Public Health

Most outlets argue that before the United States can reopen safely, a new massive workforce needs to be in place to trace the contacts of people diagnosed with COVID-19, according to a report from the Johns Hopkins Center for Health Security.

Save Time On Research and Writing
Hire a Pro to Write You a 100% Plagiarism-Free Paper.
Get My Paper

Contact tracing is laborious, detailed and time-consuming detective work: Trained staff interview people who have been diagnosed with a contagious disease to figure out who they may have recently been in contact with. Then, they go tell those people they may have been exposed, sometimes encouraging them to quarantine themselves to prevent spreading the disease any further. Think of it as part public health work, and part investigation.  The technique is a “cornerstone” of preventative medicine, says Dr. Laura Breeher, medical director of occupational health services at the Mayo Clinic. “Contact tracing, it’s having a moment of glory right now with COVID because of the crucial importance of identifying those individuals who have been exposed quickly and isolating or quarantining them,” she says.

A long-used public health tool, contact tracing aims to break the chain of transmission of a contagious disease by identifying and alerting those who may have been exposed to it. Traditionally, a trained contact tracer will interview an individual diagnosed with the disease to determine all of their recent contacts, then reach out to those contacts to provide further information—which may include a recommendation to self-quarantine. In the past, this meticulous strategy has been used to help control Ebola, SARS, sexually transmitted infections, and tuberculosis, among other communicable diseases.

With the global outbreak of COVID-19, public health experts believe contact tracing will be a critical step for containing the virus, alongside social distancing and widespread testing. Many countries have already deployed extensive contact tracing, including New Zealand, Iceland, Taiwan, Singapore, and South Korea.

The United States, too, is gradually ramping up efforts—including a new hiring surge funded by the CDC, a Google/Apple tech partnership, and statewide programs. In New York, Gov. Andrew Cuomo is working with Bloomberg Philanthropies to launch their contact tracing program that will include online training from the Johns Hopkins Bloomberg School of Public Health.

Save Time On Research and Writing
Hire a Pro to Write You a 100% Plagiarism-Free Paper.
Get My Paper

“In the absence of a vaccine, we think this is really the big public health tool we have to control transmission of COVID-19,” according to Crystal Watson, lead author of the report and Assistant Professor in the Bloomberg School. “We need to push hard for this.”

Is this true that “contact tracing” is essential and valuable or is it an academic exercise? Write your discussion based on your readings on the applications and challenges of contact tracing; address the pro’s and con’s of contact tracing since this pandemic is no longer in its early stages of spread, the vast majority of infected individuals are asymptomatic and in New York, for example more that 60% of new infected individuals were sequestered “at home”.  Also take into consideration, privacy, costs, and issues with the timing and accuracy of current diagnostic tests.  With all of your passion, argument for or against massive contact tracing for COVID-19.

APA format, in-text citation, references include, 1 1/2 pages

Full Terms & Conditions of access and use can be found at
https://www.tandfonline.com/action/journalInformation?journalCode=fint20

Intelligence and National Security

ISSN: 0268-4527 (Print) 1743-9019 (Online) Journal homepage: https://www.tandfonline.com/loi/fint20

Improving ‘Five Eyes’ health security intelligence
capabilities: leadership and governance challenges

Patrick F. Walsh

To cite this article: Patrick F. Walsh (2020): Improving ‘Five Eyes’ health security intelligence
capabilities: leadership and governance challenges, Intelligence and National Security, DOI:
10.1080/02684527.2020.1750156

To link to this article: https://doi.org/10.1080/02684527.2020.1750156

Published online: 22 Apr 2020.

Submit your article to this journal

Article views: 676

View related articles

View Crossmark data

https://www.tandfonline.com/action/journalInformation?journalCode=fint20

https://www.tandfonline.com/loi/fint20

https://www.tandfonline.com/action/showCitFormats?doi=10.1080/02684527.2020.1750156

https://doi.org/10.1080/02684527.2020.1750156

https://www.tandfonline.com/action/authorSubmission?journalCode=fint20&show=instructions

https://www.tandfonline.com/action/authorSubmission?journalCode=fint20&show=instructions

https://www.tandfonline.com/doi/mlt/10.1080/02684527.2020.1750156

https://www.tandfonline.com/doi/mlt/10.1080/02684527.2020.1750156

http://crossmark.crossref.org/dialog/?doi=10.1080/02684527.2020.1750156&domain=pdf&date_stamp=2020-04-22

http://crossmark.crossref.org/dialog/?doi=10.1080/02684527.2020.1750156&domain=pdf&date_stamp=2020-04-22

ARTICLE

Improving ‘Five Eyes’ health security intelligence capabilities:
leadership and governance challenges
Patrick F. Walsh

ABSTRACT
This article explores common organizational pressure points for ‘Five Eyes’
intelligence communities in their ability to understand, prevent and dis-
rupt potential emerging bio-threats and risks. The acceleration in the
development of synthetic biology and biotechnology for legitimate mar-
kets (e.g. pharmaceuticals, food production and energy) is moving faster
than current intelligence communities’ ability to identify and understand
potential bio-threats and risks.

The article surveys several political leadership and intelligence govern-
ance challenges responsible for the current sub-optimal development of
health security intelligence capabilities and identifies possible policy sug-
gestions to ameliorate challenges.

  • Introduction
  • For some practitioners across ‘Five Eyes’ Intelligence Communities (IC) the mention of ‘health
    security’ and ‘intelligence’ together still seem a strange combination of words. For others in the IC
    even if ‘health security threats and risks’ are seen as legitimate collection and analytical priorities akin
    to others such as counter-terrorism and cyber–there is a reticence to invest in them. This somewhat
    disengaged perspective is explainable partly because there is yet no clear trajectory for bio-threats
    and risks. Disengagement is also arguably due–in part because historically ICs have not had a good
    understanding of bio-threats and risks from the Cold War up to and after the Coalition invasion of
    Iraq in 2003.

    Based on recent research, this article investigates how ‘Five Eyes’ partner countries can improve
    their health security intelligence capabilities to gain a deeper understanding of emerging bio-threats
    and risks. It concludes that sustained and coordinated capability improvements are largely a function
    of two aspects of intelligence governance: political leadership/external governance and internal IC
    governance issues. The article explores the significance of both factors before assessing the future
    outlook of health security intelligence capability over the next five years (2020-2025). Before
    addressing the central question of how to improve health security intelligence capability, however,
    it is important first to define how ‘health security intelligence’ is used in this article.

    Health security intelligence

    There are multiple interpretations on what constitutes ‘health security intelligence’ based largely on
    the diverse disciplinary backgrounds of scholars who seek to define it. In particular, the literature
    highlights this diversity between how public health specialists, clinicians, epidemiologists use the
    term compared to intelligence and security specialists. Aldis provides a useful discussion on the lack

    CONTACT Patrick F. Walsh pawalsh@csu.edu.au

    INTELLIGENCE AND NATIONAL SECURITY
    https://doi.org/10.1080/02684527.2020.1750156

    2020 Informa UK Limited, trading as Taylor & Francis Group

    http://www.tandfonline.com

    https://crossmark.crossref.org/dialog/?doi=10.1080/02684527.2020.1750156&domain=pdf&date_stamp=2020-04-12

    of agreement and understanding, particularly between developed and developing countries, on the
    concept of health security. He stated that the term ‘health security,’ like ‘biosecurity,’

    ‘sits at the intersection of several disciplines which do not share a commonmethodology (e.g., practitioners from
    security studies, foreign policy, IR [international relations], development theory and practice of UN agencies).’1

    As in biosecurity, inconsistencies arise in how health security is defined. Bernard has referred to
    a ‘tribalism’ between the public health and security sectors, which has prevented both from under-
    standing each other and perceiving common priorities.2 Historically, this has led to different
    disciplines talking past each other and not seeing how their perspectives can provide potentially
    a more accurate and inclusive definition of health security intelligence. As a former intelligence
    analyst and intelligence studies scholar, my own perspectives of health security intelligence has
    evolved over time. In my most recent work in this area – Intelligence, Biosecurity and Bioterrorism the
    focus is largely on biosecurity and the intentional weaponising of biology.3 However, I concluded
    that biosecurity should be seen as one aspect of a broader health security community–the other
    being public health. The human security agenda, which emerged from the international relations
    and security studies literature in the 1990s provides a unifying concept for understanding health
    security intelligence. Human security scholars sought to broaden traditional notions of security–
    beyond being solely about war and peace between states. Human security also emphasised the
    security of individuals within and between states, and included security against pandemics, poverty,
    access to education, and safe water amongst other basic human rights.4

    In the context of a growing human security agenda, several scholars working in public health,
    particularly clinicians and sociologists begun to argue increasingly the response to global pandemics
    (HIV, SARs, H1N1, Zika and Ebola) as not just regional/global health emergencies, but also grave
    health security events.5 Such events not only had the potential to make many sick, but their
    implications globally – particularly for developing countries with fragile public sector infrastructure –
    had security implications for states involved. The 2014–16 Ebola outbreak in Western Africa, which
    killed 10,000 people illustrates how a major public health emergency not only has catastrophic
    health outcomes, but can also destabilise the capacities of nation-states to respond to such crises.6

    Similarly, since 2018 another Ebola outbreak has occurred in the Democratic Republic of Congo’s
    North Kivu and Ituri provinces. Further at the time of writing, there has been an outbreak of corona
    virus (COVID-19) first reported fromWuhan, China on 31 December 2019, which also underscores the
    fragility of even economically wealthy states’ ability to respond to regional and global health security
    crises.

