Skip directly to site content Skip directly to page options Skip directly to A-Z link Skip directly to A-Z link Skip directly to A-Z link
Issue Cover for Volume 19, Number 8—August 2013

Volume 19, Number 8—August 2013

[PDF - 10.46 MB - 179 pages]

Perspective

The New Global Health [PDF - 271 KB - 6 pages]
K. M. De Cock et al.

Global health reflects the realities of globalization, including worldwide dissemination of infectious and noninfectious public health risks. Global health architecture is complex and better coordination is needed between multiple organizations. Three overlapping themes determine global health action and prioritization: development, security, and public health. These themes play out against a background of demographic change, socioeconomic development, and urbanization. Infectious diseases remain critical factors, but are no longer the major cause of global illness and death. Traditional indicators of public health, such as maternal and infant mortality rates no longer describe the health status of whole societies; this change highlights the need for investment in vital registration and disease-specific reporting. Noncommunicable diseases, injuries, and mental health will require greater attention from the world in the future. The new global health requires broader engagement by health organizations and all countries for the objectives of health equity, access, and coverage as priorities beyond the Millennium Development Goals are set.

EID De Cock KM, Simone PM, Davison V, Slutsker L. The New Global Health. Emerg Infect Dis. 2013;19(8):1192-1197. https://doi.org/10.3201/eid1908.130121
AMA De Cock KM, Simone PM, Davison V, et al. The New Global Health. Emerging Infectious Diseases. 2013;19(8):1192-1197. doi:10.3201/eid1908.130121.
APA De Cock, K. M., Simone, P. M., Davison, V., & Slutsker, L. (2013). The New Global Health. Emerging Infectious Diseases, 19(8), 1192-1197. https://doi.org/10.3201/eid1908.130121.

Norovirus Disease in the United States [PDF - 665 KB - 8 pages]
A. J. Hall et al.

Although recognized as the leading cause of epidemic acute gastroenteritis across all age groups, norovirus has remained poorly characterized with respect to its endemic disease incidence. Use of different methods, including attributable proportion extrapolation, population-based surveillance, and indirect modeling, in several recent studies has considerably improved norovirus disease incidence estimates for the United States. Norovirus causes an average of 570–800 deaths, 56,000–71,000 hospitalizations, 400,000 emergency department visits, 1.7–1.9 million outpatient visits, and 19–21 million total illnesses per year. Persons >65 years of age are at greatest risk for norovirus-associated death, and children <5 years of age have the highest rates of norovirus-associated medical care visits. Endemic norovirus disease occurs year round but exhibits a pronounced winter peak and increases by ≤50% during years in which pandemic strains emerge. These findings support continued development and targeting of appropriate interventions, including vaccines, for norovirus disease.

EID Hall AJ, Lopman BA, Payne DC, Patel MM, Gastañaduy PA, Vinjé J, et al. Norovirus Disease in the United States. Emerg Infect Dis. 2013;19(8):1198-1205. https://doi.org/10.3201/eid1908.130465
AMA Hall AJ, Lopman BA, Payne DC, et al. Norovirus Disease in the United States. Emerging Infectious Diseases. 2013;19(8):1198-1205. doi:10.3201/eid1908.130465.
APA Hall, A. J., Lopman, B. A., Payne, D. C., Patel, M. M., Gastañaduy, P. A., Vinjé, J....Parashar, U. D. (2013). Norovirus Disease in the United States. Emerging Infectious Diseases, 19(8), 1198-1205. https://doi.org/10.3201/eid1908.130465.
Research

Medscape CME Activity
Extended-Spectrum β-Lactamase–producing Enterobacteriaceae among Travelers from the Netherlands [PDF - 2.23 MB - 8 pages]
S. Paltansing et al.

A prospective cohort study was performed among travelers from the Netherlands to investigate the acquisition of carbapenemase-producing Enterobacteriaceae (CP-E) and extended-spectrum β-lactamase–producing Enterobacteriaceae (ESBL-E) and associated risk factors. Questionnaires were administered and rectal swabs were collected and tested before and after return. Of 370 travelers, 32 (8.6%) were colonized with ESBL-E before travel; 113 (30.5%) acquired an ESBL-E during travel, and 26 were still colonized 6 months after return. No CP-E were found. Independent risk factors for ESBL-E acquisition were travel to South and East Asia. Multilocus sequence typing showed extensive genetic diversity among Escherichia coli. Predominant ESBLs were CTX-M enzymes. The acquisition rate, 30.5%, of ESBL-E in travelers from the Netherlands to all destinations studied was high. Active surveillance for ESBL-E and CP-E and contact isolation precautions may be recommended at admission to medical facilities for patients who traveled to Asia during the previous 6 months.

EID Paltansing S, Vlot JA, Kraakman M, Mesman R, Bruijning ML, Bernards AT, et al. Extended-Spectrum β-Lactamase–producing Enterobacteriaceae among Travelers from the Netherlands. Emerg Infect Dis. 2013;19(8):1206-1213. https://doi.org/10.3201/eid1908.130257
AMA Paltansing S, Vlot JA, Kraakman M, et al. Extended-Spectrum β-Lactamase–producing Enterobacteriaceae among Travelers from the Netherlands. Emerging Infectious Diseases. 2013;19(8):1206-1213. doi:10.3201/eid1908.130257.
APA Paltansing, S., Vlot, J. A., Kraakman, M., Mesman, R., Bruijning, M. L., Bernards, A. T....Veldkamp, K. (2013). Extended-Spectrum β-Lactamase–producing Enterobacteriaceae among Travelers from the Netherlands. Emerging Infectious Diseases, 19(8), 1206-1213. https://doi.org/10.3201/eid1908.130257.

Emergency Department Visit Data for Rapid Detection and Monitoring of Norovirus Activity, United States [PDF - 1.36 MB - 8 pages]
B. Rha et al.

Noroviruses are the leading cause of gastroenteritis in the United States, but timely measures of disease are lacking. BioSense, a national-level electronic surveillance system, assigns data on chief complaints (patient symptoms) collected during emergency department (ED) visits to 78 subsyndromes in near real-time. In a series of linear regression models, BioSense visits mapped by chief complaints of diarrhea and nausea/vomiting subsyndromes as a monthly proportion of all visits correlated strongly with reported norovirus outbreaks from 6 states during 2007–2010. Higher correlations were seen for diarrhea (R = 0.828–0.926) than for nausea/vomiting (R = 0.729–0.866) across multiple age groups. Diarrhea ED visit proportions exhibited winter seasonality attributable to norovirus; rotavirus contributed substantially for children <5 years of age. Diarrhea ED visit data estimated the onset, peak, and end of norovirus season within 4 weeks of observed dates and could be reliable, timely indicators of norovirus activity.

EID Rha B, Burrer S, Park S, Trivedi T, Parashar UD, Lopman BA. Emergency Department Visit Data for Rapid Detection and Monitoring of Norovirus Activity, United States. Emerg Infect Dis. 2013;19(8):1214-1221. https://doi.org/10.3201/eid1908.130483
AMA Rha B, Burrer S, Park S, et al. Emergency Department Visit Data for Rapid Detection and Monitoring of Norovirus Activity, United States. Emerging Infectious Diseases. 2013;19(8):1214-1221. doi:10.3201/eid1908.130483.
APA Rha, B., Burrer, S., Park, S., Trivedi, T., Parashar, U. D., & Lopman, B. A. (2013). Emergency Department Visit Data for Rapid Detection and Monitoring of Norovirus Activity, United States. Emerging Infectious Diseases, 19(8), 1214-1221. https://doi.org/10.3201/eid1908.130483.

