Download Introduction - UC Davis School of Veterinary Medicine

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Oesophagostomum wikipedia , lookup

Orthohantavirus wikipedia , lookup

Ebola virus disease wikipedia , lookup

African trypanosomiasis wikipedia , lookup

Influenza A virus wikipedia , lookup

Sexually transmitted infection wikipedia , lookup

Middle East respiratory syndrome wikipedia , lookup

Marburg virus disease wikipedia , lookup

Bioterrorism wikipedia , lookup

Antiviral drug wikipedia , lookup

Neglected tropical diseases wikipedia , lookup

Henipavirus wikipedia , lookup

Eradication of infectious diseases wikipedia , lookup

Cross-species transmission wikipedia , lookup

Syndemic wikipedia , lookup

Pandemic wikipedia , lookup

Transcript
excerpted from
Reducing Pandemic Risk, Promoting Global Health
For the full report go to http://report.predict.global
PHOTO BY RICHARD GIDDINGS
INTRODUCTION
E
merging infectious diseases (EIDs) pose substantial threats to the health of
animals and people and economies globally. Over the past several decades,
the emergence and spread of viruses such as severe acute respiratory syndrome
(SARS) coronavirus, Ebola virus, and H5N1 and H1N1 pandemic influenza A viruses,
have had profound global health and economic impacts, illustrating our vulnerability
to the emergence and re-emergence of zoonotic diseases.
Zoonotic diseases result in millions of deaths annually, and the economic losses from a single
outbreak can amount to tens of billions of dollars (e.g. SARS; Lee and McKibbin 2004;
Keusch et al. 2009; World Bank 2012). Prevention and early control of outbreaks is key to
reducing their impact, but there remains a critical need to improve the capacity of developing
countries to effectively implement prevention and control activities as demonstrated by the
2014 Ebola virus disease outbreak in West Africa.
Zoonotic pathogens shared with wildlife or livestock account for the majority of emerging
infectious diseases of people, and viruses comprise 25-44% of these emerging and re-emerging
pathogens (Jones et al. 2008; Taylor et al. 2001). Because of their diversity and rapid evolution,
viruses, in particular RNA viruses, are perceived to pose the greatest global pandemic threat
(Morse 1995; Cleaveland et al. 2001; Pulliam 2008). For instance, two of the most devastating
pandemics in human history, the 1918 influenza pandemic (50 million deaths) and the HIV/
AIDS pandemic (35 million deaths as of 2012; Morens et al. 2008; Fauci and Folkers 2012),
were caused by zoonotic RNA viruses.
Despite greater recognition of EIDs on a global scale,
there is limited understanding of the underlying causes
for emergence and spread of zoonotic pathogens in
people (Murray and Daszak 2013). Factors associated
with emergence, re-emergence, and spread of pathogens
have been categorized into four broad domains worthy
of investigation: 1) genetic and biological factors of
pathogens and hosts, such as microbial adaptation
and human susceptibility to infection; 2) physical
environmental factors (e.g. climate variability and
weather); 3) ecological factors (e.g. land-use change);
and 4) social, political, and economic factors, including
poverty and animal and public health infrastructure
(Bogich et al. 2012; Daszak et al. 2001; Smolinski et
al. 2003). For example, factors such as rapid human
population growth, land use change, intensification of
livestock production, natural resource extraction, and
Indochinese
silvered langur
(Trachypithecus
germaini) kept as
a pet alongside
domestic dogs in
Cambodia.
PHOTO BY WILDLIFE CONSERVATION SOCIETY
REDUCING PANDEMIC RISK, PROMOTING GLOBAL HEALTH
17
Figure 1: Emergence of pandemic zoonotic disease. From Morse et al. 2012.
Stage 1 is a pre-emergence state, in which microbes are transmitted between animal
reservoirs. Ecological disturbances (e.g. due to land use change) modify the dynamics of
