Announcement

Collapse
No announcement yet.

NEJM - Update on Avian Influenza A (H5N1) Virus Infection in Humans

Collapse
X
 
  • Filter
  • Time
  • Show
Clear All
new posts

  • NEJM - Update on Avian Influenza A (H5N1) Virus Infection in Humans

    Update on Avian Influenza A (H5N1) Virus Infection in Humans
    <!-- AUTHOR_DISPLAY --> <center> Writing Committee of the Second World Health Organization Consultation on Clinical Aspects of Human Infection with Avian Influenza A (H5N1) Virus


    The unprecedented epizootic of avian influenza A (H5N1) viruses<sup> </sup>among birds continues to cause human disease with high mortality<sup> </sup>and to pose the threat of a pandemic. This review updates a<sup> </sup>2005 report<sup>1</sup> and incorporates information recently published<sup> </sup>or presented at the Second World Health Organization (WHO) Consultation<sup> </sup>on Clinical Aspects of Human Infection with Avian Influenza<sup> </sup>A (H5N1) Virus.<sup>2</sup><sup> </sup> Viral Ecology
    Highly pathogenic avian influenza A (H5N1) viruses are entrenched<sup> </sup>among poultry in parts of Asia, Africa, and perhaps the Middle<sup> </sup>East. The highly pathogenic avian influenza H5 hemagglutinin<sup> </sup>has evolved into many phylogenetically distinct clades and subclades<sup> </sup>(Figure 1)<sup>4</sup><sup>,</sup><sup>5</sup> that generally correlate with antigenic differences<sup> </sup>that must be considered in the selection of candidates for H5N1<sup> </sup>vaccines.<sup>6</sup><sup>,</sup><sup>7</sup> These diverse lineages have been largely separate<sup> </sup>geographically since 2005 (Figure 1),<sup>5</sup> although clade 2.3 viruses<sup> </sup>from China have recently circulated in other Southeast Asian<sup> </sup>countries.<sup>8</sup><sup> </sup>
    <!-- null --> <table cellpadding="0" cellspacing="0"><tbody><tr bgcolor="#e8e8d1"><td><table cellpadding="2" cellspacing="2"><tbody><tr bgcolor="#e8e8d1"><td align="center" bgcolor="#ffffff" valign="top">
    View larger version (71K):
    <nobr>[in this window]
    [in a new window]

    </nobr> </td><td align="left" valign="top"> Figure 1. Evolution of the Hemagglutinin and Other Key Mutations Associated with Virulence or Drug Resistance in Avian Influenza A (H5N1) Virus. The phylogenetic tree is for the hemagglutinin gene of highly pathogenic avian influenza A (H5N1) viruses. The geographic distributions refer to avian isolates, and the tree is based on publicly available sequences. Clade 0 includes viruses that were first recognized to cause human infections in Hong Kong Special Administrative Region in 1997. Viruses from clades and subclades 0, 1, 2.1, 2.2, 2.3, and 7 have caused human disease. Clade 1 viruses predominated in Vietnam, Thailand, and Cambodia in the early phase of the outbreak (2004?2005), and clade 2.1 viruses are endemic in Indonesia. Clade 2.2 viruses were associated with a major outbreak of H5N1 disease in migratory birds in Qinghai Lake, China, and have since spread, causing avian disease in Central and South Asia, the Middle East, Europe, and Africa and human disease in western Asia, the Middle East, and Africa. Clade 2.3 has become dominant in southern China and has also been detected in adjacent countries. (Modified from the WHO Web site: www.who.int/csr/disease/avian_influenza/guidelines/nomenclature/en/index.html.) The influenza genome contains eight individual segments of RNA, several of which encode two proteins. Within clade 1 or clade 2.1 viruses, polymerase basic protein 2 (PB2) Glu627Lys is observed in some isolates of human viruses but not in avian viruses.<sup>3</sup> Some human clade 1 viruses without PB2 627Lys have PB2 701Asn; clade 2.2 viruses of both human and avian origin have PB2 Glu627Lys.<sup>4</sup> The importance of sequence variations in NS1, in which most influenza A (H5N1) viruses contain a carboxyl-terminus?sequence motif that mediates binding to various cellular proteins bearing a PDZ domain, remains to be determined.