    In the case of the recent Ebola outbreak, there have been over 1900 deaths and although over
    80,000 people have been immunized with a new Ebola vaccine, public health responders have been
    attacked by rebel militia and prevented from tracing and treating population in surrounding areas.
    This situation provides another example of the critical link between public health and security.7 In
    summary, if naturally occurring public health incidents and security are linked then it follows that
    intentional public health (biosecurity incidents involving bio-crimes and bio-terrorism) need to be
    seen not only as intelligence and security priorities, but also clearly as public health incidents. For
    example, the release of anthrax into a metropolitan transport hub, will require both intelligence and
    public health personnel coming together to assess whether the threat/risk is primarily a biosecurity
    or public health emergency. ‘Health security’ thus becomes a useful over-arching terminology to
    capture both biosecurity and public health dimensions of health incidents in order to assess the
    security implications of both. Clearly not all health security incidents as they unfold will rely on
    a similar composition of public health, intelligence and security personnel. However, both dimen-
    sions will remain co-dependent on each other as discussed in greater detail below.

    With health security defined, the last aspect of ‘health security intelligence’ requiring clarification
    is ‘intelligence.’ Again depending on the context (e.g. national security, policing, military, private
    sector), there are many diverse perspectives on what ‘intelligence’ is and does. Leaving aside these
    debates, and for the objectives of this article, I argue that intelligence has at least three unique

    2 P. F. WALSH

    defining attributes: secrecy, surveillance and the security environment.8 All these terms are relative in
    the sense that in certain contexts (e.g. the Central Intelligence Agency, CIA) some intelligence
    collected would be considered highly secret with profound impacts on a given nation’s interests if
    it was shared widely – perhaps even in that nation’s intelligence community. Whereas a local policing
    agency could possess material on file classified as ‘secret’ or ‘protected’ (or more accurately
    privileged information), but the consequences of its release may be less deleterious. The point is
    secrecy is a relative term, but in its broadest sense most intelligence practice regardless of context
    cannot operate without a relative level of secrecy. The second term surveillance underscores another
    primary objective of all intelligence practice regardless of context. Intelligence activities can include
    an array of activities including research and information management. However, intelligence would
    not be intelligence without an ability to warn decision-makers about impending risks and threats.
    This requires active and sometimes passive surveillance of threat actors and risks in the security
    environment, which is the third defining characteristic of intelligence. The security environment again
    depending on the context in which intelligence is practiced is large, diverse and ever changing. It
    includes an open-ended list of threats such as state based military threats, terrorism, cyber, failed
    states and bio-threats and risks–the subject of this article.

    With health security intelligence now defined, I now turn to a brief assessment of emerging bio-
    threats and risk across ‘Five Eyes’ countries. Given limited space, the focus here is squarely on post 9/
    11 emerging bio-threats and risks because it is the capability of ‘Five Eyes’ countries in managing
    both contemporary and emerging threats and risks that is most germane to our discussion. This is not
    to suggest that understanding the evolution of bio-threat and risk trajectories pre-9/11 is not
    relevant to situating current or emerging threats. Indeed the rapid development of modern micro-
    biology and the industrialisation of various biological weapons in state based programs (e.g. Soviet
    Union, USA, UK and Canada) since 1945 – and the attempt by rogue states (particularly Iraq but also
    Iran, North Korea and Syria) to do the same – provides a foundation upon which more recent bio-
    threat and risk typologies can be understood. Additionally, the rapid development of synthetic
    biology and biotechnology from the 1990s to the present and the potential weaponisation of
    aspects of these technological advancements should also be examined against the broader historical
    context of bio-threats and risk from 1945, throughout the Cold War and the immediate pre 9/
    11 period. But these developments are chronicled in detail elsewhere for readers seeking this
    background material.9

    Discussion here of contemporary and emerging bio-threats and risks starts with the Amerithrax
    incident of 2001. I argue Amerithrax represented a water shed moment in helping ICs and policy-
    makers recalibrate their understanding about future bio-threats trajectories. In September and
    October 2001, seven envelopes containing a dried powder form of anthrax spores were posted to
    several media outlets and to the US Senate offices of Senators Thomas Daschle and Patrick Leahy.
    The letters resulted in 22 cases of anthrax–five of which led to fatal inhalational anthrax. The anthrax
    letters also resulted in the contamination and closure of several major US postal offices.10 The
    Amerithrax attack came only a week after 9/11 so the United States was already unsettled about
    the possibility of more terrorist attacks. The incident resulted in a 7 year long investigation, where the
    US Department of Justice (DOJ) determined a single spore batch created by anthrax specialist
    Dr Bruce E Ivins at the US Army Medical Research Institute of Infectious Diseases (USAMRID) was
    the parent material for the letter spores. In July 2008 Ivins committed suicide before being indicted.
    However, since the FBI officially closed the investigation in 2010 several biologists and chemists
    disagree on whether the Bureau got the right perpetrator based on the presence of silicon and tin
    coating on the anthrax spores. In the opinion of some experts this suggest a greater complexity of
    manufacturing beyond the scope of what Ivins could do in his lab. Additionally, earlier in the
    investigation another army research scientist Steven Hatfield was targeted, but later exonerated
    with the DOJ paying a 4.6 million dollar legal settlement to the scientist.11 The DOJ has not changed
    its position that Ivins was responsible and complex investigations such as Amerithrax underscore the
    difficulties in attribution of individuals involved in bio-attacks.

    INTELLIGENCE AND NATIONAL SECURITY 3

    Amerithrax while not a major casualty attack was significant for a number of reasons. First it
    influenced IC assessments about how easy it would be for non-state actors such as Al Qaeda to
    produce and disseminate biological agents as weapons. Amerithrax showed the difficulties an
    anthrax scientist had in producing the spores in ideal laboratory conditions. Al Qaeda and other
    jihadist groups may have held an interest in developing bio-weapons, yet for the most part had
    limited access to similar scientific expertise or a safe laboratory setting for their development. In
    summary, the incident injected more subtlety and complexity into policy discussions given that
    during the late 1990 s and early 2000 s a dominant strand in the political discourse was that it would
    be relatively easily for terrorist groups to develop bio-weapons. Second, and more importantly,
    Amerithrax demonstrated that the terrorist may be ‘an insider’ – for example a scientist going rogue.
    Amerithrax therefore, ignited another arc of the debate about the most likely profile of bio-threat
    actors that continues to the present.

    Despite the lessons contained in Amerithrax, a comprehensive understanding of contemporary
    bio-threats and risks remains unclear across the ‘Five Eyes’ countries and the broader scientific
    community. However, there are two broad threat/risk typologies: stolen biological agents and dual
    use research and synthetic biology, which illustrate where concerns exist for the potential malicious
    use of biology into the future. I will focus more on the latter as this area is currently of greatest
    concern by ICs and the scientific community.

    Stolen biological agents
    Stealing a controlled biological agent appears rare due to increased bio-safety regulations and
    control of physical space (BSL 3 and 4 labs) that has developed since the Amerithrax incident in all
    ‘Five Eyes’ countries.12 Legislation and bio-safety regulations, which also include more stringent
    security background checks for employees working with select biological agents (such as anthrax,
    tularaemia, the plague) has provided greater protection. However, despite enhanced security
    measures in the US and to some extent the UK, there have been a number of handling and
    transportation safety episodes (particularly in the 2014 USD–2015), which demonstrate vulnerabil-
    ities in safety practices remain. Such episodes involved the US Department of Defense and the
    Centers for Disease Control and Prevention (CDC) and included safety protocols not being followed
    fully in transporting select biological agents across the United States and internationally as well as
    the shipping of bio-culture equipment from BSL 3 and 4 labs to lower safety rated labs without being
    sufficiently decontaminated. In July 2019, safety concerns have again been highlighted when the US
    Army Medical Research Institute of Infectious Diseases (USAMRID) – a key centre for the develop-
    ment of biological countermeasures against bioterrorism suspended operations under the authority
    of the Federal Select Agent Program for failing to meet federal biosafety requirements.13

    Despite bio-safety lapses and given more stringent physical and personnel safety measures, it
    seems that this bio threat/risk typology (stolen biological agents) is likely to remain very low across
    ‘Five Eyes’ countries. While it is unclear if there is a criminal market for the theft of controlled bio-
    agents from secure labs, it would seem that organised criminal groups would have other compara-
    tively less risky and more immediately profitable markets to pursue such as drug trafficking. In
    particular, given the attribution of many controlled biological agents in BSL 3 to 4 labs can be
    assessed forensically, criminal interest in stealing such substances is likely to be further dissuaded.14

    The other main threat scenario of a terrorist group breaking in and stealing a controlled bio-agent
    also seems less likely.

    While 9/11 represented a sophisticated and impactful attack modality, most lethal attacks since
    (at least in Western countries) have been simpler using readily available materials and weapons such
    as improvised explosive devices, vehicles and knives. None of these require the same expertise as
    stealing, handling or using controlled biological substances. Nonetheless, there are two areas of
    potential concern in this bio-threat/risk typology (stolen biological agents), which ‘Five Eyes’ coun-
    tries still need to collect and assess against: intellectual property theft by an ‘insider’ and the potential
    for theft from some BSL 3 to 4 labs in fragile or vulnerable states.