microbial transmission, which can lead to a heightened risk of pathogen spillover to wildlife
or livestock hosts.
Stage 2 is localized emergence, either through self-limiting spillover events (green peaks
and troughs, representing increases and decreases in the numbers of infected people with
time) or large-scale spillover (red peaks, representing spikes in the number of infected
people with time), that leads to person-to-person transmission for a few pathogen
generations.
In stage 3, some spillover events might lead to sustained person-to-person outbreaks,
international or global spread, and the emergence of a pandemic. The size, spread, and
potential effect of events increase from stage 1 to stage 3, but the frequency falls so
that full stage 3 pandemics are quite rare. By dissecting this process and analyzing the
interactions of the underlying drivers with the risk of spillover and spread, development
of a more structured approach to pandemic prevention is possible. The ultimate goal of
successful pandemic prevention is to target control at stage 1.
18
USAID PREDICT
the demands for global trade result in greater interactions between animals and people and
therefore more opportunities for cross species transmission and disease emergence (Daszak et
al. 2001; Smolinski et al. 2003). International travel has also contributed to the spread of many
infectious diseases regionally or globally with one of the most notable being the spread of SARS
coronavirus from Hong Kong to North America, Latin America, Europe, Asia, and Australia
resulting in 8,422 documented cases and 916 deaths worldwide (Heymann and Rodier 2004).
Wildlife play a major role in the emergence of infectious diseases in human populations. Analysis
of over 300 EID events occurring between 1940 and 2004 revealed that more than 70% of
emerging zoonotic pathogens originated in wildlife, and that the incidence of EID events caused
by wildlife pathogens has significantly increased over time (Jones et al. 2008). Certain wildlife
taxa are known to harbor a high proportion of zoonotic pathogens. Among assessed wildlife taxa,
zoonotic viruses have been most commonly detected in rodents, bats, primates, ungulates, and
birds (Woolhouse and Gowtage-Sequeria 2005; Calisher et al. 2006; Luis et al. 2013; Smith and
Wang 2013). Therefore, focusing surveillance efforts on these wildlife hosts is likely to enhance
the identification of viruses with pathogenic and disease emergence potential prior to spillover to
people (Parrish et al. 2008; Morse et al. 2012). Efforts in this realm must also be cognizant of the
need for clear messaging to not vilify wildlife, as the mechanisms for disease spillover tend to be
primarily human-induced, not animal-driven, and biodiversity and conservation goals must also
be maintained for a healthy world.
Rodents being sold
for consumption in
an African market.
The seriousness of these threats has led to the recognition
that a shift from a conventional, reactive approach toward
a proactive, predictive approach is necessary for EID
prevention and timely control (Figure 1; Karesh et al.
2012; Morse et al. 2012). Infrastructure improvements
and technological advances have dramatically and rapidly
improved our ability to identify high-risk interfaces for
disease transmission and to detect novel pathogens before
widespread spillover occurs. These advances include
improvements in information technology, molecular
diagnostics, and risk modeling. The challenge lies in
developing a strategic framework to identify zoonotic
pathogens of pandemic potential that have not yet emerged
in human populations.
To this end, USAID initiated the Emerging Pandemic
Threats (EPT) program in 2009. The initial goal of the
EPT program was to strengthen capacities in developing
countries to prevent, detect, and control infectious