    </td></tr></tbody></table></td></tr></tbody></table>
    The influenza A (H5N1) viruses that have infected humans have<sup> </sup>been entirely avian in origin, and they reflect strains circulating<sup> </sup>locally among poultry and wild birds. Avian influenza viruses<sup> </sup>can be maintained, amplified, and disseminated in live-poultry<sup> </sup>markets. Migratory birds may spread A (H5N1) viruses to new<sup> </sup>geographic regions, but their importance as an ecologic reservoir<sup> </sup>is uncertain. The spread of influenza A (H5N1) viruses appears<sup> </sup>to be principally related to the movement of poultry and poultry<sup> </sup>products,<sup>9</sup><sup>,</sup><sup>10</sup> although recent outbreaks of clade 2.2 virus infection<sup> </sup>in sub-Saharan Africa,<sup>11</sup> Egypt, and Europe may indicate introduction<sup> </sup>of the virus by wild birds. The risk of the introduction of<sup> </sup>influenza A (H5N1) viruses into North America by birds migrating<sup> </sup>through Alaska appears to be low.<sup>12</sup><sup> </sup>
    Epidemiology of Human Infections
    Incidence and Demographic Characteristics
    Despite widespread exposures to poultry infected with avian<sup> </sup>influenza A (H5N1) viruses,<sup>13</sup><sup>,</sup><sup>14</sup> influenza A (H5N1) disease<sup> </sup>in humans remains very rare. Since May 2005, the numbers of<sup> </sup>both affected countries<sup>13</sup> and confirmed cases of influenza A<sup> </sup>(H5N1) virus infection (340 cases as of December 14, 2007) have<sup> </sup>increased, in part because of the spread of clade 2.2 viruses<sup> </sup>across Eurasia and to Africa<sup>5</sup><sup>,</sup><sup>15</sup> (Fig. 1 of the Supplementary Appendix,<sup> </sup>available with the full text of this article at www.nejm.org).<sup> </sup>
    The median age of patients with influenza A (H5N1) virus infection<sup> </sup>is approximately 18 years, with 90% of patients 40 years of<sup> </sup>age or younger and older adults underrepresented.<sup>16</sup> The overall<sup> </sup>case fatality proportion is 61%; it is highest among persons<sup> </sup>10 to 19 years of age and lowest among persons 50 years of age<sup> </sup>or older.<sup>16</sup> Whether preexisting immunity, differences in exposure,<sup> </sup>or other factors might contribute to the apparently lower frequency<sup> </sup>of infection and lethal illness among older adults is uncertain.<sup> </sup>Most patients with influenza A (H5N1) virus infection were previously<sup> </sup>healthy. Of six affected pregnant women, four have died, and<sup> </sup>the two survivors had a spontaneous abortion.<sup>17</sup><sup> </sup>
    Increases in human cases of influenza A (H5N1) have been observed<sup> </sup>during cooler months in association with increases in outbreaks<sup> </sup>among poultry (see Fig. 1 of the Supplementary Appendix).<sup>18</sup><sup> </sup>However, because cases have occurred year-round, clinicians<sup> </sup>must be alert to possible human infection at any time, especially<sup> </sup>in countries with outbreaks of influenza A (H5N1) among birds.<sup> </sup>To date, no cases of influenza A (H5N1) illness have been identified<sup> </sup>among short-term travelers visiting countries affected by outbreaks<sup> </sup>among poultry or wild birds,<sup>19</sup> although clinicians in unaffected<sup> </sup>countries should consider this possibility in travelers with<sup> </sup>exposures to poultry.<sup> </sup>
    Surveillance for cases of influenza A (H5N1) has focused on<sup> </sup>patients with severe illness, but milder illnesses in children,<sup> </sup>which are not pneumonic,<sup>20</sup><sup>,</sup><sup>21</sup> occur. Limited seroepidemiologic<sup> </sup>studies conducted since 2003 involving villagers living with<sup> </sup>backyard poultry, workers in live-poultry markets, and health<sup> </sup>care workers suggest that asymptomatic or mild human influenza<sup> </sup>A (H5N1) virus infection is rare (Table 1 of the Supplementary Appendix).<sup>14</sup><sup> </sup>
    Transmission
    Direct avian-to-human H5N1 virus transmission is the predominant<sup> </sup>means of human infection, although the exact mode and sites<sup> </sup>of influenza A (H5N1) virus acquisition in the respiratory tract<sup> </sup>are incompletely understood. Handling of sick or dead poultry<sup> </sup>during the week before the onset of illness is the most commonly<sup> </sup>recognized risk factor.<sup>22</sup><sup>,</sup><sup>23</sup> Most patients have acquired A (H5N1)<sup> </sup>infection from poultry raised inside or outside their houses.<sup> </sup>Slaughtering, defeathering, or preparing sick poultry for cooking;<sup> </sup>playing with or holding diseased or dead poultry; handling fighting<sup> </sup>cocks or ducks that appear to be well; and consuming raw or<sup> </sup>undercooked poultry or poultry products have all been implicated<sup> </sup>as potential risk factors.<sup>21</sup><sup>,</sup><sup>22</sup><sup>,</sup><sup>23</sup><sup>,</sup><sup>24</sup> The defeathering of dead<sup> </sup>wild swans was implicated in one case cluster.<sup>25</sup><sup> </sup>
    The influenza A (H5N1) virus can also infect multiple mammalian<sup> </sup>hosts,<sup>26</sup><sup>,</sup><sup>27</sup> including domestic cats<sup>28</sup> and dogs.<sup>29</sup> None have<sup> </sup>been implicated in influenza A (H5N1) virus transmission to<sup> </sup>humans yet, but any animal infected with the virus theoretically<sup> </sup>poses a risk of transmission and of being a host for viral adaptation<sup> </sup>to mammals.<sup>26</sup><sup> </sup>
    Clusters of human influenza A (H5N1) illness with at least two<sup> </sup>epidemiologically linked cases have been identified in 10 countries<sup> </sup>and have accounted for approximately one quarter of cases.<sup>20</sup><sup>,</sup><sup>21</sup><sup>,</sup><sup>24</sup><sup>,</sup><sup>30</sup><sup>,</sup><sup>31</sup><sup>,</sup><sup>32</sup><sup> </sup>Most clusters have involved two or three persons; the largest<sup> </sup>affected eight. More than 90% of case clusters have occurred<sup> </sup>among blood-related family members, suggesting possible genetic<sup> </sup>susceptibility, although one statistical model indicated that<sup> </sup>these clusters might have occurred because of chance alone.<sup>33</sup><sup> </sup>Most persons in case clusters probably acquired infection from<sup> </sup>common-source exposures to poultry, but limited, nonsustained<sup> </sup>human-to-human transmission has probably occurred during very<sup> </sup>close, unprotected contact with a severely ill patient.<sup>20</sup><sup>,</sup><sup>30</sup><sup>,</sup><sup>32</sup><sup> </sup>In the largest cluster, transmission probably occurred from<sup> </sup>the index case to six blood-related family members and subsequently<sup> </sup>to another family member.<sup>32</sup> Respiratory secretions and all bodily<sup> </sup>fluids, including feces, should be considered potentially infectious.<sup> </sup>
    In one quarter or more of patients with influenza A (H5N1) virus<sup> </sup>infection, the source of exposure is unclear, and environment-to-human<sup> </sup>transmission remains possible.<sup>20</sup><sup>,</sup><sup>24</sup> For some patients, the only<sup> </sup>identified risk factor was visiting a live-poultry market.<sup>34</sup><sup>,</sup><sup>35</sup><sup> </sup>Plausible transmission routes include contact with virus-contaminated<sup> </sup>fomites or with fertilizer containing poultry feces, followed<sup> </sup>by self-inoculation of the respiratory tract or inhalation of<sup> </sup>aerosolized infectious excreta. It is unknown whether influenza<sup> </sup>A (H5N1) virus infection can begin in the human gastrointestinal<sup> </sup>tract. In several patients, diarrheal disease preceded respiratory<sup> </sup>symptoms,<sup>36</sup> and virus has been detected in feces.<sup>3</sup><sup>,</sup><sup>37</sup> Acquisition<sup> </sup>of influenza A (H5N1) virus infection in the gastrointestinal<sup> </sup>tract has been implicated in other mammals.<sup>26</sup> Drinking potable<sup> </sup>water and eating properly cooked foods are not considered to<sup> </sup>be risk factors, but ingestion of virus-contaminated products<sup> </sup>or swimming or bathing in virus-contaminated water might pose<sup> </sup>a risk.<sup> </sup>
    Incubation Period
    After exposure to infected poultry, the incubation period generally<sup> </sup>appears to be 7 days or less, and in many cases this period<sup> </sup>is 2 to 5 days. In clusters in which limited, human-to-human<sup> </sup>transmission has probably occurred, the incubation period appears<sup> </sup>to be approximately 3 to 5 days, although in one cluster it<sup> </sup>was estimated to be 8 to 9 days.<sup>20</sup><sup>,</sup><sup>30</sup><sup> </sup>
    Pathogenesis
    Viral Factors
    The viral and host factors that determine host-restriction and<sup> </sup>disease manifestations are incompletely understood.<sup>38</sup> Preferential<sup> </sup>binding of the influenza A (H5N1) virus to 2,3-linked sialic<sup> </sup>acid receptors on avian cells<sup>39</sup> is thought to be key in preventing<sup> </sup>influenza A (H5N1) and other avian influenza viruses from readily<sup> </sup>infecting humans. Some influenza A (H5N1) viruses isolated from<sup> </sup>humans have acquired mutations that permit binding to both 2,3-linked<sup> </sup>sialic acid receptors and 2,6-linked sialic acid receptors,<sup>40</sup><sup> </sup>but these mutations appear to be insufficient for efficient<sup> </sup>human-to-human transmission. To date, influenza A (H5N1) viruses<sup> </sup>have shown no transmissibility or poor transmissibility between<sup> </sup>ferrets and between swine, and reassortment between an influenza<sup> </sup>A (H5N1) virus and an influenza A (H3N2) virus did not confer<sup> </sup>transmissibility in ferrets.<sup>41</sup> Changes in multiple viral genes<sup> </sup>are probably required to generate a potentially pandemic influenza<sup> </sup>A (H5N1) virus.<sup> </sup>
    All recent influenza A (H5N1) viruses retain a polybasic amino<sup> </sup>acid motif at the HA1?HA2 connecting peptide that is characteristic<sup> </sup>of highly pathogenic avian influenza viruses. Geographic variations<sup> </sup>in this motif have not been associated with obvious changes<sup> </sup>in the virulence of infection in humans. Amino acid substitutions<sup> </sup>in the polymerase basic protein 2 (PB2) gene are associated<sup> </sup>with mammalian adaptation, virulence in mice, and replication<sup> </sup>at temperatures present in the upper respiratory tract (Figure 1).<sup>42</sup><sup> </sup>However, these mutations do not correlate with obvious differences<sup> </sup>in mortality among humans with this viral infection.<sup>3</sup><sup>,</sup><sup>21</sup><sup> </sup>
    Viral Replication
    The primary pathologic process that causes death is fulminant<sup> </sup>viral pneumonia. The target cells for replication of the influenza<sup> </sup>A (H5N1) virus include type 2 alveolar pneumocytes and macrophages.<sup>17</sup><sup>,</sup><sup>43</sup><sup>,</sup><sup>44</sup><sup> </sup>Bronchiolar and alveolar cells, but not epithelia from the trachea<sup> </sup>or upper respiratory tract, express detectable 2,3-linked sialic<sup> </sup>acid receptors.<sup>43</sup><sup>,</sup><sup>44</sup><sup>,</sup><sup>45</sup> However, influenza A (H5N1) viruses<sup> </sup>replicate in ex vivo organ cultures of the upper respiratory<sup> </sup>tract,<sup>44</sup> postmortem studies show virus in tracheal epithelia,<sup>17</sup><sup>,</sup><sup>46</sup><sup> </sup>and high titers of virus are detectable in specimens of throat<sup> </sup>and tracheal aspirates from humans infected with influenza A<sup> </sup>(H5N1) virus.<sup>3</sup> These findings suggest that the initial infection<sup> </sup>may occur in either the upper or lower respiratory tract, although<sup> </sup>the latter may support more efficient replication.<sup> </sup>
    Limited data show that patients with influenza A (H5N1) disease<sup> </sup>may have detectable viral RNA in the respiratory tract for up<sup> </sup>to 3 weeks, presumably because of negligible preexisting immunity<sup> </sup>and possibly viral evasion of immune responses.<sup>3</sup> One patient<sup> </sup>with fatal infection treated with both antiviral agents and<sup> </sup>corticosteroids had viral antigen and RNA in tracheal samples<sup> </sup>on day 27 after the onset of illness.<sup>17</sup> Viral loads in the pharynx<sup> </sup>are higher and plasma viral RNA is detected more often in patients<sup> </sup>with fatal disease than in those with nonfatal disease, indicating<sup> </sup>that levels of viral replication influence the outcome.<sup>3</sup> The<sup> </sup>reported presence of infectious virus in the blood, cerebrospinal<sup> </sup>fluid, or viscera of several patients with fatal disease indicates<sup> </sup>that, as in birds and several mammalian species, disseminated<sup> </sup>infection occurs in some humans.<sup>3</sup><sup>,</sup><sup>17</sup><sup>,</sup><sup>36</sup><sup>,</sup><sup>37</sup><sup>,</sup><sup>46</sup> A fatal influenza<sup> </sup>A (H5N1) infection in one pregnant woman who received corticosteroids<sup> </sup>for treatment of the disease was associated with virus infection<sup> </sup>of the brain, placenta, and fetus.<sup>17</sup> Infectious virus and viral<sup> </sup>RNA have been detected in feces and intestines, suggesting that<sup> </sup>the virus sometimes replicates in the gastrointestinal tract.<sup>1</sup><sup>,</sup><sup>3</sup><sup>,</sup><sup>36</sup><sup>,</sup><sup>37</sup><sup>,</sup><sup>46</sup><sup> </sup>
    Pathological Findings
    The few reported autopsies of patients with influenza A (H5N1)<sup> </sup>virus infection have shown diffuse alveolar damage with hyaline<sup> </sup>membrane formation, patchy interstitial lymphoplasmacytic infiltrates,<sup> </sup>bronchiolitis with squamous metaplasia, and pulmonary congestion<sup> </sup>with varying degrees of hemorrhage.<sup>17</sup><sup>,</sup><sup>46</sup><sup>,</sup><sup>47</sup> Acute exudative,<sup> </sup>diffuse alveolar damage with macrophages, neutrophils, and activated<sup> </sup>lymphocytes has been detected in patients who died within 2<sup> </sup>weeks after the onset of illness. Apoptosis in alveolar cells<sup> </sup>and infiltrating leukocytes are prominent findings.<sup>46</sup> Lymphocyte<sup> </sup>depletion occurs in the spleen, lymph nodes, and tonsils; histiocytic<sup> </sup>hyperplasia and reactive hemophagocytosis presumably result<sup> </sup>from host cytokine responses and viral infection. Edema and<sup> </sup>degeneration of myocytes in the heart and extensive acute tubular<sup> </sup>necrosis in the kidney have been observed.<sup> </sup>
    Host Responses
    Higher plasma levels of macrophage and neutrophil-attractant<sup> </sup>chemokines and both proinflammatory and antiinflammatory cytokines<sup> </sup>(interleukin-6, interleukin-10, and interferon-) have been observed<sup> </sup>in patients with influenza A (H5N1) virus infection ?<sup> </sup>particularly in patients with fatal infection ? than in<sup> </sup>patients with conventional influenza.<sup>3</sup> Plasma levels of cytokines<sup> </sup>and chemokines correlate positively with pharyngeal viral loads,<sup>3</sup><sup> </sup>suggesting that these responses are driven by high-level viral<sup> </sup>replication. In vitro experiments involving primary human macrophages<sup> </sup>and lung pneumocytes show differential up-regulation of multiple<sup> </sup>cytokines by influenza A (H5N1) virus as compared with human<sup> </sup>influenza viruses,<sup>48</sup> indicating that viral hyperinduction probably<sup> </sup>contributes to hypercytokinemia.<sup> </sup>
    In mouse models of influenza A (H5N1) virus infection, mice<sup> </sup>with deficient induction of interleukin-6, macrophage inflammatory<sup> </sup>protein 1, or tumor necrosis factor or its receptors<sup>49</sup><sup>,</sup><sup>50</sup> and<sup> </sup>mice treated with glucocorticoids,<sup>50</sup> had similar mortality as<sup> </sup>compared with wild-type animals; mice without interleukin-1<sup> </sup>receptors had increased mortality.<sup>49</sup> Tissue damage in human<sup> </sup>influenza A (H5N1) disease probably results from the combined<sup> </sup>effects of unrestrained viral infection and inflammatory responses<sup> </sup>induced by influenza A (H5N1) infection. Knowledge of the mechanisms<sup> </sup>of hypercytokinemia is insufficient to guide safe, rational<sup> </sup>immunomodulatory treatment at present.<sup> </sup>
    Clinical Features
    Currently, illness due to influenza A (H5N1) viruses typically<sup> </sup>manifests as severe pneumonia that often progresses rapidly<sup> </sup>to the acute respiratory distress syndrome. The time from the<sup> </sup>onset of illness to presentation (median, 4 days) or to death<sup> </sup>(median, 9 to 10 days) has remained unchanged from 2003 through<sup> </sup>2006 (Table 1).<sup>16</sup> Observed differences in mortality among patients<sup> </sup>with presumed clade 1 and clade 2 virus infections (Table 1<sup> </sup>and Table 2)<sup>1</sup><sup>,</sup><sup>21</sup><sup>,</sup><sup>24</sup><sup>,</sup><sup>35</sup><sup>,</sup><sup>51</sup> are difficult to interpret because<sup> </sup>of variations in medical practices and the time from the onset<sup> </sup>of illness to treatment among affected countries.<sup> </sup>
    <!-- null --> <table cellpadding="0" cellspacing="0"><tbody><tr bgcolor="#e8e8d1"><td><table cellpadding="2" cellspacing="2"><tbody><tr bgcolor="#e8e8d1"><td align="center" bgcolor="#ffffff" valign="top"> View this table:
    <nobr>[in this window]
    [in a new window]