    4 P. F. WALSH

    Given the exponential rise in synthetic biology, biotechnology research and commercialisation
    both in the public and private sector – state and non-state sponsored theft of intellectual property
    will likely increase. Such theft will be linked in many instances to broader cyber security and
    information security threats that are currently impacting on ‘Five Eyes’ economies. Again, the IC’s
    ability to manage the rising complexity in the broader cyber threat space remains a challenge. It is
    likely then that ICs will continue to confront expertise and knowledge gaps in how the bio-economy
    is being attacked and IP stolen by cyber-threat actors.15

    The second area of concern (theft of controlled biological substances) in fragile or vulnerable
    states, particularly those in the Middle East and Africa (e.g. Iraq, Syria, Pakistan) is also another area
    where greater focus by ‘Five Eyes’ ICs is warranted. Such locations intersect hotspots for jihadist
    inspired instability. It is possible that bio-safety lapses in research facilities near unstable locations or
    radicalised individuals working within biological institutions could steal bio-agents for terrorist
    attacks. Legitimate biological research facilities in vulnerable states require ongoing multi-lateral
    global security efforts to promote bio-safety. ‘Five Eyes’ defence and foreign ministries have funded
    bio-safety initiatives in vulnerable states in the past, but there is scope for a better coordinated
    approach to bilateral and multilateral programs into the future.16

    Dual use research and synthetic biology
    Dual use research is defined here as research of dangerous biological agents that might be
    weaponised, and the publication of that research, which theoretically could be disseminated to bio-
    criminals or terrorists for their own nefarious objectives.17 There are a growing number of perspec-
    tives of whether criminals and terrorists will exploit biotechnology and synthetic biology research
    developed for legitimate purposes (e.g. health care, energy and food supply) for harm or profit.18 The
    debates are complex and care should be exercised in not over-simplifying them, however it is
    possible to discern two main intellectual strands from them. These are: technological (determinism)
    and socio-technological perspectives. The technological determinists argue the upsurge in biotech-
    nological advancements will make the access, use and exploitation of relevant knowledge and skills
    easier and cheaper for those with malevolent intentions.19 Some of the consequences of the
    industrialisation of biology adds weight to their arguments about easier access and use of biotech-
    nology. For example, the entire human genome sequenced by 2013 took a team of scientists
    13 years and 500 USD million to identify 20,500 genes. Today, the human genome can be sequenced
    in a day using bench top equipment costing around 1000 USD. 20

    In contrast, the socio-technologists while not discounting that criminals and terrorists may exploit
    advances in synthetic biology and technology – argue that access to knowledge and skill is not the
    same as being able to adeptly exploit these in order to produce a bio-weapon. Accessing, develop-
    ing, manipulating or exploiting knowledge and skills still requires significant training – often in more
    than one of the biological sciences (e.g. microbiology, molecular biology, genetics, and medicine).
    Training requires not just being able to read and replicate the latest experiments but is also
    dependent on applying skills intuitively in a social and collaborative sense frequently working with
    a team of scientists. In summary, the ability of a threat actor to weaponise any dual use and synthetic
    biology is dependent also on the inter-play of other social, economic and psychological variables.21

    Both intellectual perspectives are not sterile academic debates. They matter because they inform
    how policy makers, scientists and the intelligence community conceptualise how dual use research
    and synthetic biology might be exploited by threat actors.

    Shortly, the focus will turn to a brief summary of examples of bio-threats that may arise with the
    exploitation of dual use research and synthetic biology. First however, in order to understand how bio-
    threats may manifest, it is useful to distinguish between two broad threat trajectories for dual use
    research and synthetic biology. The first (‘insiders’) refers to skilled scientists, researchers and techni-
    cians, who use knowledge and skills to create dangerous bio-agents under the veil of legitimate
    research. The second (‘outsiders’) refers to the exploitation of legitimate advances in biology, synthetic
    biology and biotechnology for dangerous and criminal ends. In the second case, it could be one or

    INTELLIGENCE AND NATIONAL SECURITY 5

    more of the recent ‘gain of function’ (GOF) experiments, where genetic material has beenmanipulated
    to encourage the development of dangerous pathogens thought to be rare, absent or dormant in the
    general population (e.g. small pox, reconstruction of 1918 influenza virus) – or experiments that make
    biological agents more virulent (e.g. mouse pox, horse pox, H5N1 avian influenza transmissible
    between human surrogates such as ferrets).22 Such experiments are often promoted as benefiting
    medicine by identifying vaccines or better ones against hard to treat viruses and bacteria. The
    November 2018 announcement by Chinese scientist He Jiankui that he had allegedly created the
    word’s first genetically edited babies that could be resistant to HIV is another recent example of many
    scientific, ethical and security concerns raised about both GOF and gene editing experiments.23

    Given both the insider and outsider threat trajectories, the critical question remains what types of
    specific bio-threats are evolving? There is no satisfactory answer to this question, and debates
    continue amongst scholars, scientists and to some extent within the ICs about where bio-threats
    are likely to emerge within dual use research and synthetic biology. I argue in this article and
    elsewhere that assessing bio-threats and risks will rely on a more strategic and comprehensive
    assembling of scientific evidence on how developments in biotechnology and synthetic capabilities
    could be exploited by threat actors. Additionally, identifying threats and risks will also rely on ICs
    gaining a greater understanding of the intentions of particular actors to select capabilities as
    weapons or for profit over other more conventional sources. Assessing intention and capability to
    misuse dual use research and synthetic biology remains difficult because the science is moving
    rapidly – which in turn makes it difficult to get a ‘fix’ on where vulnerabilities for malevolent
    exploitation are located. Such difficulties are also a product of some of the ongoing capability issues
    across ‘Five Eyes’ countries in adapting to technologically enabled threats more broadly (e.g. cyber).

    Leaving aside the ongoing difficulties in assessing emerging bio-threats and risks, I summarise here
    three threat cluster areas where ‘Five Eyes’ countries might consider developing better knowledge of
    bio-threats and risks. These are: bio-unabombers, identify theft and bio-piracy. As noted earlier, biotech-
    nology is big business particularly in the United States and other ‘Five Eyes’ countries.24 Given the
    massive acceleration in biotechnology and synthetic biological sciences – probability alone suggests
    that there will be more individuals (some mentally unstable) with a range of grievances (e.g. personal,
    psychological, political and religious) working in the biological sciences and some may escalate these
    to acts of violence. Disgruntled insiders (scientists), who can make a synthetic organism and sidestep
    theft of natural organisms under lock and key in highly regulated high containment labs are uniquely
    placed under the disguise of a bigger legitimate scientific project to become bio-unabombers. As
    discussed earlier, the alleged involvement of scientist Bruce Ivins in the Amerithrax case is illustrative
    here.25 Secondly, ‘identify theft’ – long an enabler of crime and terrorism in other contexts is also likely
    to be a developing concern. The increase in DNA holdings in various government departments and the
    private sector provide greater opportunities to steal or manipulate DNA. For example, it is possible that
    individuals could assume someone’s identity by stealing their DNA, or leaving someone else’s blood
    and saliva at a crime scene by genetic cloning to frustrate national security (terrorist) and law
    enforcement investigations.26 Thirdly, bio-piracy could become a greater bio-threat and risk in ‘Five
    Eyes’ countries. Recent developments in drugs, vaccines andmedicines can bemanipulated. Moreover,
    in the last few years the development of gene editing techniques like CRISPR, Finger Nuclease and
    Talen could be exploited criminally.27 Though currently there is an increasing ‘hype’ over the threat
    significance of CRISPR–further fueled by a national assessment by the US IC during the Obama
    Administration, which declared that it could be a national security threat.28 But it remains difficult to
    know what if any threat actors, for example jihadists, ultra-nationalists, anarchists, bio-hackers, envir-
    onmentally motivated individuals will exploit gene editing techniques like CRISPR.

  • Capability gaps
  • As noted earlier, there remain both gaps in scientific evidence and intelligence on what may
    constitute emerging bio-threats and risks, particularly those arising from dual use research and

    6 P. F. WALSH

    synthetic biology. The acceleration of scientific discovery in biotechnology and synthetic biology
    make assessing threats and risks in any comprehensive way difficult and this is unlikely to change in
    the future. However, it is possible that some threats and risks might be ‘knowable’ – at least
    sufficiently to provide decision-makers with warning. However, this will rely on each ‘Five Eyes’
    country’s ability to reflect on current capabilities to assess bio-threats and risks in order to identify
    capability gaps. With the above outline on potential bio-threat and risk trajectories in mind, this
    section explores what are the principle capability gaps across the ‘Five Eyes’ countries. The final
    section (outlook) will assess how, or indeed whether the gaps identified can be closed in the near
    future.

    Before describing the key capability gaps within and across ‘Five Eyes’ intelligence communities, it
    is useful to clarify first how I will be using the term ‘capability.’ Based on previous research on
    intelligence reform issues, I argue that capability is the sum of two inter-related factors. The first
    I refer to as core intelligence processes and the second key enabling activities. Both factors are critical
    to effective intelligence capabilities at the agency and community levels.29 In simple terms, core
    intelligence processes include the key stages in the production process of intelligence products that
    hopefully will influence and support decision-maker’s policy deliberations. These stages are: tasking
    and coordination, collection, analysis, production and evaluation. The core intelligence processes
    can be thought of as the functional aspects of intelligence production. In contrast, the key enabling
    activities provide the structural foundation under which the core intelligence processes can operate
    effectively. Intelligence product is not developed in a vacuum and it is the agency and community
    key enabling activities (governance, ICT, human resources, legislation and research) that support the
    ‘intelligence production line.’30 In this section, I argue that health intelligence capability gaps include
    a range of issues across core intelligence processes and key enabling activities. In the remaining
    space, I will summarise some of the key capability gaps in both the core intelligence processes and
    key enabling activities.