Aichi Virus in Sewage and Surface Water, the Netherlands [PDF - 1.17 MB - 9 pages]
W. J. Lodder et al.

Detection of Aichi virus in humans was initially reported in Japan in 1989. To establish a timeline for the prevalence of Aichi virus infection among humans in the Netherlands, we conducted molecular analysis of archival water samples from 1987–2000 and 2009–2012. Aichi virus RNA was detected in 100% (8/8) of sewage samples and 100% (7/7) of surface water samples collected during 1987–2000 and 100% (8/8) of sewage samples and 71% (5/7) of surface water samples collected during 2009–2012. Several genotype A and B Aichi virus lineages were observed over the 25-year period studied, but the time course of viral genetic diversity showed recent expansion of the genotype B population over genotype A. Our results show that Aichi virus has been circulating among the human population in the Netherlands since before its initial detection in humans was reported and that genotype B now predominates in this country.

EID Lodder WJ, Rutjes SA, Takumi K, Husman A. Aichi Virus in Sewage and Surface Water, the Netherlands. Emerg Infect Dis. 2013;19(8):1222-1230. https://doi.org/10.3201/eid1908.130312
AMA Lodder WJ, Rutjes SA, Takumi K, et al. Aichi Virus in Sewage and Surface Water, the Netherlands. Emerging Infectious Diseases. 2013;19(8):1222-1230. doi:10.3201/eid1908.130312.
APA Lodder, W. J., Rutjes, S. A., Takumi, K., & Husman, A. (2013). Aichi Virus in Sewage and Surface Water, the Netherlands. Emerging Infectious Diseases, 19(8), 1222-1230. https://doi.org/10.3201/eid1908.130312.

Medscape CME Activity
Effects and Clinical Significance of GII.4 Sydney Norovirus, United States, 2012–2013 [PDF - 661 KB - 8 pages]
E. Leshem et al.

During 2012, global detection of a new norovirus (NoV) strain, GII.4 Sydney, raised concerns about its potential effect in the United States. We analyzed data from NoV outbreaks in 5 states and emergency department visits for gastrointestinal illness in 1 state during the 2012–13 season and compared the data with those of previous seasons. During August 2012–April 2013, a total of 637 NoV outbreaks were reported compared with 536 and 432 in 2011–2012 and 2010–2011 during the same period. The proportion of outbreaks attributed to GII.4 Sydney increased from 8% in September 2012 to 82% in March 2013. The increase in emergency department visits for gastrointestinal illness during the 2012–13 season was similar to that of previous seasons. GII.4 Sydney has become the predominant US NoV outbreak strain during the 2012–13 season, but its emergence did not cause outbreak activity to substantially increase from that of previous seasons.

EID Leshem E, Wikswo M, Barclay L, Brandt E, Storm W, Salehi E, et al. Effects and Clinical Significance of GII.4 Sydney Norovirus, United States, 2012–2013. Emerg Infect Dis. 2013;19(8):1231-1238. https://doi.org/10.3201/eid1908.130458
AMA Leshem E, Wikswo M, Barclay L, et al. Effects and Clinical Significance of GII.4 Sydney Norovirus, United States, 2012–2013. Emerging Infectious Diseases. 2013;19(8):1231-1238. doi:10.3201/eid1908.130458.
APA Leshem, E., Wikswo, M., Barclay, L., Brandt, E., Storm, W., Salehi, E....Hall, A. J. (2013). Effects and Clinical Significance of GII.4 Sydney Norovirus, United States, 2012–2013. Emerging Infectious Diseases, 19(8), 1231-1238. https://doi.org/10.3201/eid1908.130458.

Outbreak-associated Salmonella enterica Serotypes and Food Commodities, United States, 1998–2008 [PDF - 365 KB - 6 pages]
B. R. Jackson et al.

Salmonella enterica infections are transmitted not only by animal-derived foods but also by vegetables, fruits, and other plant products. To clarify links between Salmonella serotypes and specific foods, we examined the diversity and predominance of food commodities implicated in outbreaks of salmonellosis during 1998–2008. More than 80% of outbreaks caused by serotypes Enteritidis, Heidelberg, and Hadar were attributed to eggs or poultry, whereas >50% of outbreaks caused by serotypes Javiana, Litchfield, Mbandaka, Muenchen, Poona, and Senftenberg were attributed to plant commodities. Serotypes Typhimurium and Newport were associated with a wide variety of food commodities. Knowledge about these associations can help guide outbreak investigations and control measures.

EID Jackson BR, Griffin PM, Cole D, Walsh KA, Chai SJ. Outbreak-associated Salmonella enterica Serotypes and Food Commodities, United States, 1998–2008. Emerg Infect Dis. 2013;19(8):1239-1244. https://doi.org/10.3201/eid1908.121511
AMA Jackson BR, Griffin PM, Cole D, et al. Outbreak-associated Salmonella enterica Serotypes and Food Commodities, United States, 1998–2008. Emerging Infectious Diseases. 2013;19(8):1239-1244. doi:10.3201/eid1908.121511.
APA Jackson, B. R., Griffin, P. M., Cole, D., Walsh, K. A., & Chai, S. J. (2013). Outbreak-associated Salmonella enterica Serotypes and Food Commodities, United States, 1998–2008. Emerging Infectious Diseases, 19(8), 1239-1244. https://doi.org/10.3201/eid1908.121511.

Comparison of 2 Assays for Diagnosing Rotavirus and Evaluating Vaccine Effectiveness in Children with Gastroenteritis [PDF - 543 KB - 8 pages]
J. E. Tate et al.

We compared rotavirus detection rates in children with acute gastroenteritis (AGE) and in healthy controls using enzyme immunoassays (EIAs) and semiquantitative real-time reverse transcription PCR (qRT-PCR). We calculated rotavirus vaccine effectiveness using different laboratory-based case definitions to determine which best identified the proportion of disease that was vaccine preventable. Of 648 AGE patients, 158 (24%) were EIA positive, and 157 were also qRT-PCR positive. An additional 65 (10%) were qRT-PCR positive but EIA negative. Of 500 healthy controls, 1 was EIA positive and 24 (5%) were qRT-PCR positive. Rotavirus vaccine was highly effective (84% [95% CI 71%–91%]) in EIA-positive children but offered no significant protection (14% [95% CI −105% to 64%]) in EIA-negative children for whom virus was detected by qRT-PCR alone. Children with rotavirus detected by qRT-PCR but not by EIA were not protected by vaccination, suggesting that rotavirus detected by qRT-PCR alone might not be causally associated with AGE in all patients.