diseases. Efforts were focused on early detection and
response to potentially high-consequence animal viruses
before they became significant public health threats in
regional “hotspots” for emerging diseases, such as central
Africa, South and Southeast Asia, and Latin America
(USAID 2013).
PHOTO BY PIOTR NASKRECKI
REDUCING PANDEMIC RISK, PROMOTING GLOBAL HEALTH
19
PREDICT, a surveillance and virus discovery component of the EPT program, focused on
building capacity to identify potential zoonotic disease threats at high-risk wildlife-human disease
transmission interfaces where diseases are most likely to emerge. PREDICT was implemented
in more than 20 countries, including Bangladesh, Bolivia, Brazil, Cambodia, Cameroon,
China, Democratic Republic of Congo, Gabon, Indonesia, Laos, Malaysia, Mexico, Nepal,
Peru, Republic of Congo, Rwanda, Tanzania, Thailand, Uganda, and Vietnam. The program
aimed to improve early detection and response to disease threats through five main objectives: 1)
strengthening disease surveillance systems; 2) improving viral discovery by developing laboratory
and disease outbreak response capacity; 3) characterizing drivers of pathogen spillover and spread
from animals to people; 4) optimizing predictive models for disease emergence and spread, and
5) deploying cutting-edge information management and communication tools to advance a more
integrated, global approach to emerging zoonotic diseases.
Through EPT, PREDICT worked to enhance in-country capacity for response and prevention
of pathogen spillovers by strengthening laboratory and diagnostic systems and increasing
connectivity among government sectors (wildlife, livestock, and human health) to operationalize
a One Health platform and promote early detection. In addition, PREDICT conducted
surveillance and monitoring for viruses in wildlife at high-risk animal-human interfaces to detect
potential zoonotic threats and prevent their emergence in human or livestock populations.
20
USAID PREDICT
REFERENCES
Bogich, T.L., R. Chunara, D. Scales, E. Chan, L. Pinheiro, A. Chmura,
D. Carroll, P. Daszak, and J.S. Brownstein. 2012. Preventing
pandemics via international development: a systems approach.
PLoS Medicine 9(12): e1001354.
Calisher, C.H., J.E. Childs, H.E. Field, K.V. Holmes, and T. Schountz.
2006. Bats: important reservoir hosts of emerging viruses.
Clinical Microbiology Reviews 19:531-545.
Cleaveland, S., M.K. Laurenson, and L.H. Taylor. 2001. Diseases
of humans and their domestic mammals: pathogen
characteristics, host range, and the risk of emergence.
Philosophical Transactions of the Royal Society of London B
356: 991-999.
Daszak, P., A.A. Cunningham, and A.D. Hyatt. 2001.
Anthropogenic environmental change and the emergence of
infectious diseases in wildlife. Acta Tropica 78: 103-116.
Fauci, A.S., and G.K. Folkers. 2012. The world must build
on three decades of scientific advances to enable a new
generation to live free of HIV/AIDS. Health Affairs 31:
1529–1536.
Heymann, D.L., and G. Rodier. 2004. SARS: lessons from a new
disease. In Learning from SARS: Preparing for the Next Disease
Outbreak: Workshop Summary 71-82.
Jones, K.E., N.G. Patel, M.A. Levy, A. Storeygard, D. Balk, J.L.
Gittleman, and P. Daszak. 2008. Global trends in emerging
infectious diseases. Nature 451: 990–994.
Kamins, A.O., O. Restif, Y. Ntiamoa-Baidu, R. Suu-Ire, D.T.S.
Hayman, A.A. Cunningham, J.N.L. Wood, and J. Rowcliffe. 2011.
Uncovering the fruit bat bushmeat commodity chain and
the true extent of fruit bat hunting in Ghana, West Africa.
Biological Conservation 144, 3000–3008. (doi:10.1016/j.
biocon.2011.09.003).
Karesh, W.B., A. Dobson, J.O. Lloyd-Smith, J. Lubroth, M.A. Dixon,
M. Bennett, S. Aldrich, T. Harrington, P. Furmenty, E.H. Loh, C.
Machalaba, M.J. Thomas, and D.L. Heymann. 2012. The ecology
of zoonoses: their natural and unnatural histories. The Lancet
380:1936-1945.