    </nobr> </td><td align="left" valign="top"> Table 1. Case Fatality Proportion According to Clade or Subclade and Median Time from Onset of Illness to Hospitalization or Death in Patients with Confirmed Influenza A (H5N1) Illness.
    </td></tr></tbody></table></td></tr></tbody></table>
    <!-- null --> <table cellpadding="0" cellspacing="0"><tbody><tr bgcolor="#e8e8d1"><td><table cellpadding="2" cellspacing="2"><tbody><tr bgcolor="#e8e8d1"><td align="center" bgcolor="#ffffff" valign="top"> View this table:
    <nobr>[in this window]
    [in a new window]

    </nobr> </td><td align="left" valign="top"> Table 2. Clinical and Common Laboratory Features of Influenza A (H5N1) Disease at Hospital Admission.
    </td></tr></tbody></table></td></tr></tbody></table>
    Febrile upper respiratory illnesses without pneumonia in children<sup> </sup>have been reported more frequently since 2005.<sup>20</sup><sup>,</sup><sup>21</sup> Early consultation<sup> </sup>and antiviral therapy may have altered the clinical course of<sup> </sup>these illnesses. Less frequent gastrointestinal symptoms have<sup> </sup>been reported since 2005 (Table 2), suggesting that some manifestations<sup> </sup>of clade 1 and 2 virus infections may differ from each other.<sup> </sup>Leukopenia, lymphopenia, mild-to-moderate thrombocytopenia,<sup> </sup>and elevated levels of aminotransferases are common but not<sup> </sup>universal (Table 2). Lymphopenia and increased levels of lactate<sup> </sup>dehydrogenase at presentation have been associated with a poor<sup> </sup>prognosis.<sup>1</sup><sup>,</sup><sup>3</sup><sup>,</sup><sup>21</sup><sup>,</sup><sup>37</sup> Other reported abnormalities include elevated<sup> </sup>levels of creatine phosphokinase, hypoalbuminemia, and increased<sup> </sup>d-dimer levels and changes indicative of disseminated intravascular<sup> </sup>coagulopathy.<sup>20</sup><sup>,</sup><sup>21</sup><sup> </sup>
    The nonspecific clinical presentation of influenza A (H5N1)<sup> </sup>disease has often resulted in misdiagnosis of subsequently confirmed<sup> </sup>cases (Table 3); influenza A (H5N1) virus infection has been<sup> </sup>suspected in only a small number of patients. Health care staff<sup> </sup>should include influenza A (H5N1) virus infection in the differential<sup> </sup>diagnosis for patients who present with epidemiologic risk factors<sup> </sup>and unusual courses of illness, especially rapidly progressing<sup> </sup>pneumonia (see Fig. 2 of the Supplementary Appendix).<sup> </sup>
    <!-- null --> <table cellpadding="0" cellspacing="0"><tbody><tr bgcolor="#e8e8d1"><td><table cellpadding="2" cellspacing="2"><tbody><tr bgcolor="#e8e8d1"><td align="center" bgcolor="#ffffff" valign="top"> View this table:
    <nobr>[in this window]
    [in a new window]