EID Tate JE, Mijatovic-Rustempasic S, Tam K, Lyde FC, Payne DC, Szilagyi P, et al. Comparison of 2 Assays for Diagnosing Rotavirus and Evaluating Vaccine Effectiveness in Children with Gastroenteritis. Emerg Infect Dis. 2013;19(8):1245-1252. https://doi.org/10.3201/eid1908.130461
AMA Tate JE, Mijatovic-Rustempasic S, Tam K, et al. Comparison of 2 Assays for Diagnosing Rotavirus and Evaluating Vaccine Effectiveness in Children with Gastroenteritis. Emerging Infectious Diseases. 2013;19(8):1245-1252. doi:10.3201/eid1908.130461.
APA Tate, J. E., Mijatovic-Rustempasic, S., Tam, K., Lyde, F. C., Payne, D. C., Szilagyi, P....Parashar, U. D. (2013). Comparison of 2 Assays for Diagnosing Rotavirus and Evaluating Vaccine Effectiveness in Children with Gastroenteritis. Emerging Infectious Diseases, 19(8), 1245-1252. https://doi.org/10.3201/eid1908.130461.

Extended-Spectrum β-Lactamase– and AmpC-Producing Enterobacteria in Healthy Broiler Chickens, Germany [PDF - 775 KB - 7 pages]
F. Reich et al.

During 2010, we evaluated the presence of extended-spectrum β-lactamase– and AmpC-producing enterobacteria in broiler chickens at slaughter. Samples (70 carcasses and 51 ceca) from 4 flocks were analyzed by direct plating and after enrichment. Extended-spectrum β-lactamase producers were found in 88.6% and 72.5% of carcasses and ceca, respectively; AmpC producers were found in 52.9% and 56.9% of carcasses and ceca, respectively. Most isolates were identified as Escherichia coli; Enterobacter cloacae (cecum) and Proteus mirabilis (carcass) were found in 2 samples each. Molecular characterization revealed the domination of CTX-M genes; plasmidic AmpC was CIT-like. Phylogenetic grouping of E. coli showed types A (31.5%), B1 (20.2%), B2 (13.5%), and D (34.8%). These findings provide evidence that healthy broilers in Germany are a source for the dissemination of transmissible resistance mechanisms in enterobacteria brought from the rearing environment into the food chain during slaughtering.

EID Reich F, Atanassova V, Klein G. Extended-Spectrum β-Lactamase– and AmpC-Producing Enterobacteria in Healthy Broiler Chickens, Germany. Emerg Infect Dis. 2013;19(8):1253-1259. https://doi.org/10.3201/eid1908.120879
AMA Reich F, Atanassova V, Klein G. Extended-Spectrum β-Lactamase– and AmpC-Producing Enterobacteria in Healthy Broiler Chickens, Germany. Emerging Infectious Diseases. 2013;19(8):1253-1259. doi:10.3201/eid1908.120879.
APA Reich, F., Atanassova, V., & Klein, G. (2013). Extended-Spectrum β-Lactamase– and AmpC-Producing Enterobacteria in Healthy Broiler Chickens, Germany. Emerging Infectious Diseases, 19(8), 1253-1259. https://doi.org/10.3201/eid1908.120879.

Duration of Immunity to Norovirus Gastroenteritis [PDF - 482 KB - 8 pages]
K. Simmons et al.

The duration of immunity to norovirus (NoV) gastroenteritis has been believed to be from 6 months to 2 years. However, several observations are inconsistent with this short period. To gain better estimates of the duration of immunity to NoV, we developed a mathematical model of community NoV transmission. The model was parameterized from the literature and also fit to age-specific incidence data from England and Wales by using maximum likelihood. We developed several scenarios to determine the effect of unknowns regarding transmission and immunity on estimates of the duration of immunity. In the various models, duration of immunity to NoV gastroenteritis was estimated at 4.1 (95% CI 3.2–5.1) to 8.7 (95% CI 6.8–11.3) years. Moreover, we calculated that children (<5 years) are much more infectious than older children and adults. If a vaccine can achieve protection for duration of natural immunity indicated by our results, its potential health and economic benefits could be substantial.

EID Simmons K, Gambhir M, Leon J, Lopman B. Duration of Immunity to Norovirus Gastroenteritis. Emerg Infect Dis. 2013;19(8):1260-1267. https://doi.org/10.3201/eid1908.130472
AMA Simmons K, Gambhir M, Leon J, et al. Duration of Immunity to Norovirus Gastroenteritis. Emerging Infectious Diseases. 2013;19(8):1260-1267. doi:10.3201/eid1908.130472.
APA Simmons, K., Gambhir, M., Leon, J., & Lopman, B. (2013). Duration of Immunity to Norovirus Gastroenteritis. Emerging Infectious Diseases, 19(8), 1260-1267. https://doi.org/10.3201/eid1908.130472.

Accuracy of Diagnostic Methods and Surveillance Sensitivity for Human Enterovirus, South Korea, 1999–2011 [PDF - 546 KB - 8 pages]
J. Hyeon et al.

The epidemiology of enteroviral infection in South Korea during 1999–2011 chronicles nationwide outbreaks and changing detection and subtyping methods used over the 13-year period. Of 14,657 patients whose samples were tested, 4,762 (32.5%) samples were positive for human enterovirus (human EV); as diagnostic methods improved, the rate of positive results increased. A seasonal trend of outbreaks was documented. Genotypes enterovirus 71, echovirus 30, coxsackievirus B5, enterovirus 6, and coxsackievirus B2 were the most common genotypes identified. Accurate test results correlated clinical syndromes to enterovirus genotypes: aseptic meningitis to echovirus 30, enterovirus 6, and coxsackievirus B5; hand, foot and mouth disease to coxsackievirus A16; and hand, foot and mouth disease with neurologic complications to enterovirus 71. There are currently no treatments specific to human EV infections; surveillance of enterovirus infections such as this study provides may assist with evaluating the need to research and develop treatments for infections caused by virulent human EV genotypes.

EID Hyeon J, Hwang S, Kim H, Song J, Ahn J, Kang B, et al. Accuracy of Diagnostic Methods and Surveillance Sensitivity for Human Enterovirus, South Korea, 1999–2011. Emerg Infect Dis. 2013;19(8):1268-1275. https://doi.org/10.3201/eid1908.130496
AMA Hyeon J, Hwang S, Kim H, et al. Accuracy of Diagnostic Methods and Surveillance Sensitivity for Human Enterovirus, South Korea, 1999–2011. Emerging Infectious Diseases. 2013;19(8):1268-1275. doi:10.3201/eid1908.130496.
APA Hyeon, J., Hwang, S., Kim, H., Song, J., Ahn, J., Kang, B....Cheon, D. (2013). Accuracy of Diagnostic Methods and Surveillance Sensitivity for Human Enterovirus, South Korea, 1999–2011. Emerging Infectious Diseases, 19(8), 1268-1275. https://doi.org/10.3201/eid1908.130496.
Policy Review

Impact of 2003 State Regulation on Raw Oyster–associated Vibrio vulnificus Illnesses and Deaths, California, USA [PDF - 1.00 MB - 5 pages]
D. J. Vugia et al.

US vibriosis rates have increased since 1996, and many Vibrio vulnificus infections are fatal. In April 2003, California implemented a regulation restricting the sale of raw oysters harvested from the Gulf of Mexico during April 1–October 31, unless they were processed to reduce V. vulnificus to nondetectable levels. We analyzed California cases of V. vulnificus infection before and after the regulation’s implementation and compared case data with data from other states. The annual number of reported V. vulnificus infections and deaths in California with patient’s sole exposure to raw oysters dropped from 0 to 6 cases and 0 to 5 deaths per year during 1991–2002, before implementation, to 0 during 2003–2010, after implementation (p = 0.0005 for both). In other states, median annual numbers of similar cases and deaths increased slightly after 2002. The data strongly suggest that the 2003 regulation led to a significant reduction in reported raw oyster–associated V. vulnificus illnesses and deaths.