Keele, B.F., F. Van Heuverswyn, Y. Li, E. Bailes, J. Takehisa, M.L.
Santiago, F. Bibollet-Ruche, Y. Chen, L.V. Wain, F. Liegeois, S. Loul,
E.M. Ngole, Y. Bienvenue, E. Delaporte, J.F.Y. Brookfield, P.M.
Sharp, G.M. Shaw, M. Peeters, and B.H. Hahn. 2006. Chimpanzee
reservoirs of pandemic and nonpandemic HIV-1. Science
313:523–526.
Keusch, G.T., M. Pappaioanou, M.C. Gonzalez, K.A. Scott, and
P. Tsai. 2009. Sustaining global surveillance and response to
emerging zoonotic diseases. The National Academies Press,
Washington, D.C.
Korber B, M. Muldoon, J. Theiler, F. Gao, R. Gupta, A. Lapedes,
B.H. Hahn, S. Wolinsky, and T. Bhattacharya. 2000. Timing the
ancestor of HIV-1 pandemic strains. Science 288:1789–1796.
Lederberg J, R.E. Shope, S.C. and Oaks. 1992. Emerging
infections: microbial threats to health in the United States.
Washington, DC: National Academy Press.
Mickleburgh, S., K. Waylen, and P. Racey. 2009. Bats as
bushmeat: a global review. Oryx 43, 217–234. (doi:10.1017/
S0030605308000938).
Morens, D.M., G.K. Folkers, and A.S. Fauci. 2008. Emerging
infections: a perpetual challenge. Lancet Infectious Diseases 8:
710–719.
Morse, S.S. 1995. Factors in the emergence of infectious
diseases. Emerging Infectious Diseases 1: 7-15.
Morse, S.S., J. Mazet, M. Woolhouse, C.R. Parrish, D. Carroll, W.B.
Karesh, C. Zambrana-Torrelio, W.I. Lipkin, and P. Daszak. 2012.
Prediction and prevention of the next pandemic zoonosis.
The Lancet 380:1956-1965.
Murray, K.A., and P. Daszak. 2013. Human ecology in pathogenic
landscapes: two hypotheses on how land use change drives
viral emergence. Current Opinion in Virology 3:79-83.
Parrish, C.R., E.C. Holmes, D.M. Morens, E.C. Park, D.S. Burke,
C.H. Calisher, C.A. Laughlin, L.J. Saif, and P. Daszak. 2008.
Cross-species virus transmission and the emergence of
new epidemic diseases. Microbiology and Molecular Biology
Reviews 72: 457–470.
Pulliam, J.R.C. 2008. Viral host jumps: moving toward a
predictive framework. Ecohealth 5: 80–91.
Smith, I., and L-F. Wang. 2013. Bats and their virome: an
important source of emerging viruses capable of infecting
humans. Current Opinion in Virology 3:84-91.
Smolinski, M.S., M.A. Hamburg, J. Lederberg (editors), and
Committee on emerging microbial threats to health in the
21st Century. 2003. Microbial threats to health: emergence,
detection, and response. Washington, DC: The National
Academies Press.
Streicker, D.G., A.S. Turmelle, M.J. Vonhof, I.V. Kuzmin, G.F.
McCracken, and C.E. Rupprecht. 2010. Host phylogeny
constrains cross-species emergence and establishment of
rabies virus in bats. Science 329: 676–679.
Taylor, L.H., S.M. Latham, and M.E.J. Woolhouse. 2001. Risk
factors for human disease emergence. Philosophical
Transactions of the Royal Society of London B 356: 983-989.
United States Agency for International Development. 2013.
Emerging Pandemic Threats Program. Available at: http://www.
usaid.gov/news-information/fact-sheets/emerging-pandemicthreats-program.
Weiss, R.A., and A.J. McMichael. 2004. Social and environmental
risk factors in the emergence of infectious diseases. Nature
Medicine 10:S70-S76.
Woolhouse, M.E.J., and S. Gowtage-Sequeria. 2005. Host range
and emerging and reemerging pathogens. Emerging Infectious
Diseases 11(12): 1842-1847.
World Bank. 2012. People, Pathogens, and our Planet: The
Economics of One Health. Washington D.C.P
Lee, J-W, and W.J. McKibbin. 2004. Globalization and disease:
the case of SARS. Asian Economic Papers 3(1): 113-131.
Luis, A.D., D.T.S. Hayman, T.J. O’Shea, P.M. Cryan, A.T. Gilbert,
J.R.C. Pulliam, J.N. Mills, M.E. Timonin, C.K.R. Willis, A.A.
Cunningham, A.R. Fooks, C.E. Rupprecht, J.L.N. Wood, and C.T.
Webb. 2013. A comparison of bats and rodents as reservoirs
of zoonotic viruses: are bats special? Proceedings of the
Royal Society B 280: 20122753. http://dx.doi.org/10.1098/
rspb.2012.2753.
REDUCING PANDEMIC RISK, PROMOTING GLOBAL HEALTH
21