    </nobr> </td><td align="left" valign="top"> Table 3. Initial Diagnosis in Patients with Confirmed Influenza A (H5N1) Virus Infection.
    </td></tr></tbody></table></td></tr></tbody></table>
    Laboratory Diagnosis
    Detection of viral RNA by means of conventional or real-time<sup> </sup>reverse-transcriptase polymerase chain reaction remains the<sup> </sup>best method for the initial diagnosis of influenza A (H5N1).<sup>52</sup><sup> </sup>These assays can provide results within 4 to 6 hours and can<sup> </sup>be performed under biosafety level 2 conditions. The genetic<sup> </sup>variability of influenza A (H5N1) viruses<sup>7</sup><sup>,</sup><sup>8</sup> calls for frequent<sup> </sup>updating of primers and probes. Consequently, access to sequences<sup> </sup>from recent influenza A (H5N1) viral isolates is essential.<sup> </sup>To detect other influenza A virus infections and reduce false<sup> </sup>negative results due to mutations in the H5 hemagglutinin gene,<sup> </sup>a conserved influenza A gene (e.g., matrix or nucleoprotein)<sup> </sup>should also be targeted.<sup> </sup>
    Diagnostic yields are higher with throat specimens than with<sup> </sup>nasal swabs because of higher viral loads of influenza A (H5N1)<sup> </sup>in the throat.<sup>1</sup><sup>,</sup><sup>3</sup> However, nasal swabs are useful for detecting<sup> </sup>human influenza viruses, so collection of both specimens is<sup> </sup>recommended. If they are available, tracheal aspirates have<sup> </sup>higher viral titers and yields than specimens from the upper<sup> </sup>respiratory tract.<sup>3</sup> Negative results in single respiratory specimens<sup> </sup>do not rule out influenza A (H5N1) virus infection,<sup>21</sup> and repeated<sup> </sup>collection of multiple specimen types is recommended.<sup>52</sup> Previous<sup> </sup>antiviral treatment may reduce the diagnostic yield. Detection<sup> </sup>of influenza A (H5N1) viral RNA in feces or blood may provide<sup> </sup>prognostic information,<sup>3</sup> but it has lower diagnostic sensitivity<sup> </sup>than influenza A (H5N1) viral RNA in respiratory specimens.<sup> </sup>
    Commercially available rapid assays for influenza-antigen detection<sup> </sup>have poor clinical sensitivity for the detection of influenza<sup> </sup>A (H5N1) virus (Table 2 of the Supplementary Appendix),<sup>1</sup><sup>,</sup><sup>20</sup><sup>,</sup><sup>21</sup><sup> </sup>and they do not differentiate between human and avian subtypes<sup> </sup>of influenza A viruses. Although rapid antigen tests have similar<sup> </sup>analytic sensitivity for detecting human and avian influenza<sup> </sup>A (H5N1) viruses, they require 1000 times higher levels of virus<sup> </sup>than viral cultures to be positive.<sup>53</sup><sup> </sup>
    The detection of anti-H5 antibodies is essential for epidemiologic<sup> </sup>investigations and may provide retrospective diagnostic confirmation<sup> </sup>in patients. Seroconversion generally occurs 2 to 3 weeks after<sup> </sup>infection. Microneutralization assays are the most reliable<sup> </sup>methods for detecting antibodies to avian viruses, but they<sup> </sup>are labor-intensive and require biosafety level 3 facilities<sup> </sup>and appropriate strains of influenza A (H5N1) viruses. As compared<sup> </sup>with initial samples, elevations of four times or more or single<sup> </sup>titers of 1:80 or more in convalescent-phase samples are considered<sup> </sup>to be diagnostic.<sup>52</sup> Modified nonpathogenic influenza A (H5N1)<sup> </sup>virus generated by reverse genetics or lentivirus pseudotyped<sup> </sup>with H5 hemagglutinin<sup>54</sup> may provide alternatives for performing<sup> </sup>neutralization tests in biosafety level 2 facilities. Hemagglutination-inhibition<sup> </sup>assays with the use of horse erythrocytes show promising results<sup> </sup>but require further validation.<sup> </sup>
    Treatment
    Antiviral Agents
    Susceptibility to current antiviral agents varies among circulating<sup> </sup>strains of influenza A (H5N1) viruses. Clade 1 viruses and most<sup> </sup>clade 2 viruses from Indonesia are fully resistant to M2 inhibitors,<sup> </sup>whereas clade 2 viruses from the lineages in other parts of<sup> </sup>Eurasia and Africa are usually susceptible (Klimov A: personal<sup> </sup>communication). As compared with influenza A (H5N1) viruses<sup> </sup>from 1997 or influenza A (H1N1) viruses in vitro,<sup>55</sup> clade 1<sup> </sup>viruses generally show enhanced susceptibility to oseltamivir<sup> </sup>carboxylate, but the high-level replication of some oseltamivir-susceptible<sup> </sup>strains requires higher doses or more prolonged administration,<sup> </sup>or both, in animal models.<sup>55</sup><sup>,</sup><sup>56</sup> Clade 1 viruses appear to be<sup> </sup>15 to 30 times more sensitive to oseltamivir than clade 2 isolates<sup> </sup>from Indonesia and Turkey,<sup>56</sup><sup>,</sup><sup>57</sup> although the possible clinical<sup> </sup>relevance of such differences in oseltamivir susceptibility<sup> </sup>remains to be determined. During oseltamivir therapy, the emergence<sup> </sup>of highly resistant variants with an H274Y neuraminidase mutation<sup> </sup>may be associated with a fatal outcome.<sup>58</sup> Infection by influenza<sup> </sup>A (H5N1) viruses containing an N294S mutation that causes a<sup> </sup>reduction in oseltamivir susceptibility by a factor of 12 to<sup> </sup>15 times was reported to be present in two Egyptian patients<sup> </sup>with fatal disease before therapy,<sup>59</sup> and avian influenza A (H5N1)<sup> </sup>viruses with reduced susceptibility to neuraminidase inhibitors<sup> </sup>are occasionally detected.<sup>60</sup><sup> </sup>
    Early treatment with oseltamivir is recommended,<sup>61</sup><sup>,</sup><sup>62</sup> and data<sup> </sup>from uncontrolled clinical trials suggest that it improves survival<sup> </sup>(Table 4), although the optimal dose and duration of therapy<sup> </sup>are uncertain. Mortality remains high despite administration<sup> </sup>of oseltamivir; late initiation of therapy appears to be a major<sup> </sup>factor. Uncontrolled viral replication, as reflected in the<sup> </sup>detection of persistent pharyngeal RNA after completion of standard<sup> </sup>therapy, is associated with a poor prognosis.<sup>58</sup> Higher levels<sup> </sup>of viral replication and slower clearance of infection probably<sup> </sup>occur in the lower respiratory tract.<sup>3</sup> The oral bioavailability<sup> </sup>of oseltamivir in patients with severe diarrhea or gastrointestinal<sup> </sup>dysfunction related to influenza A (H5N1) virus infection or<sup> </sup>those in whom the drug has been administered extemporaneously<sup> </sup>(e.g., by means of a nasogastric tube) is uncertain.<sup> </sup>
    <!-- null --> <table cellpadding="0" cellspacing="0"><tbody><tr bgcolor="#e8e8d1"><td><table cellpadding="2" cellspacing="2"><tbody><tr bgcolor="#e8e8d1"><td align="center" bgcolor="#ffffff" valign="top"> View this table:
    <nobr>[in this window]
    [in a new window]