EID Vugia DJ, Tabnak F, Newton AE, Hernandez M, Griffin PM. Impact of 2003 State Regulation on Raw Oyster–associated Vibrio vulnificus Illnesses and Deaths, California, USA. Emerg Infect Dis. 2013;19(8):1276-1280. https://doi.org/10.3201/eid1908.121861
AMA Vugia DJ, Tabnak F, Newton AE, et al. Impact of 2003 State Regulation on Raw Oyster–associated Vibrio vulnificus Illnesses and Deaths, California, USA. Emerging Infectious Diseases. 2013;19(8):1276-1280. doi:10.3201/eid1908.121861.
APA Vugia, D. J., Tabnak, F., Newton, A. E., Hernandez, M., & Griffin, P. M. (2013). Impact of 2003 State Regulation on Raw Oyster–associated Vibrio vulnificus Illnesses and Deaths, California, USA. Emerging Infectious Diseases, 19(8), 1276-1280. https://doi.org/10.3201/eid1908.121861.
Dispatches

Macrolide Resistance of Mycoplasma pneumoniae, South Korea, 2000–2011 [PDF - 509 KB - 4 pages]
K. Hong et al.

In Korea, Mycoplasma pneumoniae was detected in 255/2,089 respiratory specimens collected during 2000–2011; 80 isolates carried 23S rRNA gene mutations, and 69/123 culture-positive samples with the mutation were resistant to 5 macrolides. During 2000–2011, prevalence of the mutation increased substantially. These findings have critical implications for the treatment of children with mycoplasma pneumonia.

EID Hong K, Choi E, Lee H, Lee S, Cho E, Choi J, et al. Macrolide Resistance of Mycoplasma pneumoniae, South Korea, 2000–2011. Emerg Infect Dis. 2013;19(8):1281-1284. https://doi.org/10.3201/eid1908.121455
AMA Hong K, Choi E, Lee H, et al. Macrolide Resistance of Mycoplasma pneumoniae, South Korea, 2000–2011. Emerging Infectious Diseases. 2013;19(8):1281-1284. doi:10.3201/eid1908.121455.
APA Hong, K., Choi, E., Lee, H., Lee, S., Cho, E., Choi, J....Lee, J. (2013). Macrolide Resistance of Mycoplasma pneumoniae, South Korea, 2000–2011. Emerging Infectious Diseases, 19(8), 1281-1284. https://doi.org/10.3201/eid1908.121455.

Recombinant Coxsackievirus A2 and Deaths of Children, Hong Kong, 2012 [PDF - 383 KB - 4 pages]
C. Yip et al.

A natural recombinant of coxsackievirus A2 was found in 4 children with respiratory symptoms in Hong Kong, China, during the summer of 2012. Two of these children died. Vigilant monitoring of this emerging recombinant enterovirus is needed to prevent its transmission to other regions.

EID Yip C, Lau S, Woo P, Wong S, Tsang T, Lo J, et al. Recombinant Coxsackievirus A2 and Deaths of Children, Hong Kong, 2012. Emerg Infect Dis. 2013;19(8):1285-1288. https://doi.org/10.3201/eid1908.121498
AMA Yip C, Lau S, Woo P, et al. Recombinant Coxsackievirus A2 and Deaths of Children, Hong Kong, 2012. Emerging Infectious Diseases. 2013;19(8):1285-1288. doi:10.3201/eid1908.121498.
APA Yip, C., Lau, S., Woo, P., Wong, S., Tsang, T., Lo, J....Yuen, K. (2013). Recombinant Coxsackievirus A2 and Deaths of Children, Hong Kong, 2012. Emerging Infectious Diseases, 19(8), 1285-1288. https://doi.org/10.3201/eid1908.121498.

Monitoring Avian Influenza A(H7N9) Virus through National Influenza-like Illness Surveillance, China [PDF - 445 KB - 4 pages]
C. Xu et al.

In China during March 4–April 28, 2013, avian influenza A(H7N9) virus testing was performed on 20,739 specimens from patients with influenza-like illness in 10 provinces with confirmed human cases: 6 (0.03%) were positive, and increased numbers of unsubtypeable influenza-positive specimens were not seen. Careful monitoring and rapid characterization of influenza A(H7N9) and other influenza viruses remain critical.

EID Xu C, Havers F, Wang L, Chen T, Shi J, Wang D, et al. Monitoring Avian Influenza A(H7N9) Virus through National Influenza-like Illness Surveillance, China. Emerg Infect Dis. 2013;19(8):1289-1292. https://doi.org/10.3201/eid1908.130662
AMA Xu C, Havers F, Wang L, et al. Monitoring Avian Influenza A(H7N9) Virus through National Influenza-like Illness Surveillance, China. Emerging Infectious Diseases. 2013;19(8):1289-1292. doi:10.3201/eid1908.130662.
APA Xu, C., Havers, F., Wang, L., Chen, T., Shi, J., Wang, D....Shu, Y. (2013). Monitoring Avian Influenza A(H7N9) Virus through National Influenza-like Illness Surveillance, China. Emerging Infectious Diseases, 19(8), 1289-1292. https://doi.org/10.3201/eid1908.130662.

Norovirus Surveillance among Callers to Foodborne Illness Complaint Hotline, Minnesota, USA, 2011–2013 [PDF - 382 KB - 4 pages]
A. A. Saupe et al.

Norovirus is the leading cause of foodborne disease in the United States. During October 2011–January 2013, we conducted surveillance for norovirus infection in Minnesota among callers to a complaint-based foodborne illness hotline who reported diarrhea or vomiting. Of 241 complainants tested, 127 (52.7%) were positive for norovirus.

EID Saupe AA, Kaehler D, Cebelinski EA, Nefzger B, Hall AJ, Smith KE. Norovirus Surveillance among Callers to Foodborne Illness Complaint Hotline, Minnesota, USA, 2011–2013. Emerg Infect Dis. 2013;19(8):1293-1296. https://doi.org/10.3201/eid1908.130462
AMA Saupe AA, Kaehler D, Cebelinski EA, et al. Norovirus Surveillance among Callers to Foodborne Illness Complaint Hotline, Minnesota, USA, 2011–2013. Emerging Infectious Diseases. 2013;19(8):1293-1296. doi:10.3201/eid1908.130462.
APA Saupe, A. A., Kaehler, D., Cebelinski, E. A., Nefzger, B., Hall, A. J., & Smith, K. E. (2013). Norovirus Surveillance among Callers to Foodborne Illness Complaint Hotline, Minnesota, USA, 2011–2013. Emerging Infectious Diseases, 19(8), 1293-1296. https://doi.org/10.3201/eid1908.130462.

Travel-associated Diseases, Indian Ocean Islands, 1997–2010 [PDF - 439 KB - 5 pages]
H. Savini et al.

Data collected by the GeoSentinel Surveillance Network for 1,415 ill travelers returning from Indian Ocean islands during 1997–2010 were analyzed. Malaria (from Comoros and Madagascar), acute nonparasitic diarrhea, and parasitoses were the most frequently diagnosed infectious diseases. An increase in arboviral diseases reflected the 2005 outbreak of chikungunya fever.