    Core intelligence processes

    There are capability gap issues in three core intelligence processes: tasking and coordination,
    collection and analysis. The gaps in each area impact upon one another and as will be demonstrated
    shortly also influence how capability gaps arise in the key enabling activities. Tasking and coordina-
    tion include a range of activities by decision-makers and internally within ICs about how intelligence
    assets particularly collection and analytical resources are to be applied against various threats and
    risks. Given intelligence is a decision-maker’s support tool, priorities about the tasking and coordina-
    tion of intelligence resources are formally determined at heads of government and cabinet level in
    ‘Five Eyes’ countries. The identification of other collection and assessment priorities not currently
    tasked by senior cabinet ministers obviously are also made by ‘Five Eyes’ intelligence communities
    on a regular basis. However, some recent (albeit tentative) research on tasking and coordination
    against emerging bio-threats and risks suggest that both decision-makers and the intelligence
    communities themselves remain relatively disengaged from these threats.31 This is not to suggest
    some bio-threat and risk issues are not getting tasking and coordination attention both by govern-
    ments and as part of the routine internal mechanisms of ‘Five Eyes’ communities. There still seems to
    be some interest in state sponsored WMD (as it relates to bio) and bioterrorism, however this interest
    is limited compared to earlier periods such as Amerithrax and the faulty WMD assessments that
    preluded the coalition invasion of Iraq in 2003. There are any number of comparatively more ‘urgent’
    threats and risks vying for the attention of ‘Five Eyes’ leaders such as conventional terrorism, cyber,
    the resurgence of Russia, the rise of China and North Korea’s nuclear proliferation. In comparison,
    emerging bio-threats and risks are akin to cyber threats – they are deeply technical, which com-
    pounds the difficulty by IC and decision-makers understanding them. Many are also currently
    assessed (rightly or wrongly) as low probability/high impact threats, which influences the amount
    of tasking such issues receive by both political decision-makers and from within the ‘Five Eyes’

    INTELLIGENCE AND NATIONAL SECURITY 7

    intelligence machinery. Low levels of tasking have a knock on effect in the other two stages of the
    core intelligence processes: collection and analysis.

    From the perspective of collection, the first capability issue is that low or ad hoc levels of tasking
    impact on ‘Five Eyes’ countries’ ability to develop better risk and threat methodologies that would
    allow more accurate assessment of emerging bio-threats and risks. Poor ability to assess bio-threats
    and risks circuitously results in even lower and ad hoc levels of tasking and therefore collection
    against these issues by both decision-makers and within ICs. This latter point also raises a broader
    question of whether current collection capabilities are suited ideally for gaining better calibration
    against emerging bio-threats and risks. As noted earlier, emerging bio-threats and risks may well
    arise from dual use research and synthetic biology, but given these areas are predominantly focused
    on legitimate research and commercial development, will traditional and more covert IC collection
    platforms (e.g. SIGINT, HUMINT and Geo-spatial intelligence) be best placed to ‘discover’ the illicit
    weaponisation of biology in these sectors? The answer to this question is not clear. Clearly threat and
    risk context will be important. Much will depend on the nature of the bio-threat and risk. Is it local,
    national or transnational and is it at the planning, operational or attack stage? Given the emerging
    nature of some bio-threats and risks, traditional collection platforms may be less helpful (at least
    initially) compared to other open source intelligence such as scientific peer reviewed sources, social
    media, epidemiology and microbial forensics in improving assessments of such threats and risks.32

    However, current research suggests that while progress has been made in ‘Five Eyes’ countries to
    link into and learn from these open sources, a more systematic and strategic approach needs to be
    adopted to accessing bio-threat and risk related open source intelligence.33 Additionally, capability
    improvements are needed in many open source intelligence areas derived from the scientific
    community such as microbial forensics. Collection capabilities in the bio-threat and risk context
    are also limited by other issues impacting on broader collection efforts across all ‘Five Eyes’ com-
    munities. Of particular importance is the post Snowden impact of threat actors ‘going dark’ due to
    them becoming aware of how to reduce their exposure to traditional IC collection platforms. The
    increased encryption of communications via Apps and the use of the dark web present ongoing
    collection capability challenges – not just in the bio-threat and risk area but across the entire security
    environment.34

    The third core intelligence process area where there are capability gaps is analysis. Similar to the
    collection capability issues discussed some of the gaps in analysis on bio-threats and risks are not
    unique to assessing these issues. They are symptomatic of broader concerns and debates across ‘Five
    Eyes’ countries about how one improves analytical capabilities.35

    The way ICs build analytical capability is influenced by external and internal factors. As noted
    earlier in the discussion on tasking and coordination if political leaders do not demonstrate an
    appetite for reading intelligence assessments on specialised or less ‘bread and butter’ issues then the
    capability investment in these areas becomes depleted. While the exact headcount of analysts
    working on bio-threat and risk issues across all ‘Five Eyes’ countries is unclear, discussions with
    people with more intimate knowledge of the ICs work flows suggest that numbers within agencies
    can be counted with the fingers on one or two hands.36 It seems that many analysts working on bio-
    threat and risk issues are likely working on other accounts, including the broader suite of WMD issues
    (e.g. chemical and nuclear). There are pockets of excellence and specialisation for example, in the
    United States the FBI WMD Directorate Outreach program and in recent years within the Defense
    Intelligence Agency (DIA). However, analytical expertise in emerging bio-threats and risks generally
    remains shallow and fragmented within and across agencies. In summary, if decision-makers are not
    calling for product on bio-threats and risks, then the internal leadership within ICs are less compelled
    to increase the depth and breadth of analytical expertise in these areas. This in turn further reduces
    both the skills and repository of knowledge in bio-threat and risk areas. This phenomenon of
    intelligence leaders reducing the depth and breadth of analytical coverage in some areas has played
    out in different agencies across ‘Five Eyes’ countries. For example, CIA agency leaders have in recent
    years focused on getting more analysts to work flexibly across a range of areas to meet the ever

    8 P. F. WALSH

    increasing demand of current intelligence rather than necessarily cultivate subject matter
    expertise.37 Flexibility has some merits, though it can prevent a deeper more sustained investment
    in analysts with expertise in one specialised area, who arguably can write better current and strategic
    products based on a deeper knowledge of the subject matter.38

    Leadership decisions about where to invest in building analytical capability leads us to the main key
    enabling activity related capability issue – governance. As discussed earlier, governance is only one of
    five key enabling activities (governance, ICT, human resources, legislation and research). In recent
    research, I found that there were also capability gaps in the other four key enabling activities, including
    in human resources and legislation.39 However, given space limitations the focus here is on governance
    issues as they are themost consequential for understanding capability gaps arising in other key enabling
    activity areas. For example, poor governance can lead to poor human resource planning as well as
    inadequate or not fit for purpose legislation in which to manage more effectively bio-threats and risks.

    Governance

    Intelligence governance is a term I have used in research on IC capability issues since 2011. From
    research resulting in the book Intelligence and Intelligence Analysis, I developed an effective intelligence
    framework. The framework showed that sound and adaptable IC capability relied on how well both
    core intelligence processes and key enabling activities worked together to produce intelligence out-
    comes decision-makers needed.40 In this research governance was defined as ‘attributes and rules
    pertaining to strong sustainable leadership, doctrine design, evaluation, and effective coordination,
    cooperation and integration of intelligence processes.’41 In simple terms, governance is about effective
    leadership and it has an external dimension (‘the political leadership’) and an internal one (heads of IC
    agencies and communities). It’s logical that sound governance (in both its external and internal
    dimensions) should result in better intelligence capabilities. It is also follows that effective capabilities
    allow the creation of a working environment where higher standard products inform decision-making.

    Research into the governance capability issues as they relate to bio-threats and risks is ongoing so
    the observations here are generalised, incomplete and require further validation across each ‘Five
    Eyes’ country. However, based on discussions with some IC personnel and secondary literature there
    is evidence that there are both external and internal governance capability issues. These have
    a knock-on effect on how bio-threats and risks are coordinated and prioritised across each ‘Five
    Eyes’ country. At this stage, the evidence is strongest in the US IC, where both public health
    stakeholders and current and former members of IC and the broader security community have
    been calling in recent years for better leadership both at the political and IC agency level over
    managing bio-threats and risks. The United States Blue Ribbon Panel Report of 2015 is one example
    of concerns raised about the lack of consistent leadership of broader health security issues including
    the role of the IC.42 In the United States, several executive policy declarations on health security and
    bioterrorism have been produced by successive administrations from George W Bush to the current
    Trump administrations. However, what seems to bemissing is a clear articulation of how any national
    health security strategy will be operationalised generally and how political and IC leadership will
    coordinate effectively the limited resources to collect and assess against bio-threats and risks.
    Additionally, and less clear, is how do ICs reach out more explicitly and strategically to important
    stakeholders (e.g. scientists, public health specialists, clinicians) in order to improve the quality of
    collection and analytical efforts? In the final section below, the paper revisits the capability gaps
    discussed here to determine if they can be closed. It also provides an assessment of where ‘Five Eyes’
    ICs capabilities might be on managing bio-threats and risks in five years’ time.