    </nobr> </td><td align="left" valign="top"> Table 4. Effects of Treatment and Time to Treatment with Oseltamivir on Survival among Patients with Influenza A (H5N1) Infection.
    </td></tr></tbody></table></td></tr></tbody></table>
    A higher dose of oseltamivir (e.g., 150 mg twice daily in adults)<sup> </sup>and an increased duration of therapy, for a total of 10 days,<sup> </sup>may be reasonable, given the high levels of replication of the<sup> </sup>influenza A (H5N1) virus, observations of progressive disease<sup> </sup>despite early administration of standard-dose oseltamivir (75<sup> </sup>mg twice daily for 5 days in adults) within 1 to 3 days after<sup> </sup>the onset of the illness, and the proven safety of higher doses<sup> </sup>in adults with seasonal influenza, especially if there is pneumonic<sup> </sup>disease at presentation or evidence of clinical progression.<sup>62</sup><sup> </sup>In mouse models of amantadine-sensitive influenza A (H5N1) virus<sup> </sup>infection, as compared with monotherapy, the combination of<sup> </sup>oseltamivir and amantadine significantly increased survival<sup> </sup>rates and inhibited viral replication in the internal organs.<sup>64</sup><sup> </sup>No adverse pharmacologic interactions have been shown in humans.<sup>65</sup><sup> </sup>In areas where influenza A (H5N1) viruses are likely to be susceptible<sup> </sup>to amantadine, combination treatment with oseltamivir would<sup> </sup>be reasonable, especially in seriously ill patients.<sup> </sup>
    Although zanamivir is active against oseltamivir-resistant variants<sup> </sup>with N1 neuraminidase mutations at H274Y<sup>66</sup> or N294S, the value<sup> </sup>of inhaled zanamivir has not been studied in human influenza<sup> </sup>A (H5N1) disease. Suboptimal delivery to sites of infection<sup> </sup>in patients with pneumonic or extrapulmonary disease is a concern.<sup> </sup>Parenteral delivery of zanamivir or the neuraminidase inhibitor<sup> </sup>peramivir results in antiviral activity in animal models of<sup> </sup>influenza A (H5N1) virus infection; these agents and others<sup> </sup>are under clinical development (Table 3 of the Supplementary Appendix).<sup> </sup>
    Other Treatments
    Supportive care with correction of hypoxemia and treatment of<sup> </sup>nosocomial complications remains fundamental in the management<sup> </sup>of influenza A (H5N1) disease.<sup>2</sup><sup>,</sup><sup>62</sup> Corticosteroids should not<sup> </sup>be used routinely.<sup>62</sup> Corticosteroid therapy has thus far not<sup> </sup>been shown to be effective in patients with influenza A (H5N1)<sup> </sup>virus infection,<sup>1</sup> and prolonged or high-dose corticosteroid<sup> </sup>therapy can result in serious adverse events, including opportunistic<sup> </sup>infections such as central nervous system toxoplasmosis (Soeroso<sup> </sup>S: unpublished data). In northern Vietnam, mortality was 59%<sup> </sup>among 29 recipients of corticosteroids, as compared with 24%<sup> </sup>among 38 persons who did not receive corticosteroids (P=0.004)<sup> </sup>(Cao T, Thanh Liem N: personal communication). The possible<sup> </sup>value of other immunomodulators remains to be determined.<sup> </sup>
    Prevention
    Avian influenza A viruses are readily inactivated by a variety<sup> </sup>of chemical agents and physical conditions, including soaps,<sup> </sup>detergents, alcohols, and chlorination.<sup>67</sup><sup>,</sup><sup>68</sup> Guidelines for<sup> </sup>the prevention of infection with influenza A (H5N1) virus in<sup> </sup>various risk groups, including poultry workers, travelers, and<sup> </sup>health care workers, are available from the U.S. Centers for<sup> </sup>Disease Control and Prevention and the WHO.<sup> </sup>
    Antiviral Chemoprophylaxis
    WHO guidelines for the use of antiviral agents for prophylaxis<sup> </sup>in persons who have been exposed to influenza A (H5N1) viruses<sup> </sup>in the current pandemic-alert period have been published.<sup>61</sup><sup> </sup>Mathematical models of an emerging outbreak of influenza A (H5N1)<sup> </sup>in rural Asia predict that a strategy of mass, targeted antiviral<sup> </sup>chemoprophylaxis and social-distancing measures might extinguish<sup> </sup>or delay pandemic spread of the virus. The WHO has a stockpile<sup> </sup>of oseltamivir for this purpose and is working with partners<sup> </sup>for implementation of its distribution in the event of an outbreak.<sup>69</sup><sup> </sup>
    Immunization
    Safe and immunogenic inactivated H5 vaccines have been developed.<sup>6</sup><sup> </sup>Reverse genetics permits the rapid generation of seed viruses<sup> </sup>with attenuated virulence, but the changing antigenicity of<sup> </sup>circulating strains of influenza A (H5N1) viruses calls for<sup> </sup>new candidate vaccines from different lineages<sup>6</sup> and the development<sup> </sup>of vaccines that elicit cross-clade immunogenicity. H5 hemagglutinin<sup> </sup>appears to be a weak human immunogen. For subvirion vaccines<sup> </sup>without adjuvants, persons who have not received a priming dose<sup> </sup>require two doses with a high hemagglutinin antigen content<sup> </sup>(Table 4 of the Supplementary Appendix). As compared with conventional<sup> </sup>subunit vaccines, certain oil-in-water adjuvant agents<sup>6</sup><sup>,</sup><sup>70</sup><sup>,</sup><sup>71</sup><sup> </sup>or the use of whole-virus H5N1 vaccines<sup>6</sup><sup>,</sup><sup>72</sup><sup>,</sup><sup>73</sup> can substantially<sup> </sup>reduce the amount of vaccine antigen required to induce immune<sup> </sup>responses in persons who have not received a priming dose, and<sup> </sup>they can induce immune responses to antigenically drifted viruses.<sup> </sup>However, the specific adjuvant, formulation, dose, stability,<sup> </sup>and ratio with the antigen are important variables that require<sup> </sup>clinical testing for each candidate vaccine. Alum adjuvants<sup> </sup>have not consistently improved the responses to H5 vaccines,<sup>6</sup><sup>,</sup><sup>73</sup><sup>,</sup><sup>74</sup><sup> </sup>whereas certain proprietary adjuvants (e.g., MF59 and AS03)<sup> </sup>appear to be highly effective and allow for considerable antigen-sparing<sup> </sup>and cross-reactive antibody responses.<sup>6</sup><sup>,</sup><sup>70</sup><sup>,</sup><sup>71</sup> These adjuvants<sup> </sup>have also been associated with increased rates of local and<sup> </sup>sometimes systemic reactogenicity.<sup> </sup>
    The antibody levels required for protection against human influenza<sup> </sup>A (H5N1) illness are unclear. The durability of antibody responses<sup> </sup>is limited, but boosting with a homologous vaccine<sup>70</sup> or virus<sup> </sup>vaccine with viral antigen from another clade<sup>75</sup> appears to be<sup> </sup>effective in persons who have received two priming doses. Prepriming<sup> </sup>might allow single doses of a homologous vaccine to be sufficient<sup> </sup>for an antigenically drifted pandemic virus. However, decisions<sup> </sup>regarding the use of vaccine before a pandemic and stockpiling<sup> </sup>require complex risk?benefit and cost?benefit analyses<sup> </sup>that include effects on the seasonal capacity of vaccine production,<sup> </sup>because the timing and cause of the next influenza pandemic<sup> </sup>are unknown, and it is unclear whether immunization of large<sup> </sup>populations could have adverse consequences.<sup> </sup>
    Initial studies in children and elderly persons suggest that<sup> </sup>antibody responses to subvirion vaccines at high doses (45 or<sup> </sup>90 ?g) are similar to those in young adults. Approximately<sup> </sup>15 to 20% of older adults have some baseline neutralizing antibodies<sup> </sup>to H5N1 virus and may have a response to a single dose.<sup>6</sup> The<sup> </sup>mechanisms leading to these antibodies are uncertain. Other<sup> </sup>studies to date have shown that intradermal H5 vaccines at low<sup> </sup>doses are poorly immunogenic and may be associated with injection-site<sup> </sup>reactions.<sup>6</sup> Intranasal live attenuated H5 vaccines are highly<sup> </sup>effective in animal models,<sup>76</sup> but they show a variable ability<sup> </sup>to replicate in humans and to induce immune responses. Various<sup> </sup>investigational approaches, including conserved antigen vaccines,<sup> </sup>vectored H5 vaccines, and other adjuvants, are being explored.<sup> </sup>
    <sup> </sup>
    <sup> </sup>
    <sup> </sup>
    Dr. Chotpitayasunondh reports receiving grant support from Sanofi<sup> </sup>Pasteur and lecture fees from Sanofi Pasteur, GlaxoSmithKline,<sup> </sup>and Merck; and Dr. Peiris, consulting fees from GlaxoSmithKline<sup> </sup>and Novartis and travel expenses and lecture fees from Novartis,<sup> </sup>Roche, and Sanofi Pasteur. No other potential conflict of interest<sup> </sup>relevant to this article was reported.<sup> </sup>
    Two authors (Drs. Hayden and Shindo) are staff members of the<sup> </sup>WHO. The authors alone are responsible for the views expressed<sup> </sup>in this article, and they do not necessarily represent the decisions<sup> </sup>or the stated policy of the WHO. The views expressed in this<sup> </sup>article do not necessarily reflect those of the other organizations<sup> </sup>whose staff participated in the WHO consultation.<sup> </sup>
    ? World Health Organization 2008. All rights reserved.<sup> </sup>Published with permission from the World Health Organization.<sup> </sup>
    We thank Drs. Christoph Steffen and Kaat Vandermaele of the<sup> </sup>WHO for their help with access to data and development of the<sup> </sup>management algorithm, Diane Ramm of the University of Virginia<sup> </sup>for her assistance in the preparation of an earlier version<sup> </sup>of the manuscript, and our colleagues in countries affected<sup> </sup>by A (H5N1) virus for their willingness to share unpublished<sup> </sup>clinical data for this article.<sup> </sup>
    <!-- null --> <sup>*</sup> Affiliations of the writing committee are listed in the Appendix.<sup> </sup>The participants in the meeting of the Second World Health Organization<sup> </sup>Consultation on Clinical Aspects of Human Infection with Avian<sup> </sup>Influenza A (H5N1) Virus, Antalya, Turkey, March 19?21,<sup> </sup>2007, are listed in the Supplementary Appendix, which is available<sup> </sup>with the full text of this article at www.nejm.org.<sup> </sup>