EID Savini H, Gautret P, Gaudart J, Field V, Castelli F, López-Vélez R, et al. Travel-associated Diseases, Indian Ocean Islands, 1997–2010. Emerg Infect Dis. 2013;19(8):1297-1301. https://doi.org/10.3201/eid1908.121739
AMA Savini H, Gautret P, Gaudart J, et al. Travel-associated Diseases, Indian Ocean Islands, 1997–2010. Emerging Infectious Diseases. 2013;19(8):1297-1301. doi:10.3201/eid1908.121739.
APA Savini, H., Gautret, P., Gaudart, J., Field, V., Castelli, F., López-Vélez, R....Simon, F. (2013). Travel-associated Diseases, Indian Ocean Islands, 1997–2010. Emerging Infectious Diseases, 19(8), 1297-1301. https://doi.org/10.3201/eid1908.121739.

Whole Genome Sequencing of an Unusual Serotype of Shiga Toxin–producing Escherichia coli [PDF - 368 KB - 3 pages]
T. Dallman et al.

Shiga toxin–producing Escherichia coli serotype O117:K1:H7 is a cause of persistent diarrhea in travelers to tropical locations. Whole genome sequencing identified genetic mechanisms involved in the pathoadaptive phenotype. Sequencing also identified toxin and putative adherence genes flanked by sequences indicating horizontal gene transfer from Shigella dysenteriae and Salmonella spp., respectively.

EID Dallman T, Cross L, Bishop C, Perry N, Olesen B, Grant KA, et al. Whole Genome Sequencing of an Unusual Serotype of Shiga Toxin–producing Escherichia coli. Emerg Infect Dis. 2013;19(8):1302-1304. https://doi.org/10.3201/eid1908.130016
AMA Dallman T, Cross L, Bishop C, et al. Whole Genome Sequencing of an Unusual Serotype of Shiga Toxin–producing Escherichia coli. Emerging Infectious Diseases. 2013;19(8):1302-1304. doi:10.3201/eid1908.130016.
APA Dallman, T., Cross, L., Bishop, C., Perry, N., Olesen, B., Grant, K. A....Jenkins, C. (2013). Whole Genome Sequencing of an Unusual Serotype of Shiga Toxin–producing Escherichia coli. Emerging Infectious Diseases, 19(8), 1302-1304. https://doi.org/10.3201/eid1908.130016.

Acute Gastroenteritis Surveillance through the National Outbreak Reporting System, United States [PDF - 616 KB - 5 pages]
A. J. Hall et al.

Implemented in 2009, the National Outbreak Reporting System provides surveillance for acute gastroenteritis outbreaks in the United States resulting from any transmission mode. Data from the first 2 years of surveillance highlight the predominant role of norovirus. The pathogen-specific transmission pathways and exposure settings identified can help inform prevention efforts.

EID Hall AJ, Wikswo ME, Manikonda K, Roberts VA, Yoder JS, Gould L. Acute Gastroenteritis Surveillance through the National Outbreak Reporting System, United States. Emerg Infect Dis. 2013;19(8):1305-1309. https://doi.org/10.3201/eid1908.130482
AMA Hall AJ, Wikswo ME, Manikonda K, et al. Acute Gastroenteritis Surveillance through the National Outbreak Reporting System, United States. Emerging Infectious Diseases. 2013;19(8):1305-1309. doi:10.3201/eid1908.130482.
APA Hall, A. J., Wikswo, M. E., Manikonda, K., Roberts, V. A., Yoder, J. S., & Gould, L. (2013). Acute Gastroenteritis Surveillance through the National Outbreak Reporting System, United States. Emerging Infectious Diseases, 19(8), 1305-1309. https://doi.org/10.3201/eid1908.130482.

Duck Liver–associated Outbreak of Campylobacteriosis among Humans, United Kingdom, 2011 [PDF - 378 KB - 4 pages]
M. Abid et al.

Campylobacter­ spp.–related gastroenteritis in diners at a catering college restaurant was associated with consumption of duck liver pâté. Population genetic analysis indicated that isolates from duck samples were typical of isolates from farmed poultry. Campylobacter spp. contamination of duck liver may present a hazard similar to the increasingly recognized contamination of chicken liver.

EID Abid M, Wimalarathna H, Mills J, Saldana L, Pang W, Richardson JF, et al. Duck Liver–associated Outbreak of Campylobacteriosis among Humans, United Kingdom, 2011. Emerg Infect Dis. 2013;19(8):1310-1313. https://doi.org/10.3201/eid1908.121535
AMA Abid M, Wimalarathna H, Mills J, et al. Duck Liver–associated Outbreak of Campylobacteriosis among Humans, United Kingdom, 2011. Emerging Infectious Diseases. 2013;19(8):1310-1313. doi:10.3201/eid1908.121535.
APA Abid, M., Wimalarathna, H., Mills, J., Saldana, L., Pang, W., Richardson, J. F....McCarthy, N. D. (2013). Duck Liver–associated Outbreak of Campylobacteriosis among Humans, United Kingdom, 2011. Emerging Infectious Diseases, 19(8), 1310-1313. https://doi.org/10.3201/eid1908.121535.

Diarrhetic Shellfish Poisoning, Washington, USA, 2011 [PDF - 322 KB - 3 pages]
J. K. Lloyd et al.

Diarrhetic shellfish poisoning is a gastrointestinal illness caused by consumption of bivalves contaminated with dinophysistoxins. We report an illness cluster in the United States in which toxins were confirmed in shellfish from a commercial harvest area, leading to product recall. Ongoing surveillance is needed to prevent similar illness outbreaks.

EID Lloyd JK, Duchin JS, Borchert J, Quintana H, Robertson A. Diarrhetic Shellfish Poisoning, Washington, USA, 2011. Emerg Infect Dis. 2013;19(8):1314-1316. https://doi.org/10.3201/eid1908.121824
AMA Lloyd JK, Duchin JS, Borchert J, et al. Diarrhetic Shellfish Poisoning, Washington, USA, 2011. Emerging Infectious Diseases. 2013;19(8):1314-1316. doi:10.3201/eid1908.121824.
APA Lloyd, J. K., Duchin, J. S., Borchert, J., Quintana, H., & Robertson, A. (2013). Diarrhetic Shellfish Poisoning, Washington, USA, 2011. Emerging Infectious Diseases, 19(8), 1314-1316. https://doi.org/10.3201/eid1908.121824.

Genotype GI.6 Norovirus, United States, 2010–2012 [PDF - 420 KB - 4 pages]
E. Leshem et al.

We report an increase in the proportion of genotype GI.6 norovirus outbreaks in the United States from 1.4% in 2010 to 7.7% in 2012 (p<0.001). Compared with non-GI.6 outbreaks, GI.6 outbreaks were characterized by summer seasonality, foodborne transmission, and non–health care settings.

EID Leshem E, Barclay L, Wikswo M, Vega E, Gregoricus N, Parashar UD, et al. Genotype GI.6 Norovirus, United States, 2010–2012. Emerg Infect Dis. 2013;19(8):1317-1320. https://doi.org/10.3201/eid1908.130445
AMA Leshem E, Barclay L, Wikswo M, et al. Genotype GI.6 Norovirus, United States, 2010–2012. Emerging Infectious Diseases. 2013;19(8):1317-1320. doi:10.3201/eid1908.130445.
APA Leshem, E., Barclay, L., Wikswo, M., Vega, E., Gregoricus, N., Parashar, U. D....Hall, A. J. (2013). Genotype GI.6 Norovirus, United States, 2010–2012. Emerging Infectious Diseases, 19(8), 1317-1320. https://doi.org/10.3201/eid1908.130445.