  • Outlook
  • Turning first to improving tasking and coordination processes, in the next five years this will continue to
    be a difficult ad hoc process across ‘Five Eyes’ countries. Care needs to be exercised in generalising

    INTELLIGENCE AND NATIONAL SECURITY 9

    across each ‘Five Eyes’ country. Further detailed research is also required to assess how each ‘Five Eyes’
    IC arranges tasking and coordination of bio-threats and risks. However, as noted earlier, research to
    date suggest in all five countries while cross IC and broader government coordination has improved,
    decisionmakers and therefore the IC leadership do not appear to routinely task on emerging bio-threat
    and risk issues. The recent Trump Administrations’ national biodefense strategy and the UK’s Biological
    Security Strategy both include rhetoric about the need of greater coordination of IC, public health and
    other stakeholders.43 However in each case, there is little detail about how these strategic documents
    will be operationalised. In the UK case, at the time of writing (August 2019) the Joint Committee on the
    National Security Strategy has commenced an inquiry into the how the strategy is addressing
    biosecurity and human health. The launch of the inquiry does suggest that further work is required
    to fully implement it across the UK government. In particular, the US and UK policy documents, do not
    identify a leadership position within the ICs, who could be responsible for ensuring there is regular
    tasking and effective coordination of efforts in the collection and assessment of bio-threats and risks. As
    discussed earlier, the core intelligence process of tasking and coordination is closely linked to the key
    enabling activity of governance. Governance is crucially about effective political and internal leadership
    of ICs. Hence tasking and coordination on bio-threats and risks is intricately linked to whether
    improvements can be made in governance over the next five years.

    On the other two core intelligence processes (collection and analysis) and leaving aside the
    stultifying effect of a relatively low engagement in bio-threats and risks by the political leadership of
    ‘Five Eyes’ countries–there are opportunities to build on existing collection platforms in ways that
    would improve information feeds on these issues. It may not be that ICs need to build additional
    expensive collection platforms just for bio-threats and risks. In some cases, using existing covert and
    open source intelligence that are already being applied against comparatively more conventional
    threats and risks (terrorism and organised crime) will likely provide opportunities to collect inciden-
    tally against bio-threats and risks.

    Governance

    Whether tasking and coordination can be improved depends first on whether a renewed interest in
    bio-threats and risks can be garnered from the political leadership in ‘Five Eyes’ countries in the
    absence of an actual bio-attack. In the United States, the Trump Administration has developed an
    adversarial and mistrusting attitude to the US IC – making it difficult for IC leaders to discern which
    intelligence priorities the President is interested on a day to day basis let alone emerging bio-threat
    and risk issues. Additionally, effective tasking and coordination is further compounded by President
    Trump’s focus mainly on border security and political matters including the US Congress impeach-
    ment inquiry and earlier the Mueller special investigation into whether he and members of his
    campaign team colluded with Russia in ways that influenced the outcome of the 2016 presidential
    election. Combined these factors demonstrate a lack of a sustained political commitment and
    interest in improving tasking and coordination, collection and assessments on emerging bio-
    threats and risks.

    Similarly in the UK, at the time of writing the government continues to be pre-occupied with
    securing a Brexit deal from the EU, and it remains to be seen how cabinet ministers will focus on
    implementing both the Biological Security Strategy and how the strategy might improve the way
    the UK IC tasks, coordinates, collects and assesses emerging bio-threats and risks. Australia,
    Canada and New Zealand all have the necessary IC and public health bureaucratic institutions
    in place to theoretically respond to emerging bio-threats and risks. However, there is no evidence
    that these governments have developed national health security strategic plans, which clearly
    articulate how their ICs will improve their understanding of emerging bio-threats; or better
    integrate their resources with important stakeholders such as scientists and the public health
    sector.

    10 P. F. WALSH

    At the time of writing, there are major reform changes underway in the Australian Intelligence
    Community (AIC) – including the establishment in 2018 of a new Office of National Intelligence to
    better coordinate the Community. Other reforms include the establishment of a capability fund to
    help the AIC identify where future investment needs to take place.44 Though given the amount of
    other work ONI has to address across the Australian intelligence enterprise, it is unlikely the current
    reform agenda in the near term will produce a national health security strategic plan that includes
    reflection on how the AIC can play a role in understanding emerging bio-threats and risks.

    Further, given the short electoral cycles in ‘Five Eyes’ countries and the perceived low probability
    and high impact nature of bio-threats and risks by many policy makers – it seems unlikely political
    leaders of each ‘Five Eyes’ country will address in any significant way current governance issues
    relating to emerging bio-threats and risks. But the rapidly changing nature of all threats and risks
    particularly those technologically enabled ones such as cyber, underscore the need for stronger
    governance that can identify and remedy IC capability gaps in understanding bio-threats and risks.
    As a start, better governance at the political level could be facilitated by formalised high-level
    coordination. For example, the establishment of a Health Security Coordination Council led pre-
    ferably by head of government (or the very least a senior cabinet minister) would provide a more
    coherent ‘whole of government’ approach to tasking and coordination of bio-threats and risks as
    well as identifying other capability gaps discussed in the paper. Current US and UK strategic plans
    outlined earlier seem to place senior bureaucrats largely in charge of national level biosecurity,
    biodefense committees. A senior cabinet minister (from either a health or security portfolio) is a more
    appropriate choice to chair such a council. The council should also contain in addition to heads of IC,
    other senior stakeholders including public and animal health specialists, and scientific advisors.
    A broad and diverse council membership is critical to ensure national health security strategies
    articulate a whole of government (including ICs) approach. The next ideal step in improving
    governance arrangements would be for each stakeholder to develop operational plans to implement
    government tasking priorities as well as ensuring effective coordination of resources.

    For each ‘Five Eyes’ country a senior sub-cabinet level national health security intelligence officer
    should also be established. This appointee would have authority to identify and implement tasking
    and coordination, collection and analytical priorities across the entire IC. The national health security
    intelligence officer would liaise with other important stakeholders that are part of the health security
    coordination council; including public/animal health, scientists and their counter parts in other ‘Five
    Eyes’ countries to gain a national and international understanding of emerging bio-threats and risk
    as well as identifying IC capability gaps.

    Closing collection capability gaps

    A national health security intelligence officer sitting across the entire IC enterprise should be able to
    implement coherent and coordinated national collection plans for bio-threats and risks that help
    both collection and assessment agencies identify what role they can play in overall collection efforts.
    A national health security intelligence officer can consider for example, how traditional collection
    platforms (SIGINT, HUMINT, Geospatial Intelligence) are best applied to retrieve information about
    both current and emerging bio-threats. Threat and risk evolution will also be an important con-
    sideration in what collection capabilities need to be deployed. In other words, at what develop-
    mental stage is the threat? Can it be prevented, disrupted or only treated after the event? It is quite
    possible as noted earlier, that traditional collection platforms might be initially applied against more
    ‘here and now’ threats such as terrorism or organised crime – yet may also reveal in some
    circumstances an interest by these threat actors or their associates in bio-threats and risks. This in
    turn would allow a more focused application of other specialised, scientific collection tools such as
    microbial forensics.

    Arguably though with many bio-threats and risks, there are likely to be an even larger volume of
    relevant information accessible to the IC from non-security stakeholders on the prevention,

    INTELLIGENCE AND NATIONAL SECURITY 11

    disruption and treatment of bio-threats and risks. While ‘Five Eyes’ ICs have in the past reached out to
    some stakeholders, a more strategic and explicit approach to building trusted external expert
    networks would improve collection efforts, particularly in areas such as dual use research of concern
    and synthetic biology. The potential lists of relevant stakeholders is almost endless. They can be
    found at the multi-lateral, bilateral, national, regional and local levels.45 At a multi-lateral level,
    historically important security measures such as the Biological and Toxin Weapons Convention
    (BTWC) will continue to provide ICs with information, albeit not always reliable about states’
    adherence to the Convention. Other recent multilateral biosafety and biosecurity initiatives such
    as the Global Health Security Agenda established by the Obama Administration could provide
    opportunities for ‘Five Eyes’ ICs to increase awareness of potential bio-threats and risks in addition
    of course to their public health capacity-building effects.46

    In each ‘Five Eyes’ country, albeit some with more capacity (e.g. US and UK) defence departments
    or their allied defence research divisions there are biodefence research capabilities which the ICs
    could tap into more regularly. In the case of the United States, other government research bodies
    such as the Intelligence Advanced Research Projects Activity (IARPA) also sponsor biodefense
    research that is directly relevant to how ICs may participate in the future in the identification and
    prevention of weaponised dual use research and synthetic biology. For example, in July 2017, IARPA
    commissioned the Project Felix program to fund research that can detect signals of bio-engineering
    including types of changes, location and possibly in the future where changes were made. Such work
    could be used in cases where suspicious circumstances suggest the malicious exploitation of bio-
    engineered material is occurring.47 Leaving aside what the defence, security and intelligence
    research groups offer the IC in this area, it’s clear that the medical community (epidemiology and
    public health) are also naturally indispensable partners for the ICs. Medical, public health and
    epidemiological knowledge provides the ICs with a common understanding of how to identify
    natural and intentional disease outbreaks and in some cases can inform attribution about what
    threat actors may be responsible. Finally, first responders have critical knowledge about the physical
    environment(s), where an attack occurs as well as forensic evidence, which can help inform tactical
    and operational intelligence collection and analysis.