    Source Information
    The members of the writing committee (Abdel-Nasser Abdel-Ghafar, M.D., Tawee Chotpitayasunondh, M.D., Zhancheng Gao, M.D., Ph.D., Frederick G. Hayden, M.D., Nguyen Duc Hien, M.D., Ph.D., Menno D. de Jong, M.D., Ph.D., Azim Naghdaliyev, M.D., J.S. Malik Peiris, M.D., Nahoko Shindo, M.D., Santoso Soeroso, M.D., and Timothy M. Uyeki, M.D.) assume responsibility for the overall content and integrity of the article.
    Address reprint requests to Dr. Hayden at the Global Influenza Program, Department of Epidemic and Pandemic Alert and Response, World Health Organization, 20 Ave. Appia, Ch-1211, Geneva 27, Switzerland, or at haydenf@who.int<script type="text/javascript"><!-- var u = "haydenf", d = "who.int"; document.getElementById("em0").innerHTML = '<a href="mailto:' + u + '@' + d + '">' + u + '@' + d + '<\/a>'//--></script>.
    References
    1. <!-- null -->
    2. The Writing Committee of the World Health Organization (WHO) Consultation on Human Influenza A/H5. Avian influenza A (H5N1) infection in humans. N Engl J Med 2005;353:1374-1385. [Erratum, N Engl J Med 2006;354:884.]<!-- HIGHWIRE ID="358:3:261:1" --> <nobr>[Free Full Text]</nobr><!-- /HIGHWIRE --><!-- null -->
    3. World Health Organization. Summary of the second WHO consultation on clinical aspects of human infection with avian influenza A (H5N1) virus. (Accessed December 20, 2007, at http://www.who.int/csr/disease/avian.../en/index.html.)<!-- HIGHWIRE ID="358:3:261:2" --><!-- /HIGHWIRE --><!-- null -->
    4. de Jong M, Simmons CP, Thanh TT, et al. Fatal outcome of human influenza A (H5N1) is associated with high viral load and hypercytokinemia. Nat Med 2006;12:1203-1207.<!-- HIGHWIRE ID="358:3:261:3" --> [CrossRef][ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    5. Chen H, Smith GJD, Li KS, et al. Establishment of multiple sublineages of H5N1 influenza virus in Asia: implications for pandemic control. Proc Natl Acad Sci U S A 2006;103:2845-2850.<!-- HIGHWIRE ID="358:3:261:4" --> <nobr>[Free Full Text]</nobr><!-- /HIGHWIRE --><!-- null -->
    6. Webster RG, Govorkova EA. H5N1 Influenza -- continuing evolution and spread. N Engl J Med 2006;355:2174-2177.<!-- HIGHWIRE ID="358:3:261:5" --> <nobr>[Free Full Text]</nobr><!-- /HIGHWIRE --><!-- null -->
    7. World Health Organization. Third WHO meeting on evaluation of pandemic influenza prototype vaccines in clinical trials, Geneva, 15-16 February 2007. (Accessed December 20, 2007, at http://www.who.int/vaccine_research/...ing_150207/en/.)<!-- HIGHWIRE ID="358:3:261:6" --><!-- /HIGHWIRE --><!-- null -->
    8. Idem. Antigenic and genetic characteristics of H5N1 viruses and candidate H5N1 vaccine viruses developed for potential use as pre-pandemic vaccines. March 2007. (Accessed December 20, 2007, at http://www.who.int/csr/disease/avian.../en/index.html.)<!-- HIGHWIRE ID="358:3:261:7" --><!-- /HIGHWIRE --><!-- null -->
    9. Smith GJD, Fan XH, Wang J, et al. Emergence and predominance of an H5N1 influenza variant in China. Proc Natl Acad Sci U S A 2006;103:16936-16941.<!-- HIGHWIRE ID="358:3:261:8" --> <nobr>[Free Full Text]</nobr><!-- /HIGHWIRE --><!-- null -->
    10. Gauthier-Clerc M, Lebarbenchon C, Thomas F. Recent expansion of highly pathogenic avian influenza H5N1: a critical review. Ibis 2007;149:202-14.<!-- HIGHWIRE ID="358:3:261:9" --><!-- /HIGHWIRE --><!-- null -->
    11. Kilpatrick AM, Chmura AA, Gibbons DW, Fleischer RC, Marra PP, Daszak P. Predicting the global spread of H5N1 avian influenza. Proc Natl Acad Sci U S A 2006;103:19368-19373.<!-- HIGHWIRE ID="358:3:261:10" --> <nobr>[Free Full Text]</nobr><!-- /HIGHWIRE --><!-- null -->
    12. Ducatez MF, Olinger CM, Owoade AA, et al. Avian flu: multiple introductions of H5N1 in Nigeria. Nature 2006;442:37-37.<!-- HIGHWIRE ID="358:3:261:11" --> [CrossRef][Medline]<!-- /HIGHWIRE --><!-- null -->
    13. Winker K, McCracken KG, Gibson D, et al. Movements of birds and avian influenza from Asia into Alaska. Emerg Infect Dis 2007;13:547-552.<!-- HIGHWIRE ID="358:3:261:12" --> [ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    14. Fielding R, Bich TH, Quang LN, et al. Live poultry exposures, Hong Kong and Hanoi, 2006. Emerg Infect Dis 2007;13:1065-1067.<!-- HIGHWIRE ID="358:3:261:13" --> [ISI]<!-- /HIGHWIRE --><!-- null -->
    15. Vong S, Coghlan B, Mardy S, et al. Low frequency of poultry-to-human H5N1 virus transmission, Southern Cambodia, 2005. Emerg Infect Dis 2006;12:1542-1547.<!-- HIGHWIRE ID="358:3:261:14" --> [ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    16. World Health Organization. Cumulative number of confirmed human cases of avian influenza A/(H5N1) reported to WHO. (Accessed December 20, 2007, at http://www.who.int/csr/disease/avian.../en/index.html.)<!-- HIGHWIRE ID="358:3:261:15" --><!-- /HIGHWIRE --><!-- null -->
    17. Update: WHO-confirmed human cases of avian influenza A(H5N1) infection, 25 November 2003-24 November 2006. Wkly Epidemiol Rec 2007;82:41-48.www.who.int/wer/2007/wer8206.pdf<!-- HIGHWIRE ID="358:3:261:16" --> <!-- /HIGHWIRE --><!-- null -->
    18. Gu J, Xie Z, Gao Z, et al. H5N1 infection of the respiratory tract and beyond: a molecular pathology study. Lancet 2007;370:1137-1145.<!-- HIGHWIRE ID="358:3:261:17" --> [CrossRef][ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    19. Park AW, Glass K. Dynamic patterns of avian and human influenza in east and southeast Asia. Lancet Infect Dis 2007;7:543-548.<!-- HIGHWIRE ID="358:3:261:18" --> [CrossRef][ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    20. Ortiz JR, Wallis TR, Katz MA, et al. No evidence of avian influenza A (H5N1) among returning US travelers. Emerg Infect Dis 2007;13:294-297.<!-- HIGHWIRE ID="358:3:261:19" --> [ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    21. Kandun IN, Wibisono H, Sedyaningsih ER, et al. Three Indonesian clusters of H5N1 virus infection in 2005. N Engl J Med 2006;355:2186-2194.<!-- HIGHWIRE ID="358:3:261:20" --> <nobr>[Free Full Text]</nobr><!-- /HIGHWIRE --><!-- null -->
    22. Oner AF, Bay A, Arslan S, et al. Avian influenza A (H5N1) infection in eastern Turkey in 2006. N Engl J Med 2006;355:2179-2185.<!-- HIGHWIRE ID="358:3:261:21" --> <nobr>[Free Full Text]</nobr><!-- /HIGHWIRE --><!-- null -->
    23. Dinh PN, Long HT, Tien NTK, et al. Risk factors for human infection with avian influenza A H5N1, Vietnam, 2004. Emerg Infect Dis 2006;12:1841-1847.<!-- HIGHWIRE ID="358:3:261:22" --> [ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    24. Areechokchai D, Jiraphongsa C, Laosiritaworn Y, Hanshaoworakul W, O'Reilly M. Investigation of avian influenza (H5N1) outbreak in humans -- Thailand, 2004. MMWR Morb Mortal Wkly Rep 2006;55:Suppl 1:3-6.<!-- HIGHWIRE ID="358:3:261:23" --> <!-- /HIGHWIRE --><!-- null -->
    25. Sedyaningsih ER, Isfandari S, Setiawaty V, et al. Epidemiology of cases of H5N1 virus infection in Indonesia, July 2005-June 2006. J Infect Dis 2007;196:522-527.<!-- HIGHWIRE ID="358:3:261:24" --> [CrossRef][ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    26. Human avian influenza in Azerbaijan, February-March 2006. Wkly Epidemiol Rec 2007;81:183-188.www.who.int/wer/2006/wer8118.pdf<!-- HIGHWIRE ID="358:3:261:25" --> <!-- /HIGHWIRE --><!-- null -->
    27. Thiry E, Zicola A, Addie D, et al. Highly pathogenic avian influenza H5N1 virus in cats and other carnivores. Vet Microbiol 2007;122:25-31.<!-- HIGHWIRE ID="358:3:261:26" --> [CrossRef][ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    28. Mumford E, Bishop J, Hendrickx S, Embarek PB, Perdue M. Avian influenza H5N1: Risks at the human-animal interface. Food Nutr Bull 2007;28:Suppl:S357-S363.<!-- HIGHWIRE ID="358:3:261:27" --> [ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    29. Leschnik M, Weikel J, M?stl K, et al. Subclinical infection with avian influenza A (H5N1) virus in cats. Emerg Infect Dis 2007;13:243-247.<!-- HIGHWIRE ID="358:3:261:28" --> [ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    30. Songserm T, Amonsin A, Jam-on R, et al. Fatal avian influenza A H5N1 in a dog. Emerg Infect Dis 2006;12:1744-1747.<!-- HIGHWIRE ID="358:3:261:29" --> [ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    31. Ungchusak K, Auewarakul P, Dowell SF, et al. Probable person-to-person transmission of avian influenza A (H5N1). N Engl J Med 2005;352:333-340.<!-- HIGHWIRE ID="358:3:261:30" --> <nobr>[Free Full Text]</nobr><!-- /HIGHWIRE --><!-- null -->
    32. Olsen SJ, Ungchusak K, Sovann L, et al. Family clustering of avian influenza A (H5N1). Emerg Infect Dis 2005;11:1799-1801.<!-- HIGHWIRE ID="358:3:261:31" --> [ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    33. World Health Organization. Avian influenza ? situation in Indonesia ? update 16. 2006. (Accessed December 20, 2007, at http://www.who.int/csr/don/2006_05_31/en/print.html.)<!-- HIGHWIRE ID="358:3:261:32" --><!-- /HIGHWIRE --><!-- null -->
    34. Pitzer VE, Olsen SJ, Bergstrom CT, Dowell SF, Lipsitch M. Little evidence for genetic susceptibility to influenza A (H5N1) from family clustering data. Emerg Infect Dis 2007;13:1074-1076.<!-- HIGHWIRE ID="358:3:261:33" --> [ISI]<!-- /HIGHWIRE --><!-- null -->
    35. Wang M, Di B, Zhou D-H, et al. Food markets with live birds as source of avian influenza. Emerg Infect Dis 2006;12:1773-1775.<!-- HIGHWIRE ID="358:3:261:34" --> [ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    36. Yu H, Feng Z, Zhang X, et al. Human influenza A (H5N1) cases, urban areas of People's Republic of China, 2005-2006. Emerg Infect Dis 2007;13:1061-1061.<!-- HIGHWIRE ID="358:3:261:35" --> [ISI]<!-- /HIGHWIRE --><!-- null -->
    37. de Jong MD, Cam BV, Qui PT, et al. Fatal avian influenza A (H5N1) in a child presenting with diarrhea followed by coma. N Engl J Med 2005;352:686-691.<!-- HIGHWIRE ID="358:3:261:36" --> <nobr>[Free Full Text]</nobr><!-- /HIGHWIRE --><!-- null -->
    38. Buchy P, Mardy S, Vong S, et al. Influenza A/H5N1 virus infection in humans in Cambodia. J Clin Virol 2007;39:164-168.<!-- HIGHWIRE ID="358:3:261:37" --> [CrossRef][ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    39. Peiris JSM, de Jong MD, Guan Y. Avian influenza virus (H5N1): a threat to human health. Clin Microbiol Rev 2007;20:243-267.<!-- HIGHWIRE ID="358:3:261:38" --> <nobr>[Free Full Text]</nobr><!-- /HIGHWIRE --><!-- null -->
    40. Stevens J, Blixt O, Tumpey TM, Taubenberger JK, Paulson JC, Wilson IA. Structure and receptor specificity of the hemagglutinin from an H5N1 influenza virus. Science 2006;312:404-410.<!-- HIGHWIRE ID="358:3:261:39" --> <nobr>[Free Full Text]</nobr><!-- /HIGHWIRE --><!-- null -->
    41. Yamada S, Suzuki Y, Suzuki T, et al. Haemagglutinin mutations responsible for the binding of H5N1 influenza A viruses to human-type receptors. Nature 2006;444:378-382.<!-- HIGHWIRE ID="358:3:261:40" --> [CrossRef][Medline]<!-- /HIGHWIRE --><!-- null -->
    42. Maines TR, Chen LM, Matsuoka Y, et al. Lack of transmission of H5N1 avian-human reassortant influenza viruses in a ferret model. Proc Natl Acad Sci U S A 2006;103:12121-12126.<!-- HIGHWIRE ID="358:3:261:41" --> <nobr>[Free Full Text]</nobr><!-- /HIGHWIRE --><!-- null -->