Detection of Novel Rotavirus Strain by Vaccine Postlicensure Surveillance [PDF - 444 KB - 3 pages]
G. A. Weinberg et al.

Surveillance for rotavirus-associated diarrhea after implementation of rotavirus vaccination can assess vaccine effectiveness and identify disease-associated genotypes. During active vaccine postlicensure surveillance in the United States, we found a novel rotavirus genotype, G14P[24], in a stool sample from a child who had diarrhea. Unusual rotavirus strains may become more prevalent after vaccine implementation.

EID Weinberg GA, Teel EN, Mijatovic-Rustempasic S, Payne DC, Roy S, Foytich K, et al. Detection of Novel Rotavirus Strain by Vaccine Postlicensure Surveillance. Emerg Infect Dis. 2013;19(8):1321-1323. https://doi.org/10.3201/eid1908.130470
AMA Weinberg GA, Teel EN, Mijatovic-Rustempasic S, et al. Detection of Novel Rotavirus Strain by Vaccine Postlicensure Surveillance. Emerging Infectious Diseases. 2013;19(8):1321-1323. doi:10.3201/eid1908.130470.
APA Weinberg, G. A., Teel, E. N., Mijatovic-Rustempasic, S., Payne, D. C., Roy, S., Foytich, K....Bowen, M. D. (2013). Detection of Novel Rotavirus Strain by Vaccine Postlicensure Surveillance. Emerging Infectious Diseases, 19(8), 1321-1323. https://doi.org/10.3201/eid1908.130470.

Novel G10P[14] Rotavirus Strain, Northern Territory, Australia [PDF - 573 KB - 4 pages]
D. Cowley et al.

We identified a genotype G10P[14] rotavirus strain in 5 children and 1 adult with acute gastroenteritis from the Northern Territory, Australia. Full genome sequence analysis identified an artiodactyl-like (bovine, ovine, and camelid) G10-P[14]-I2-R2-C2-M2-A11-N2-T6-E2-H3 genome constellation. This finding suggests artiodactyl-to-human transmission and strengthens the need to continue rotavirus strain surveillance.

EID Cowley D, Donato CM, Roczo-Farkas S, Kirkwood CD. Novel G10P[14] Rotavirus Strain, Northern Territory, Australia. Emerg Infect Dis. 2013;19(8):1324-1327. https://doi.org/10.3201/eid1908.121653
AMA Cowley D, Donato CM, Roczo-Farkas S, et al. Novel G10P[14] Rotavirus Strain, Northern Territory, Australia. Emerging Infectious Diseases. 2013;19(8):1324-1327. doi:10.3201/eid1908.121653.
APA Cowley, D., Donato, C. M., Roczo-Farkas, S., & Kirkwood, C. D. (2013). Novel G10P[14] Rotavirus Strain, Northern Territory, Australia. Emerging Infectious Diseases, 19(8), 1324-1327. https://doi.org/10.3201/eid1908.121653.

Primary and Secondary Human Bocavirus 1 Infections in a Family, Finland [PDF - 930 KB - 4 pages]
A. Jula et al.

Human bocavirus 1 (HBoV1) was detected in a young child hospitalized for pneumonia and subsequently in his twin brother and other family members. The mother’s nasopharyngeal samples intermittently showed HBoV1 DNA; the grandmother had HBoV1 reinfection. Findings in this family lead to consideration of HBoV virulence, latency, and reactivation.

EID Jula A, Waris M, Kantola K, Peltola V, Söderlund-Venermo M, Hedman K, et al. Primary and Secondary Human Bocavirus 1 Infections in a Family, Finland. Emerg Infect Dis. 2013;19(8):1328-1331. https://doi.org/10.3201/eid1908.130074
AMA Jula A, Waris M, Kantola K, et al. Primary and Secondary Human Bocavirus 1 Infections in a Family, Finland. Emerging Infectious Diseases. 2013;19(8):1328-1331. doi:10.3201/eid1908.130074.
APA Jula, A., Waris, M., Kantola, K., Peltola, V., Söderlund-Venermo, M., Hedman, K....Ruuskanen, O. (2013). Primary and Secondary Human Bocavirus 1 Infections in a Family, Finland. Emerging Infectious Diseases, 19(8), 1328-1331. https://doi.org/10.3201/eid1908.130074.
Commentaries

Rapid Advances in Understanding Viral Gastroenteritis from Domestic Surveillance [PDF - 222 KB - 1 page]
D. C. Payne and U. D. Parashar
EID Payne DC, Parashar UD. Rapid Advances in Understanding Viral Gastroenteritis from Domestic Surveillance. Emerg Infect Dis. 2013;19(8):1189. https://doi.org/10.3201/eid1908.130449
AMA Payne DC, Parashar UD. Rapid Advances in Understanding Viral Gastroenteritis from Domestic Surveillance. Emerging Infectious Diseases. 2013;19(8):1189. doi:10.3201/eid1908.130449.
APA Payne, D. C., & Parashar, U. D. (2013). Rapid Advances in Understanding Viral Gastroenteritis from Domestic Surveillance. Emerging Infectious Diseases, 19(8), 1189. https://doi.org/10.3201/eid1908.130449.

Beyond Discovering the Viral Agents of Acute Gastroenteritis [PDF - 394 KB - 2 pages]
R. Glass
EID Glass R. Beyond Discovering the Viral Agents of Acute Gastroenteritis. Emerg Infect Dis. 2013;19(8):1190-1191. https://doi.org/10.3201/eid1908.130773
AMA Glass R. Beyond Discovering the Viral Agents of Acute Gastroenteritis. Emerging Infectious Diseases. 2013;19(8):1190-1191. doi:10.3201/eid1908.130773.
APA Glass, R. (2013). Beyond Discovering the Viral Agents of Acute Gastroenteritis. Emerging Infectious Diseases, 19(8), 1190-1191. https://doi.org/10.3201/eid1908.130773.
Letters

Rotavirus G9P[4] in 3 Countries in Latin America, 2009–2010 [PDF - 259 KB - 2 pages]
O. Quaye et al.
EID Quaye O, McDonald S, Esona MD, Lyde FC, Mijatovic-Rustempasic S, Roy S, et al. Rotavirus G9P[4] in 3 Countries in Latin America, 2009–2010. Emerg Infect Dis. 2013;19(8):1332-1333. https://doi.org/10.3201/eid1908.130288
AMA Quaye O, McDonald S, Esona MD, et al. Rotavirus G9P[4] in 3 Countries in Latin America, 2009–2010. Emerging Infectious Diseases. 2013;19(8):1332-1333. doi:10.3201/eid1908.130288.
APA Quaye, O., McDonald, S., Esona, M. D., Lyde, F. C., Mijatovic-Rustempasic, S., Roy, S....Bowen, M. D. (2013). Rotavirus G9P[4] in 3 Countries in Latin America, 2009–2010. Emerging Infectious Diseases, 19(8), 1332-1333. https://doi.org/10.3201/eid1908.130288.