    Analysis

    In any intelligence agency, an effective operation of all core intelligence processes requires an almost
    symbiotic relationship between collection and analysis. Uncoordinated and insufficient strategic and
    operational collection planning will reduce the ability of analysts to improve the reliability and
    validity of their products. There are no ‘off the shelf’ solutions to current analytical capability gaps
    discussed earlier. Evaluating whether the analytical workforce is ‘fit for purpose’ as far as assessing
    bio-threats and risks is really a sub-set of a bigger issue about whether ‘Five Eyes’ ICs have the right
    mix of subject matter expert vs generalist analysts – against a whole range of current and emerging
    threats. Significant investment has gone into training and education of analysts across ‘Five Eyes’
    countries in the military, national security and to lesser degree the law enforcement community.48

    But debates continue within ICs and amongst teachers and scholars what skills, knowledge and
    competencies entry level analysts need to deal with the fast paced changes in the security environ-
    ment. Less consideration is currently given to how to deploy subject matter experts in specialised
    areas such as bio-threats and risks. Do ‘Five Eyes’ ICs try to develop specialised expertise in this
    subject internally or should this work be out-sourced to trusted, security cleared outsiders who
    might be scientists?

    I argue that it shouldn’t be an either/or decision given that all ‘Five Eyes’ ICs need to maintain
    corporate repositories of knowledge in specialised areas – yet at the same time it would be
    impossible for those few individuals responsible for interpreting bio-threats and risks across the
    entire IC to be experts in all bio-threat matters. Given resources are limited, another aspect of
    workforce analytical training of staff on bio-threat and risk issues is that heads of agencies and ICs

    12 P. F. WALSH

    need to also think carefully about what role subject matter experts may play. Logically some
    should be deployed in prevention roles that can provide strategic early warning of potential bio-
    threat and risk trajectories. Others would need to focus on disruption of evolving bio-threats and
    risk in operational settings; whilst others would be useful in assessing the treatment of threats and
    risks.

    Further evidence is required, but IC leaders currently do not appear to be planning in any
    systematic way the future bio-threat and risk analytical workforce. As part of work force planning,
    it is unclear whether IC leaders are considering in any strategic way what analytical capability gaps
    are best met by external stakeholders (scientists, clinicians, and public health officials)–and how to
    integrate more seamlessly this knowledge into the closed working environment within ICs. Finally,
    with respect to sub-optimal analytical capabilities, further reflection by IC leaders needs to be given
    on how potentially a diverse and voluminous amount of information collected across health security
    sector (clinicians, epidemiologists, scientists, military, national security, law enforcement, animal and
    agricultural health) can best be interrogated rapidly by analysts. Again the ability to quickly makes
    sense of large data sets and intelligence is obviously a problem writ large for ICs not just an issue to
    be resolved in the bio-threat and risk context.

    In summary, this article argues it is unlikely that each ‘Five Eyes’ country will improve dramatically
    their IC capabilities for managing bio-threats and risks in the next five years (2020–25). Barring
    a catastrophic bio-attack by a terrorist group or other health security crisis resulting in high mortality
    rates, bio-threats and risks will continue to be seen as low probability-high impact threats for political
    decision-makers. This translates within the ICs as at best a watching brief of bio-threats and risks and
    at worse them being largely ignored. At this time, bio-threats and risks only get ad-hoc attention by
    the ‘Five Eyes’ ICs. For example, ICs may focus periodically on them as part of broader WMD threat
    assessments ahead of hosting a major international event.

    In addition to low levels of political tasking and coordination, systematic and proactive
    approaches to managing bio-threats and risks at the IC level are not likely without stronger IC
    governance by its leaders. This includes a national health security intelligence officer, who can
    champion the threat and risk area and help agencies close capability gaps. In some ‘Five Eyes’
    countries there has been some capability improvements since 9/11, however, based on available
    evidence, I assess no substantial move towards a greater strategic approach to health security
    intelligence across ‘the Five Eyes’ countries during the next five years. There is no available public
    knowledge that there are any impending substantive bio-threats and risks, particularly emanating
    from dual use research and synthetic biology. Nonetheless the rise in the cyber threat trajectory and
    the struggle by ICs to understand it provides a good lesson that building capability takes time. It is
    now time to invest in capability to better manage potential and emerging bio-threats and risks.
    Hopefully the COVID-19 pandemic might provide a catalyst to build the kinds of capabilities in ICs
    discussed throughout the article.

  • Notes
  • 1. Aldis, “Health Security as a Public Health Concept,” 370.
    2. Bernard, “Health and National Security,” 157.
    3. Walsh, Intelligence Biosecurity and Bioterrorism.
    4. The Human Security Centre, The Human Security Centre.
    5. See for example, Elbe, “Pandemic Security,” 163–173.
    6. Heymann, “Global Health Security: the Wider Lessons from the West WHO.” “Ebola Virus Disease in West Africa –

    The First Nine Months of the Epidemic and Forward Projections,” 1481–1495; “Global Health Security: the Wider
    Lessons from the West African Ebola Virus Disease Epidemic,” 1884–1901; Marston, “Ebola Response Impact on
    Public Health Programs, West Africa 2014–2017”; WHO, “Ebola Virus Disease in West Africa – The First Nine
    Months of the Epidemic and Forward Projections,” 1481–1495.

    7. Soucheray, “With New Cases, Katwa Remains Epicenter of Ebola Outbreak.”
    8. Walsh, Intelligence and Intelligence Analysis, 29–32.

    INTELLIGENCE AND NATIONAL SECURITY 13

    9. Alibek, Biohazard: The Chilling True Story of the Largest Covert Biological Weapons Program in the World; Balmer,
    Britain and Biological Warfare. Expert Advice and Science Policy; Christopher, et al., “Biological Warfare: A Historical
    Perspective,” 412–417; Geissler, & Ellis van Courtland Moon, (Eds.). Biological and Toxin Weapons: Research,
    Development and Use from the Middle Ages to 1945; Koblentz, Living Weapons; and Walsh, Intelligence, Biosecurity
    and Bioterrorism.

    10. Walsh, Intelligence, Biosecurity and Bioterrorism, 30–31.
    11. Broad and Shane, “Scientist’s Analysis Disputes FBI Closing of Anthrax Case.”
    12. In BSL-3 to 4 rated labs, scientists work on pathogens that can cause serious or potentially lethal disease.

    Generally, the most lethal agents, where there is no vaccine or an unknown risk of transmission are worked on in
    BSL-4 labs.

    13. Lizotte, “Research Halted at USAMRID Over Biosecurity Issues,” CDC, Report on the Potential Exposure to Anthrax;
    CDC, 90 Day Internal Review of the Division of Select Agents and Toxins; Dennis Brady and Lena Sun, ‘FDA Found
    More than Smallpox Vials in Storage Room.’ Washington Post. 16 July 2014, https://www.washingtonpost.com/
    national/health-science/fda-found-more-than-smallpox-vials-in-storage-room/2014/07/16/850d4b12-0d22-
    11e4-8341-b8072b1e7348_story.html?utm_term=.978241b9d1f8.

    14. Walsh, “Managing Intelligence and Responding to Emerging Threats,” 837–57; Walsh, “Managing Emerging
    Health Security Threats Since 9/11: The Role of Intelligence,” 341–67.

    15. Koblentz and Mazanec, “Viral Warfare: The Security Implications of Cyber and Biological Weapons,” 418–34; and
    Murch, “Emerging New Discipline to Help Safeguard the Bioeconomy.”

    16. For a brief discussion on some of the bilateral and multi-lateral biosafety programs that ‘Five Eyes’ countries
    have been active in see, Walsh, Intelligence, Biosecurity and Bioterrorism, 179–231.

    17. Ibid., 179–231.
    18. Tucker, Innovation, Dual Use and Security; Suk et al., “Dual Use Research and Technological Diffusion.

    Reconsidering the Bioterrorism Threat Spectrum,” 1–3; Gerstein, Bioterror in the 21st Century: Emerging Threats
    in a New Global Environment; National Academy of Sciences, Human Genome Editing. Science Ethics and
    Governance; Arnason, “Synthetic Biology between Self-Regulation and Public Discourse,” 246–56.

    19. Chyba, “Biotechnology and the Challenge to Arms Control,” 11–17; Carlson, “The Pace and Proliferation of
    Biological Technologies,” 203–14; Petro and Carus, “Biological Threat Characterisation Research,” 295–308.

    20. Walsh, Intelligence, Biosecurity and Bioterrorism, 44.
    21. Kathleen Vogel’s work on the non-technical aspects of bio-threats provide a good background to all the non-

    technical variables that should be considered in assessing bio-threats and risks. See, Vogel, “Biodefense,” 227–55;
    “Intelligent Assessment: Putting Emerging Biotechnology Threats in Context,” 45–54; “Necessary Interventions.
    Expertise and Experiments in Bioweapons Intelligence Assessments,” 61–88; and, Phantom Menace or Looming
    Danger?.

    22. Walsh, Intelligence, Biosecurity and Bioterrorism, 41–51.
    23. Kolata, “Chinese Scientist Claims to Use Crispr to Make First Edited Babies”.
    24. In the US, easily over 2 million people are employed with over 73,000 businesses working across range of

    biosciences (medicine, agriculture, pharmaceuticals, research). Similarly in Australia, around 48,000 Australians
    are employed in the biotechnology sector with the sector estimated to obtain a 4.4% annual growth reaching
    8.67 billion AUD in revenues by 2021. See, Battelle. Battelle/Bio State Bioscience, Jobs, Investments and Innovation;
    Ausbiotech, Australia’s Biotechnology Organisation (website), 2018. https://www.ausbiotech.org/biotechnol
    ogy-industry/fast-facts.

    25. Walsh, Intelligence, Biosecurity and Bioterrorism, 51.
    26. Ibid., 41.
    27. Willingham, “A Fresh Threat: Will CAS9 Lead to CRISPR Bioweapons?”; National Academy of Sciences. Human

    Genome Editing. Science Ethics and Governance; Revill, “Could Gene Editing Tools Such as CRISPR be Used as
    a Biological Weapon.”