    43. Hatta M, Hatta Y, Kim JH, et al. Growth of H5N1 influenza A viruses in the upper respiratory tracts of mice. PloS Pathog 2007;3:1374-9.<!-- HIGHWIRE ID="358:3:261:42" --><!-- /HIGHWIRE --><!-- null -->
    44. van Riel D, Munster VJ, de Wit E, et al. H5N1 virus attachment to lower respiratory tract. Science 2006;312:399-399.<!-- HIGHWIRE ID="358:3:261:43" --> <nobr>[Free Full Text]</nobr><!-- /HIGHWIRE --><!-- null -->
    45. Nicholls JM, Chan MCW, Chan WY, et al. Tropism of avian influenza A (H5N1) in the upper and lower respiratory tract. Nat Med 2007;13:147-149.<!-- HIGHWIRE ID="358:3:261:44" --> [CrossRef][ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    46. Shinya K, Ebina M, Yamada S, Ono M, Kasai N, Kawaoka Y. Avian flu: influenza virus receptors in the human airway. Nature 2006;440:435-436.<!-- HIGHWIRE ID="358:3:261:45" --> [CrossRef][Medline]<!-- /HIGHWIRE --><!-- null -->
    47. Uiprasertkul M, Kitphati TC, Pathavathana P, et al. Apoptosis and pathogenesis of avian influenza A (H5N1) virus in humans. Emerg Infect Dis 2007;13:708-712.<!-- HIGHWIRE ID="358:3:261:46" --> [ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    48. Ng WF, To KF, Lam WWL, Ng TK, Lee KC. The comparative pathology of severe acute respiratory syndrome and avian influenza A subtype H5N1 -- a review. Hum Pathol 2006;37:381-390.<!-- HIGHWIRE ID="358:3:261:47" --> [CrossRef][ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    49. Chan MC, Cheung CY, Chui WH, et al. Proinflammatory cytokine responses induced by influenza A (H5N1) viruses in primary human alveolar and bronchial epithelial cells. Respir Res 2005;6:135-135.<!-- HIGHWIRE ID="358:3:261:48" --> [CrossRef][Medline]<!-- /HIGHWIRE --><!-- null -->
    50. Szretter KJ, Gangappa S, Lu X, et al. Role of host cytokine responses in the pathogenesis of avian H5N1 influenza viruses in mice. J Virol 2007;81:2736-2744.<!-- HIGHWIRE ID="358:3:261:49" --> <nobr>[Free Full Text]</nobr><!-- /HIGHWIRE --><!-- null -->
    51. Salomon R, Hoffmann E, Webster RG. Inhibition of the cytokine response does not protect against lethal H5N1 influenza infection. Proc Natl Acad Sci U S A 2007;104:12479-12481.<!-- HIGHWIRE ID="358:3:261:50" --> <nobr>[Free Full Text]</nobr><!-- /HIGHWIRE --><!-- null -->
    52. Yu H, Shu Y, Hu S, et al. The first confirmed human case of avian influenza A (H5N1) in Mainland China. Lancet 2006;367:84-84.<!-- HIGHWIRE ID="358:3:261:51" --> [CrossRef][ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    53. World Health Organization. Collecting, preserving and shipping specimens for the diagnosis of avian influenza A (H5N1) virus infection: guide for field operations. (Accessed December 20, 2007, at http://www.who.int/csr/resources/pub...csredc2004.pdf.)<!-- HIGHWIRE ID="358:3:261:52" --><!-- /HIGHWIRE --><!-- null -->
    54. Chan KH, Lam SY, Puthavathana P, et al. Comparative analytical sensitivities of six rapid influenza A antigen detection test kits for detection of influenza A subtypes H1N1, H3N2 and H5N1. J Clin Virol 2007;38:169-171.<!-- HIGHWIRE ID="358:3:261:53" --> [CrossRef][ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    55. Nefkens I, Garcia JM, Ling CS, et al. Hemagglutinin pseudotyped lentiviral particles: characterization of a new method for avian H5N1 influenza sero-diagnosis. J Clin Virol 2007;39:27-33.<!-- HIGHWIRE ID="358:3:261:54" --> [CrossRef][ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    56. Yen HL, Monto AS, Webster RG, Govorkova EA. Virulence may determine the necessary duration and dosage of oseltamivir treatment for highly pathogenic A/Vietnam/1203/04 (H5N1) influenza virus in mice. J Infect Dis 2005;192:665-672.<!-- HIGHWIRE ID="358:3:261:55" --> [CrossRef][ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    57. Govorkova EA, Ilyushina NA, Boltz DA, Douglas A, Yilmaz N, Webster RG. Efficacy of oseltamivir therapy in ferrets inoculated with different clades of H5N1 influenza virus. Antimicrob Agents Chemother 2007;51:1414-1424.<!-- HIGHWIRE ID="358:3:261:56" --> <nobr>[Free Full Text]</nobr><!-- /HIGHWIRE --><!-- null -->
    58. McKimm-Breschkin J, Selleck P, Usman TB, Johnson M. Reduced sensitivity of influenza A (H5N1) to oseltamivir. Emerg Infect Dis 2007;13:1354-1357.<!-- HIGHWIRE ID="358:3:261:57" --> [ISI]<!-- /HIGHWIRE --><!-- null -->
    59. de Jong MD, Thanh TT, Khanh TH, et al. Oseltamivir resistance during treatment of influenza A (H5N1) infection. N Engl J Med 2005;353:2667-2672.<!-- HIGHWIRE ID="358:3:261:58" --> <nobr>[Free Full Text]</nobr><!-- /HIGHWIRE --><!-- null -->
    60. Saad MD, Boynton BR, Earhart KC, et al. Detection of oseltamivir resistance mutation N294S in humans with influenza A H5N1. In: Program and abstracts of the Options for the Control of Influenza Conference, Toronto, June 17?23, 2007:228. abstract.<!-- HIGHWIRE ID="358:3:261:59" --><!-- /HIGHWIRE --><!-- null -->
    61. Hurt AC, Selleck P, Komadina N, Shaw R, Brown L, Barr IG. Susceptibility of highly pathogenic A(H5N1) avian influenza viruses to the neuraminidase inhibitors and adamantanes. Antiviral Res 2007;73:228-231.<!-- HIGHWIRE ID="358:3:261:60" --> [CrossRef][ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    62. World Health Organization. WHO rapid advice guidelines on pharmacological management of humans infected with avian influenza A (H5N1) virus. 2006. (Accessed December 20, 2007, at http://www.who.int/medicines/publica...PAR_2006.6.pdf.)<!-- HIGHWIRE ID="358:3:261:61" --><!-- /HIGHWIRE --><!-- null -->
    63. Idem. Clinical management of human infection with avian influenza A (H5N1) virus. 2007. (Accessed December 20, 2007, at http://www.who.int/csr/disease/avian...nagement07.pdf.)<!-- HIGHWIRE ID="358:3:261:62" --><!-- /HIGHWIRE --><!-- null -->
    64. Sedyaningsih ER, Isfandari S, Setyaway V, et al. Clinical features of avian influenza A (H5N1) infection in Indonesia, July 2005?April 2007. In: Abstract book for the Options for the Control of Influenza VI Conference, Toronto, June 17?23, 2007:329.<!-- HIGHWIRE ID="358:3:261:63" --><!-- /HIGHWIRE --><!-- null -->
    65. Ilyushina NA, Hoffmann E, Solomon R, Webster RG, Govorkova EA. Amantadine-oseltamivir combination therapy for H5N1 influenza virus infection in mice. Antivir Ther 2007;12:363-370.<!-- HIGHWIRE ID="358:3:261:64" --> [ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    66. Morrison D, Roy S, Rayner C, et al. A randomized, crossover study to evaluate the pharmacokinetics of amantadine and oseltamivir administered alone and in combination. PLoS Clin Trials (in press).<!-- HIGHWIRE ID="358:3:261:65" --><!-- /HIGHWIRE --><!-- null -->
    67. Le QM, Kiso M, Someya K, et al. Avian flu: isolation of drug-resistant H5N1 virus. Nature 2005;437:1108-1108. [Erratum, Nature 2005;438:754.]<!-- HIGHWIRE ID="358:3:261:66" --> [CrossRef][Medline]<!-- /HIGHWIRE --><!-- null -->
    68. De Benedictis P, Beato MS, Capua I. Inactivation of avian influenza viruses by chemical agents and physical conditions: a review. Zoonoses Public Health 2007;54:51-68.<!-- HIGHWIRE ID="358:3:261:67" --><!-- /HIGHWIRE --><!-- null -->
    69. Rice EW, Adcock NJ, Sivaganesan M, Brown JD, Stallknecht DE, Swayne D. Chlorine inactivation of highly pathogenic avian influenza virus (H5N1). Emerg Infect Dis 2007;13:1568-1570.<!-- HIGHWIRE ID="358:3:261:68" --> [ISI]<!-- /HIGHWIRE --><!-- null -->
    70. World Health Organization. WHO interim protocol: rapid operations to contain the initial emergence of pandemic influenza. Updated October 2007. (Accessed December 20, 2007, at http://www.who.int/csr/disease/avian.../en/index.html.)<!-- HIGHWIRE ID="358:3:261:69" --><!-- /HIGHWIRE --><!-- null -->
    71. Stephenson I, Bugarini R, Nicholson KG, et al. Cross-reactivity to highly pathogenic avian influenza H5N1 viruses after vaccination with nonadjuvanted and MF59-adjuvanted influenza A/Duck/Singapore/97 (H5N3) vaccine: a potential priming strategy. J Infect Dis 2005;191:1210-1215.<!-- HIGHWIRE ID="358:3:261:70" --> [CrossRef][ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    72. Leroux-Roels I, Borkowski A, Vanwolleghem T, et al. Antigen sparing and cross-reactive immunity with an adjuvanted rH5N1 prototype pandemic influenza vaccine: a randomised controlled trial. Lancet 2007;370:580-589.<!-- HIGHWIRE ID="358:3:261:71" --> [CrossRef][ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    73. Lin J, Zhang J, Dong X, et al. Safety and immunogenicity of an inactivated adjuvanted whole-virion influenza A (H5N1) vaccine: a phase I randomised controlled trial. Lancet 2006;368:991-997.<!-- HIGHWIRE ID="358:3:261:72" --> [CrossRef][ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    74. Barrett N. Safety and immunogenicity of a cell culture (vero) derived whole virus H5N1 vaccine: a Phase I/II dose escalation study. In: Program of the IX International Symposium on Respiratory Viral Infections, Hong Kong, March 3?6, 2007.<!-- HIGHWIRE ID="358:3:261:73" --><!-- /HIGHWIRE --><!-- null -->
    75. Bresson JL, Perronne C, Launay O, et al. Safety and immunogenicity of an inactivated split-virion influenza A/Vietnam/1194/2004 (H5N1) vaccine: phase I randomised trial. Lancet 2006;367:1657-1664.<!-- HIGHWIRE ID="358:3:261:74" --> [CrossRef][ISI][Medline]<!-- /HIGHWIRE --><!-- null -->
    76. Goji N, Nolan C, Hill H, Wolff M, Rowe T, Treanor J. Immune responses of healthy subjects to a single dose of intramuscular inactivated influenza A/Vietnam/1203/2004 (H5N1) vaccine after priming with an antigenic variant. In: Final program and abstracts of the 44th Annual Meeting of IDSA, Toronto, October 12?15, 2006:64.<!-- HIGHWIRE ID="358:3:261:75" --><!-- /HIGHWIRE --><!-- null -->
    77. Lu X, Edwards LE, Desheva JA, et al. Cross-protective immunity in mice induced by live-attenuated or inactivated vaccines against highly pathogenic influenza A (H5N1) viruses. Vaccine 2006;24:6588-6593.<!-- HIGHWIRE ID="358:3:261:76" --> [CrossRef][ISI][Medline]<!-- /HIGHWIRE -->
    Appendix The writing committee's affiliations are as follows: the Ministry<sup> </sup>of Health and Population, Cairo (A.-N.A.-G.); the Queen Sirikit<sup> </sup>National Institute of Child Health, Bangkok, Thailand (T.C.);<sup> </sup>the Peking University People's Hospital, Beijing (Z.G.); the<sup> </sup>Global Influenza Program, World Health Organization, Geneva<sup> </sup>(F.G.H., N.S.); the University of Virginia, Charlottesville<sup> </sup>(F.G.H.); the National Institute for Infectious and Tropical<sup> </sup>Diseases, Hanoi (N.D.H.); the Oxford University Clinical Research<sup> </sup>Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam<sup> </sup>(M.D.J.); the Abulfaz Karayev Children's Hospital No. 2, Baku,<sup> </sup>Azerbaijan (A.N.); the University of Hong Kong, Hong Kong (J.S.M.P.);<sup> </sup>the National Infectious Diseases Hospital, Jakarta, Indonesia<sup> </sup>(S.S.); and the Centers for Disease Control and Prevention,<sup> </sup>Atlanta (T.M.U.).