Recently Identified Novel Human Astroviruses in Children with Diarrhea, China [PDF - 315 KB - 3 pages]
Y. Wang et al.
EID Wang Y, Li Y, Jin Y, Li D, Li X, Duan Z. Recently Identified Novel Human Astroviruses in Children with Diarrhea, China. Emerg Infect Dis. 2013;19(8):1333-1335. https://doi.org/10.3201/eid1908.121863
AMA Wang Y, Li Y, Jin Y, et al. Recently Identified Novel Human Astroviruses in Children with Diarrhea, China. Emerging Infectious Diseases. 2013;19(8):1333-1335. doi:10.3201/eid1908.121863.
APA Wang, Y., Li, Y., Jin, Y., Li, D., Li, X., & Duan, Z. (2013). Recently Identified Novel Human Astroviruses in Children with Diarrhea, China. Emerging Infectious Diseases, 19(8), 1333-1335. https://doi.org/10.3201/eid1908.121863.

Call to Action for Dengue Vaccine Failure [PDF - 257 KB - 3 pages]
S. Mahalingam et al.
EID Mahalingam S, Herring BL, Halstead SB. Call to Action for Dengue Vaccine Failure. Emerg Infect Dis. 2013;19(8):1335-1337. https://doi.org/10.3201/eid1908.121864
AMA Mahalingam S, Herring BL, Halstead SB. Call to Action for Dengue Vaccine Failure. Emerging Infectious Diseases. 2013;19(8):1335-1337. doi:10.3201/eid1908.121864.
APA Mahalingam, S., Herring, B. L., & Halstead, S. B. (2013). Call to Action for Dengue Vaccine Failure. Emerging Infectious Diseases, 19(8), 1335-1337. https://doi.org/10.3201/eid1908.121864.

Novel Norovirus GII.4 Variant, Shanghai, China, 2012 [PDF - 337 KB - 2 pages]
Z. Shen et al.
EID Shen Z, Qian F, Li Y, Hu Y, Yuan Z, Zhang J. Novel Norovirus GII.4 Variant, Shanghai, China, 2012. Emerg Infect Dis. 2013;19(8):1337-1339. https://doi.org/10.3201/eid1908.130026
AMA Shen Z, Qian F, Li Y, et al. Novel Norovirus GII.4 Variant, Shanghai, China, 2012. Emerging Infectious Diseases. 2013;19(8):1337-1339. doi:10.3201/eid1908.130026.
APA Shen, Z., Qian, F., Li, Y., Hu, Y., Yuan, Z., & Zhang, J. (2013). Novel Norovirus GII.4 Variant, Shanghai, China, 2012. Emerging Infectious Diseases, 19(8), 1337-1339. https://doi.org/10.3201/eid1908.130026.

Human Deaths and Third-Generation Cephalosporin use in Poultry, Europe [PDF - 250 KB - 2 pages]
P. Collignon et al.
EID Collignon P, Aarestrup FM, Irwin R, McEwen S. Human Deaths and Third-Generation Cephalosporin use in Poultry, Europe. Emerg Infect Dis. 2013;19(8):1339-1340. https://doi.org/10.3201/eid1908.120681
AMA Collignon P, Aarestrup FM, Irwin R, et al. Human Deaths and Third-Generation Cephalosporin use in Poultry, Europe. Emerging Infectious Diseases. 2013;19(8):1339-1340. doi:10.3201/eid1908.120681.
APA Collignon, P., Aarestrup, F. M., Irwin, R., & McEwen, S. (2013). Human Deaths and Third-Generation Cephalosporin use in Poultry, Europe. Emerging Infectious Diseases, 19(8), 1339-1340. https://doi.org/10.3201/eid1908.120681.

Autochthonous Human Schistosomiasis, Malaysia [PDF - 301 KB - 2 pages]
B. Latif et al.
EID Latif B, Heo C, Razuin R, Shamalaa DV, Tappe D. Autochthonous Human Schistosomiasis, Malaysia. Emerg Infect Dis. 2013;19(8):1340-1341. https://doi.org/10.3201/eid1908.121710
AMA Latif B, Heo C, Razuin R, et al. Autochthonous Human Schistosomiasis, Malaysia. Emerging Infectious Diseases. 2013;19(8):1340-1341. doi:10.3201/eid1908.121710.
APA Latif, B., Heo, C., Razuin, R., Shamalaa, D. V., & Tappe, D. (2013). Autochthonous Human Schistosomiasis, Malaysia. Emerging Infectious Diseases, 19(8), 1340-1341. https://doi.org/10.3201/eid1908.121710.

Asian Musk Shrew as a Reservoir of Rat Hepatitis E Virus, China [PDF - 355 KB - 3 pages]
D. Guan et al.
EID Guan D, Li W, Su J, Fang L, Takeda N, Wakita T, et al. Asian Musk Shrew as a Reservoir of Rat Hepatitis E Virus, China. Emerg Infect Dis. 2013;19(8):1341-1343. https://doi.org/10.3201/eid1908.130069
AMA Guan D, Li W, Su J, et al. Asian Musk Shrew as a Reservoir of Rat Hepatitis E Virus, China. Emerging Infectious Diseases. 2013;19(8):1341-1343. doi:10.3201/eid1908.130069.
APA Guan, D., Li, W., Su, J., Fang, L., Takeda, N., Wakita, T....Ke, C. (2013). Asian Musk Shrew as a Reservoir of Rat Hepatitis E Virus, China. Emerging Infectious Diseases, 19(8), 1341-1343. https://doi.org/10.3201/eid1908.130069.

No Evidence for Hepatitis E Virus Genotype 3 Susceptibility in Rats [PDF - 353 KB - 3 pages]
T. Li et al.
EID Li T, Ami Y, Suzaki Y, Takeda N, Takaji W. No Evidence for Hepatitis E Virus Genotype 3 Susceptibility in Rats. Emerg Infect Dis. 2013;19(8):1343-1345. https://doi.org/10.3201/eid1908.130200
AMA Li T, Ami Y, Suzaki Y, et al. No Evidence for Hepatitis E Virus Genotype 3 Susceptibility in Rats. Emerging Infectious Diseases. 2013;19(8):1343-1345. doi:10.3201/eid1908.130200.
APA Li, T., Ami, Y., Suzaki, Y., Takeda, N., & Takaji, W. (2013). No Evidence for Hepatitis E Virus Genotype 3 Susceptibility in Rats. Emerging Infectious Diseases, 19(8), 1343-1345. https://doi.org/10.3201/eid1908.130200.

Fatal Case of Enterovirus 71 Infection and Rituximab Therapy, France, 2012 [PDF - 357 KB - 3 pages]
S. Kassab et al.
EID Kassab S, Saghi T, Boyer A, Lafon M, Gruson D, Lina B, et al. Fatal Case of Enterovirus 71 Infection and Rituximab Therapy, France, 2012. Emerg Infect Dis. 2013;19(8):1345-1347. https://doi.org/10.3201/eid1908.130202
AMA Kassab S, Saghi T, Boyer A, et al. Fatal Case of Enterovirus 71 Infection and Rituximab Therapy, France, 2012. Emerging Infectious Diseases. 2013;19(8):1345-1347. doi:10.3201/eid1908.130202.
APA Kassab, S., Saghi, T., Boyer, A., Lafon, M., Gruson, D., Lina, B....Schuffenecker, I. (2013). Fatal Case of Enterovirus 71 Infection and Rituximab Therapy, France, 2012. Emerging Infectious Diseases, 19(8), 1345-1347. https://doi.org/10.3201/eid1908.130202.