    28. Willingham, “A Fresh Threat”; Clapper, Statement for the Record. Worldwide Threat Assessment of the US
    Intelligence Community.

    29. A more detailed discussion of these factors and how they relate to theorising about effective intelligence
    frameworks can be found in Walsh, Intelligence and Intelligence Analysis, 131–147; Walsh, “Building Better
    Intelligence Frameworks Through Effective Governance,” 123–42.

    30. Ibid.
    31. Walsh, Intelligence, Biosecurity and Bioterrorism, 59–89.
    32. Ibid., 89–121.
    33. Ibid.
    34. Walsh and “Rethinking “Five Eyes” Security Intelligence Collection Policies and Practice Post Snowden,” 345–68;

    Chertoff, “A public policy perspective of the Dark Web,” 26–38; and Chen, Bioterrorism and Knowledge Mapping
    Dark Web Exploring and Data Mining the Dark Side of the Web, 335–67.

    35. Walsh, & Ratcliffe, “Strategic Criminal Intelligence Education: A Collaborative Approach,” 152–66. Walsh,
    “Teaching intelligence in the twenty-first century: towards an evidence-based approach for curriculum design,”

    14 P. F. WALSH

    https://www.washingtonpost.com/national/health-science/fda-found-more-than-smallpox-vials-in-storage-room/2014/07/16/850d4b12-0d22-11e4-8341-b8072b1e7348_story.html?utm_term=.978241b9d1f8

    https://www.washingtonpost.com/national/health-science/fda-found-more-than-smallpox-vials-in-storage-room/2014/07/16/850d4b12-0d22-11e4-8341-b8072b1e7348_story.html?utm_term=.978241b9d1f8

    https://www.washingtonpost.com/national/health-science/fda-found-more-than-smallpox-vials-in-storage-room/2014/07/16/850d4b12-0d22-11e4-8341-b8072b1e7348_story.html?utm_term=.978241b9d1f8

    https://www.ausbiotech.org/biotechnology-industry/fast-facts

    https://www.ausbiotech.org/biotechnology-industry/fast-facts

    1005–1021; Harrison, & Et al., “Tradecraft to Standards – Moving Criminal Intelligence Practice to a Profession
    through the Development of Criminal Intelligence Training and Development Continuum,” 1–13.

    36. Walsh, Intelligence, Biosecurity and Bioterrorism, 127–128.
    37. Gentry and Gordon, Strategic Warning Intelligence, 223.
    38. Ibid., 218.
    39. Walsh, Intelligence, Biosecurity and Bioterrorism.
    40. Walsh, Intelligence and Intelligence Analysis, 31–152.
    41. Ibid., 149.
    42. Blue Ribbon Study Panel, Blue Ribbon Study Panel on Biodefense. A National Blueprint for Biodefense: Leadership

    and Major Reform Needed to Optimise Efforts; Blue Ribbon Study Panel, Biodefense Indicators One Year Later.
    Events Outpacing Federal Efforts to Defend the Nation.

    43. White House, National Biodefense Strategy; HMG. UK Biological Security Strategy.
    44. For a discussion of the main aspects of the reform agenda currently underway in the AIC, see my ISA Paper:

    “Transforming the Australian Intelligence Community: Mapping Change, Impact and Governance Challenges,”
    paper given at the 60th International Studies Association Conference, 27 August 2019.

    45. For a detailed discussion of what non-security stakeholders can bring to the IC see: Walsh, Intelligence, Biosecurity
    and Bioterrorism, 179–231.

    46. Ibid., 192–3.
    47. Ibid., 183.
    48. Walsh, “Teaching intelligence in the twenty-first century,” 1005–21.

  • Disclosure statement
  • No potential conflict of interest was reported by the author.

  • Notes on contributor
  • Patrick F. Walsh, PhD, is a former intelligence analyst, and has worked in Australia’s national security and law
    enforcement environments. He is an associate professor in Intelligence and Security Studies, Charles Sturt University,
    Australia. He is also a consultant to government agencies on intelligence reform and capability issues. His research
    grants and publications focus on a range of areas related to intelligence capability; including but not limited to:
    governance, leadership, intelligence and ethics, biosecurity and cyber. His last book, Intelligence, Biosecurity and
    Bioterrorism (UK: Palgrave Macmillan, 2018), examined the challenges for ‘Five Eyes’ intelligence communities in
    understanding and managing emerging bio-threats and risks.

  • Bibliography
  • Aldis, W. “Health Security as A Public Health Concept: A Critical Analysis.” Health Policy and Planning 23, no. 6 (2008):
    369–375. doi:10.1093/heapol/czn030.

    Alibek, K. Biohazard: The Chilling True Story of the Largest Covert Biological Weapons Program in the World- Told from the
    inside by the Man Who Ran It. New York: Random House, 1999.

    Arnason, G. “Synthetic Biology between Self-Regulation and Public Discourse: Ethical Issues and the Many Roles of the
    Ethicist.” Cambridge Quarterly of Healthcare Ethics 26, no. 2 (2017): 246–256. doi:10.1017/S0963180116000840.

    Ausbiotech. 2018. Australia’s Biotechnology Organisation (Website). https://www.ausbiotech.org/biotechnology-industry
    /fast-facts.

    Balmer, B. B., and B. Warfare. Expert Advice and Science Policy, 1930-65. Basingstoke, UK: Palgrave Macmillan, 2001.
    Battelle. Battelle/Bio State Bioscience, Jobs, Investments and Innovation. Columbus, OH: Battelle, 2014.
    Bernard, K. “Health and National Security: A Contemporary Collision of Cultures.” Biosecurity and Bioterrorism: Biodefense

    Strategy, Practice, and Science 11, no. 2 (2013): 157–162. doi:10.1089/bsp.2013.8522.
    Blue Ribbon Study Panel. Blue Ribbon Study Panel on Biodefense. A National Blueprint for Biodefense: Leadership and Major

    Reform Needed to Optimise Efforts. Washington, D.C: Hudson Institute for Policy Studies, 2015.
    Blue Ribbon Study Panel. Biodefense Indicators One Year Later. Events Outpacing Federal Efforts to Defend the Nation.

    Arlington, VA.: Potomac Institute for Policy Studies, 2016.
    Brady, D., and L. Sun. 2014. “FDA Found More than Smallpox Vials in Storage Room.” Washington Post, July 16. https://

    www.washingtonpost.com/national/health-science/fda-found-more-than-smallpox-vials-in-storage-room/2014/07/
    16/850d4b12-0d22-11e4-8341-b8072b1e7348_story.html?utm_term=.978241b9d1f8.

    Broad, W., and S. Shane. 2011. “Scientist’s Analysis Disputes FBI Closing of Anthrax Case.” The New York Times, October 9.

    INTELLIGENCE AND NATIONAL SECURITY 15

    https://doi.org/10.1093/heapol/czn030

    https://doi.org/10.1017/S0963180116000840

    https://www.ausbiotech.org/biotechnology-industry/fast-facts

    https://www.ausbiotech.org/biotechnology-industry/fast-facts

    https://doi.org/10.1089/bsp.2013.8522

    https://www.washingtonpost.com/national/health-science/fda-found-more-than-smallpox-vials-in-storage-room/2014/07/16/850d4b12-0d22-11e4-8341-b8072b1e7348_story.html?utm_term=.978241b9d1f8

    https://www.washingtonpost.com/national/health-science/fda-found-more-than-smallpox-vials-in-storage-room/2014/07/16/850d4b12-0d22-11e4-8341-b8072b1e7348_story.html?utm_term=.978241b9d1f8

    https://www.washingtonpost.com/national/health-science/fda-found-more-than-smallpox-vials-in-storage-room/2014/07/16/850d4b12-0d22-11e4-8341-b8072b1e7348_story.html?utm_term=.978241b9d1f8

    Carlson, R. “The Pace and Proliferation of Biological Technologies.” Biosecurity and Bioterrorism: Biodefense Strategy,
    Practice, and Science 1, no. 3 (2004): 203–214. doi:10.1089/153871303769201851.

    CDC. Report on the Potential Exposure to Anthrax. Atlanta, GA: CDC, 2014.
    CDC. 90 Day Internal Review of the Division of Select Agents and Toxins. Atlanta, GA: CDC, 2015.
    Chen, H. Bioterrorism and Knowledge Mapping Dark Web Exploring and Data Mining the Dark Side of the Web. New York,

    N.Y.: Springer, 2011.
    Chertoff, M. “A Public Policy Perspective of the Dark Web.” Journal of Cyber Policy 2, no. 1 (2017): 26–38. doi:10.1080/

    23738871.2017.1298643.
    Christopher, G. W., T. J. Cieslak, and E. M. Eitzen Jr. “Biological Warfare: A Historical Perspective.” Journal of the American

    Medical Association 278, no. 5 (1997): 412–417. doi:10.1001/jama.1997.03550050074036.
    Chyba, C. “Biotechnology and the Challenge to Arms Control.” Arms Control Today 36 (2016): 11–17.
    Clapper, J. Statement for the Record. Worldwide Threat Assessment of the US Intelligence Community. Armed Services

    Committee, 2016. Washington,DC.
    Elbe, S. “Pandemic Security.” In The Routledge Handbook of New Security Studies, edited by J. Peter Burgess, 163–173.

    Abingdon, UK: Routledge, 2010.
    Elbe, S. “Pandemic Security.” In The Routledge Handbook of New Security Studies, edited by J. Peter Burgess, 163–173.