    </center>

  • #2
    Figure 1


    Figure 1. Evolution of the Hemagglutinin and Other Key Mutations Associated with Virulence or Drug Resistance in Avian Influenza A (H5N1) Virus.
    The phylogenetic tree is for the hemagglutinin gene of highly pathogenic avian influenza A (H5N1) viruses. The geographic distributions refer to avian isolates, and the tree is based on publicly available sequences. Clade 0 includes viruses that were first recognized to cause human infections in Hong Kong Special Administrative Region in 1997. Viruses from clades and subclades 0, 1, 2.1, 2.2, 2.3, and 7 have caused human disease. Clade 1 viruses predominated in Vietnam, Thailand, and Cambodia in the early phase of the outbreak (2004?2005), and clade 2.1 viruses are endemic in Indonesia. Clade 2.2 viruses were associated with a major outbreak of H5N1 disease in migratory birds in Qinghai Lake, China, and have since spread, causing avian disease in Central and South Asia, the Middle East, Europe, and Africa and human disease in western Asia, the Middle East, and Africa. Clade 2.3 has become dominant in southern China and has also been detected in adjacent countries. (Modified from the WHO Web site: www.who.int/csr/disease/avian_influenza/guidelines/nomenclature/en/index.html.) The influenza genome contains eight individual segments of RNA, several of which encode two proteins. Within clade 1 or clade 2.1 viruses, polymerase basic protein 2 (PB2) Glu627Lys is observed in some isolates of human viruses but not in avian viruses.<sup>3</sup> Some human clade 1 viruses without PB2 627Lys have PB2 701Asn; clade 2.2 viruses of both human and avian origin have PB2 Glu627Lys.<sup>4</sup> The importance of sequence variations in NS1, in which most influenza A (H5N1) viruses contain a carboxyl-terminus?sequence motif that mediates binding to various cellular proteins bearing a PDZ domain, remains to be determined.

    Comment


    • #3
      Table 1


      Table 1. Case Fatality Proportion According to Clade or Subclade and Median Time from Onset of Illness to Hospitalization or Death in Patients with Confirmed Influenza A (H5N1) Illness.

      Comment


      • #4
        Table 2


        Table 2. Clinical and Common Laboratory Features of Influenza A (H5N1) Disease at Hospital Admission.

        Comment


        • #5
          Table 3


          Table 3. Initial Diagnosis in Patients with Confirmed Influenza A (H5N1) Virus Infection.

          Comment


          • #6
            Table 4


            Table 4. Effects of Treatment and Time to Treatment with Oseltamivir on Survival among Patients with Influenza A (H5N1) Infection.

            Comment


            • #7
              Re: Table 2

              Originally posted by Florida1 View Post

              Table 2. Clinical and Common Laboratory Features of Influenza A (H5N1) Disease at Hospital Admission.

              I see glaring omissions in these clinical features of H5N1.

              There is no mention of meningitis/encephalitis symptoms or a rash. Is "depressed consciousness" a clinical evaluation? There have been confirmed H5N1 cases with neurological symptoms, including an inability to walk.

              These neurological symptoms should be noted.

              The 1918 virus had major neurological symptoms and these lingered for years if patients survived. See "encephalitis lethargica" at this supposed professional neurological association which does not even mention the 1918 flu pandemic has been repeatedly implicated in the disease:

              "The cause of encephalitis lethargica is unknown. Between 1917 to 1928, an epidemic of encephalitis lethargica spread throughout the world, but no recurrence of the epidemic has since been reported"




              I also believe the records of human clusters and sources of infection need further analysis.

              Can anyone analyze this article and summarize what it says is definitely known?

              There seem to be too many "unknown" statements.
              Last edited by sharon sanders; January 17, 2008, 09:05 AM. Reason: typo

              Comment

              Working...
              X