Swine influenza A at Fort Dix, New Jersey (January February 1976). II. Transmission and morbidity in units with cases. J Infect Dis 1977; 136(Suppl):S363 8. 26. Gaydos JC, Hodder RA, Top FH Jr, et al. Swine influenza A at Fort Dix, New Jersey (January February 1976). I. Case finding and clinical study of cases. J Infect Dis 1977; 136(Suppl):S356 62. 27. Gaydos JC, Top FH Jr, Hodder RA, Russell PK. Swine influenza A outbreak, Fort Dix, New Jersey, 1976. Emerg Infect Dis 2006; 12:23 8. 28. Hodder RA, Gaydos JC, Allen RG, Top FH Jr, Nowosiwsky T, Russell PK. Swine influenza A at Fort Dix, New Jersey (January February 1976). III. Extent of spread and duration of the outbreak. J Infect Dis 1977; 136(Suppl):S369 75. 29. Myers KP, Olsen CW, Setterquist SF, et al. Are swine workers in the United States at increased risk of infection with zoonotic influenza virus? Clin Infect Dis 2006; 42:14 20. 30. Olsen CW, Brammer L, Easterday BC, et al. Serologic evidence of H1 swine influenza virus infection in swine farm residents and employees. Emerg Infect Dis 2002; 8:814 9. 31. Suarez DL, Woolcock PR, Bermudez AJ, Senne DA. Isolation from turkey breeder hens of a reassortant H1N2 influenza virus with swine, human, and avian lineage genes. Avian Dis 2002; 46:111 21. 32. Wright SM, Kawaoka Y, Sharp GB, Senne DA, Webster RG. Interspecies transmission and reassortment of influenza A viruses in pigs and turkeys in the United States. Am J Epidemiol 1992; 136:488 97. 33. Choi YK, Lee JH, Erickson G, et al. H3N2 influenza virus transmission from swine to turkeys, United States. Emerg Infect Dis 2004; 10: 2156 60. 34. Altmuller A, Kunerl M, Muller K, Hinshaw VS, Fitch WM, Scholtissek C. Genetic relatedness of the nucleoprotein (NP) of recent swine, turkey, and human influenza A virus (H1N1) isolates. Virus Res 1992; 22:79 87. 35. Choi YK, Nguyen TD, Ozaki H, et al. Studies of H5N1 influenza virus infection of pigs by using viruses isolated in Vietnam and Thailand in 2004. J Virol 2005; 79:10821 5. 36. Kitikoon P, Nilubol D, Erickson BJ, et al. The immune response and maternal antibody interference to a heterologous H1N1 swine influenza virus infection following vaccination. Vet Immunol Immunopathol 2006; 112:117 28. 37. Gramer M, Rossow K. Epidemiology of swine influenza and implications of reassortment. In: Proceedings of the Allen D. Leman Swine Conference (St. Paul, MN). Perry, IA: American Association of Swine Veterinarians, 2004:69 73. 38. Richt JA, Lekcharoensuk P, Lager KM, et al. Vaccination of pigs against swine influenza viruses by using an NS1-truncated modified live-virus vaccine. J Virol 2006; 80:11009 18. 39. Erdmann MM, Crabtree B. A new swine vaccine technology: research update. In: Proceedings of the 14th annual Swine Disease Conference Swine and Avian Influenzas JID 2008:197 (Suppl 1) S23

for Swine Practitioners (Ames, IA). Perry, IA: American Association of Swine Veterinarians, 2006:83 7. 40. Wesley RD, Tang M, Lager KM. Protection of weaned pigs by vaccination with human adenovirus 5 recombinant viruses expressing the hemagglutinin and the nucleoprotein of H3N2 swine influenza virus. Vaccine 2004; 22:3427 34. 41. Heinen PP, Rijsewijk FA, de Boer-Luijtze EA, Bianchi AT. Vaccination of pigs with a DNA construct expressing an influenza virus M2-nucleoprotein fusion protein exacerbates disease after challenge with influenza A virus. J Gen Virol 2002; 83:1851 9. 42. Larsen DL, Olsen CW. Effects of DNA dose, route of vaccination, and coadministration of porcine interleukin-6 DNA on results of DNA vaccination against influenza virus infection in pigs. Am J Vet Res 2002; 63:653 9. 43. Lee CW, Senne DA, Suarez DL. Effect of vaccine use in the evolution of Mexican lineage H5N2 avian influenza virus. J Virol 2004; 78: 8372 81. 44. Swayne DE, Garcia M, Beck JR, Kinney N, Suarez DL. Protection against diverse highly pathogenic H5 avian influenza viruses in chickens immunized with a recombinant fowlpox vaccine containing an H5 avian influenza hemagglutinin gene insert. Vaccine 2000; 18:1088 95. 45. Garcia M, Suarez DL, Crawford JM, et al. Evolution of H5 subtype avian influenza A viruses in North America. Virus Res 1997; 51:115 24. 46. Capua I, Terregino C, Cattoli G, Mutinelli F, Rodriguez JF. Development of a DIVA (differentiating infected from vaccinated animals) strategy using a vaccine containing a heterologous neuraminidase for the control of avian influenza. Avian Pathol 2003; 32:47 55. S24 JID 2008:197 (Suppl 1) Thacker and Janke