Norovirus Variant GII.4/Sydney/2012, Bangladesh [PDF - 348 KB - 2 pages]
M. Rahman et al.
EID Rahman M, Nahar S, Afrad M, Faruque A, Azim T. Norovirus Variant GII.4/Sydney/2012, Bangladesh. Emerg Infect Dis. 2013;19(8):1347-1348. https://doi.org/10.3201/eid1908.130227
AMA Rahman M, Nahar S, Afrad M, et al. Norovirus Variant GII.4/Sydney/2012, Bangladesh. Emerging Infectious Diseases. 2013;19(8):1347-1348. doi:10.3201/eid1908.130227.
APA Rahman, M., Nahar, S., Afrad, M., Faruque, A., & Azim, T. (2013). Norovirus Variant GII.4/Sydney/2012, Bangladesh. Emerging Infectious Diseases, 19(8), 1347-1348. https://doi.org/10.3201/eid1908.130227.

Norovirus GII.4/Sydney/2012 in Italy, Winter 2012–2013 [PDF - 268 KB - 2 pages]
G. M. Giammanco et al.
EID Giammanco GM, De Grazia S, Tummolo F, Bonura F, Calderaro A, Buonavoglia A, et al. Norovirus GII.4/Sydney/2012 in Italy, Winter 2012–2013. Emerg Infect Dis. 2013;19(8):1348-1349. https://doi.org/10.3201/eid1908.130619
AMA Giammanco GM, De Grazia S, Tummolo F, et al. Norovirus GII.4/Sydney/2012 in Italy, Winter 2012–2013. Emerging Infectious Diseases. 2013;19(8):1348-1349. doi:10.3201/eid1908.130619.
APA Giammanco, G. M., De Grazia, S., Tummolo, F., Bonura, F., Calderaro, A., Buonavoglia, A....Medici, M. C. (2013). Norovirus GII.4/Sydney/2012 in Italy, Winter 2012–2013. Emerging Infectious Diseases, 19(8), 1348-1349. https://doi.org/10.3201/eid1908.130619.

Group C Betacoronavirus in Bat Guano Fertilizer, Thailand [PDF - 351 KB - 3 pages]
S. Wacharapluesadee et al.
EID Wacharapluesadee S, Sintunawa C, Kaewpom T, Khongnomnan K, Olival KJ, Epstein JH, et al. Group C Betacoronavirus in Bat Guano Fertilizer, Thailand. Emerg Infect Dis. 2013;19(8):1349-1352. https://doi.org/10.3201/eid1908.130119
AMA Wacharapluesadee S, Sintunawa C, Kaewpom T, et al. Group C Betacoronavirus in Bat Guano Fertilizer, Thailand. Emerging Infectious Diseases. 2013;19(8):1349-1352. doi:10.3201/eid1908.130119.
APA Wacharapluesadee, S., Sintunawa, C., Kaewpom, T., Khongnomnan, K., Olival, K. J., Epstein, J. H....Hemachudha, T. (2013). Group C Betacoronavirus in Bat Guano Fertilizer, Thailand. Emerging Infectious Diseases, 19(8), 1349-1352. https://doi.org/10.3201/eid1908.130119.
Books and Media

Real-time PCR in Food Science: Current Technology and Applications [PDF - 317 KB - 2 pages]
D. F. Talkington
EID Talkington DF. Real-time PCR in Food Science: Current Technology and Applications. Emerg Infect Dis. 2013;19(8):1352-1353. https://doi.org/10.3201/eid1908.130524
AMA Talkington DF. Real-time PCR in Food Science: Current Technology and Applications. Emerging Infectious Diseases. 2013;19(8):1352-1353. doi:10.3201/eid1908.130524.
APA Talkington, D. F. (2013). Real-time PCR in Food Science: Current Technology and Applications. Emerging Infectious Diseases, 19(8), 1352-1353. https://doi.org/10.3201/eid1908.130524.

Public Health in East and Southeast Asia: Challenges and Opportunities in the Twenty-First Century [PDF - 247 KB - 1 page]
C. Ciesielski
EID Ciesielski C. Public Health in East and Southeast Asia: Challenges and Opportunities in the Twenty-First Century. Emerg Infect Dis. 2013;19(8):1353. https://doi.org/10.3201/eid1908.130721
AMA Ciesielski C. Public Health in East and Southeast Asia: Challenges and Opportunities in the Twenty-First Century. Emerging Infectious Diseases. 2013;19(8):1353. doi:10.3201/eid1908.130721.
APA Ciesielski, C. (2013). Public Health in East and Southeast Asia: Challenges and Opportunities in the Twenty-First Century. Emerging Infectious Diseases, 19(8), 1353. https://doi.org/10.3201/eid1908.130721.

PCR Detection of Microbial Pathogens [PDF - 255 KB - 1 page]
D. F. Talkington
EID Talkington DF. PCR Detection of Microbial Pathogens. Emerg Infect Dis. 2013;19(8):1353. https://doi.org/10.3201/eid1908.130528
AMA Talkington DF. PCR Detection of Microbial Pathogens. Emerging Infectious Diseases. 2013;19(8):1353. doi:10.3201/eid1908.130528.
APA Talkington, D. F. (2013). PCR Detection of Microbial Pathogens. Emerging Infectious Diseases, 19(8), 1353. https://doi.org/10.3201/eid1908.130528.
About the Cover

Ripped, Shucked, and Scattered [PDF - 258 KB - 2 pages]
P. Potter
EID Potter P. Ripped, Shucked, and Scattered. Emerg Infect Dis. 2013;19(8):1354-1355. https://doi.org/10.3201/eid1908.ac1908
AMA Potter P. Ripped, Shucked, and Scattered. Emerging Infectious Diseases. 2013;19(8):1354-1355. doi:10.3201/eid1908.ac1908.
APA Potter, P. (2013). Ripped, Shucked, and Scattered. Emerging Infectious Diseases, 19(8), 1354-1355. https://doi.org/10.3201/eid1908.ac1908.
Etymologia

Etymologia: Campylobacter [PDF - 239 KB - 1 page]
EID Etymologia: Campylobacter. Emerg Infect Dis. 2013;19(8):1313. https://doi.org/10.3201/eid1908.et1908
AMA Etymologia: Campylobacter. Emerging Infectious Diseases. 2013;19(8):1313. doi:10.3201/eid1908.et1908.
APA (2013). Etymologia: Campylobacter. Emerging Infectious Diseases, 19(8), 1313. https://doi.org/10.3201/eid1908.et1908.
Page created: July 26, 2013
Page updated: July 26, 2013
Page reviewed: July 26, 2013
The conclusions, findings, and opinions expressed by authors contributing to this journal do not necessarily reflect the official position of the U.S. Department of Health and Human Services, the Public Health Service, the Centers for Disease Control and Prevention, or the authors' affiliated institutions. Use of trade names is for identification only and does not imply endorsement by any of the groups named above.
edit_01 ScholarOne Submission Portal
Issue Select
GO
GO

Spotlight Topics

 

Get Email Updates

To receive email updates about this page, enter your email address:

file_external