    Abingdon, UK: Routledge, 2010.
    Geissler, E., and J. E. van Courtland Moon, eds. Biological and Toxin Weapons: Research, Development and Use from the

    Middle Ages to 1945. New York: Oxford University Press, 1999.
    Gentry, J., and J. Gordon. Strategic Warning Intelligence. Washington DC: Georgetown University Press, 2019.
    Gerstein, D. Bioterror in the 21st Century: Emerging Threats in a New Global Environment. New York: Naval Institute Press, 2010.
    Harrison, M., P. F. Walsh, S. Lysons-Smith, D. Truong, C. Horan, and R. Jabbour. “Tradecraft to Standards– Moving

    Criminal Intelligence Practice to a Profession through the Development of Criminal Intelligence Training and
    Development Continuum.” Policing (2018): 1–13.

    Heymann, D. et al. Global Health Security: The Wider Lessons from the West WHO.” “Ebola Virus Disease in West Africa –
    the First Nine Months of the Epidemic and Forward Projections. The New England Journal of Medicine 371 (2015):
    1481–1495.

    Heymann, D., L. Chen, K. Takemi, D. P. Fidler, J. W. Tappero, M. J. Thomas, T. A. Kenyon. “Global Health Security: the Wider
    Lessons from the West African Ebola Virus Disease Epidemic.” The Lancet, no. 9980 (2015): 1884–1901. doi:10.1016/
    S0140-6736(15)60858-3

    HMG. UK Biological Security Strategy. London: HMG, 2018.
    The Human Security Centre. The Human Security Centre: Human Security Report. Oxford, UK: Oxford University Press,

    2005.
    Koblentz, G. Living Weapons. New York: Cornell University Press, 2009.
    Koblentz, G., and B. Mazanec. “Viral Warfare: The Security Implications of Cyber and Biological Weapons.” Comparative

    Strategy 32, no. 5 (2013): 418–434. doi:10.1080/01495933.2013.821845.
    Kolata, G., S. Wee, P. Belluck. 2018. “Chinese Scientist Claims to Use Crispr to Make First Edited Babies.” The New York

    Times, November 26. https://www.nytimes.com/2018/11/26/health/gene-editing-babies-china.html.
    Lizotte, S. 2019. “Research Halted at USAMRID over Biosecurity Issues.” Global Biodefense, August 5. https://globalbio

    defense.com/2019/08/05/research-halted-at-usamriid-over-biosafety-issues/CDC, Report on the Potential Exposure
    to Anthrax.

    Marston, B., E. Kainne Dokubo, A. van Steelandt, L. Martel, D. Williams, S. Hersey, A. Jambai, S. Keita, T. G. Nyenswah, J.
    Redd. “Ebola Response Impact on Public Health Programs, West Africa 2014-2017” Emerging Infectious Diseases
    Journal 28, no. Supplement (2017): 25–31.

    Murch, R., W. So, W. Buchholz, S. Raman, J. Peccoud. “Emerging New Discipline to Help Safeguard the Bioeconomy.”
    Frontiers in Bioengineering and Biotechnology 6, no. 39 (2018): 1–6. doi:10.3389/fbioe.2018.00039.

    National Academy of Sciences. Human Genome Editing. Science Ethics and Governance. Washington, D.C: National
    Academies Press, 2017.

    Petro, J., and S. Carus. “Biological Threat Characterisation Research: A Critical Component of National Biodefense,
    Biosecurity, and Bioterrorism.” Biodefense Strategy, Practice and Science 3 (2005): 295–308.

    Revill, J. 2017. “Could Gene Editing Tools Such as CRISPR Be Used as a Biological Weapon.” The Conversation, August 31.
    https://theconversation.com/could-gene-editing-tools-such-as-crispr-be-used-as-a-biological-weapon-82187?utm_
    source=twitter&utm_medium=twitterbutton.

    Soucheray, S. 2019. “With New Cases, Katwa Remains Epicenter of Ebola Outbreak.” CIDRAP News, February 18. http://
    www.cidrap.umn.edu/news-perspective/2019/02/new-cases-katwa-remains-epicenter-ebola-outbreak

    Suk, J., A. Zmorzynska, I. Hunger, W. Biederbick, J. Sasse, H. Maidhof, and J. Semenza. “Dual Use Research and
    Technological Diffusion. Reconsidering the Bioterrorism Threat Spectrum.” PLOS Pathogens 7, no. 1 (2011): 1–3.
    doi:10.1371/journal.ppat.1001253.

    Tucker, J., Ed. Innovation, Dual Use and Security. Cambridge, MA: The MIT Press, 2012.
    Vogel, K. “Biodefense.” In Biosecurity Interventions, edited by A. Lakoff and S. Collier, 227–255. New York: Columbia

    University, 2008.

    16 P. F. WALSH

    https://doi.org/10.1089/153871303769201851

    https://doi.org/10.1080/23738871.2017.1298643

    https://doi.org/10.1080/23738871.2017.1298643

    https://doi.org/10.1001/jama.1997.03550050074036

    https://doi.org/10.1016/S0140-6736(15)60858-3

    https://doi.org/10.1016/S0140-6736(15)60858-3

    https://doi.org/10.1080/01495933.2013.821845

    https://globalbiodefense.com/2019/08/05/research-halted-at-usamriid-over-biosafety-issues/CDC,%A0Report%A0on%A0the%A0Potential%A0Exposure%A0to%A0Anthrax

    https://globalbiodefense.com/2019/08/05/research-halted-at-usamriid-over-biosafety-issues/CDC,%A0Report%A0on%A0the%A0Potential%A0Exposure%A0to%A0Anthrax

    https://globalbiodefense.com/2019/08/05/research-halted-at-usamriid-over-biosafety-issues/CDC,%A0Report%A0on%A0the%A0Potential%A0Exposure%A0to%A0Anthrax

    https://doi.org/10.3389/fbioe.2018.00039

    https://theconversation.com/could-gene-editing-tools-such-as-crispr-be-used-as-a-biological-weapon-82187?utm_source=twitter%26utm_medium=twitterbutton

    https://theconversation.com/could-gene-editing-tools-such-as-crispr-be-used-as-a-biological-weapon-82187?utm_source=twitter%26utm_medium=twitterbutton

    http://www.cidrap.umn.edu/news-perspective/2019/02/new-cases-katwa-remains-epicenter-ebola-outbreak

    http://www.cidrap.umn.edu/news-perspective/2019/02/new-cases-katwa-remains-epicenter-ebola-outbreak

    https://doi.org/10.1371/journal.ppat.1001253

    Vogel, K. “Intelligent Assessment: Putting Emerging Biotechnology Threats in Context.” Bulletin of the Atomic Scientists
    35, no. 1 (2013): 45–54.

    Vogel, K. “Necessary Interventions. Expertise and Experiments in Bioweapons Intelligence Assessments.” Science,
    Technology and Innovation Studies 9, no. 2 (2013): 61–88.

    Vogel, K. Phantom Menace or Looming Danger? Baltimore, MD: The Johns Hopkins University Press, 2013.
    Walsh, P. F. Intelligence and Intelligence Analysis. Abingdon, UK: Routledge, 2011.
    Walsh, P. F. “Managing Intelligence and Responding to Emerging Threats: The Case of Biosecurity.” In The Handbook of

    Security, edited by M. Gill, 837–857. Basingstoke: Palgrave Macmillan, 2014.
    Walsh, P. F. “Building Better Intelligence Frameworks Through Effective Governance.” International Journal of Intelligence

    and Counterintelligence 28, no. 1 (2015): 123–142. doi:10.1080/08850607.2014.924816.
    Walsh, P. F. “Managing Emerging Health Security Threats since 9/11: The Role of Intelligence.” International Journal of

    Intelligence and Counterintelligence 29, no. 2 (2016): 341–367. doi:10.1080/08850607.2016.1121048.
    Walsh, P. F. “Teaching Intelligence in the Twenty-first Century: Towards an Evidence-based Approach for Curriculum

    Design.” Intelligence and National Security 32, no. 7 (2017): 1005–1021.
    Walsh, P. F. Intelligence Biosecurity and Bioterrorism. London: Palgrave Macmillan, 2018.
    Walsh, P. F. 2019. “Transforming the Australian Intelligence Community: Mapping Change, Impact and Governance

    Challenges.” Paper given at the 60th International Studies Association Conference, August 27. Toronto, Canada.
    Walsh, P. F., and S. Miller. “Rethinking ‘Five Eyes’ Security Intelligence Collection Policies and Practice Post Snowden.”

    Intelligence and National Security 31, no. 3 (2016): 345–368. doi:10.1080/02684527.2014.998436.
    Walsh, P. F., and J. Ratcliffe. “Strategic Criminal Intelligence Education: A Collaborative Approach.” IALEIA Journal 16

    (2005): 152–166.
    White House. National Biodefense Strategy. Washington DC: White House, 2018.
    WHO. “Ebola Virus Disease in West Africa – the First Nine Months of the Epidemic and Forward Projections.” The New

    England Journal of Medicine 371 (2015): 1481–1495.
    Willingham, D. “A Fresh Threat: Will CAS9 Lead to CRISPR Bioweapons?” Journal of Biosecurity, Biosafety, and Biodefense

    Law 9, no. 1 (2018). doi:10.1515/jbbbl-2018-0010.

    INTELLIGENCE AND NATIONAL SECURITY 17

    https://doi.org/10.1080/08850607.2014.924816

    https://doi.org/10.1080/08850607.2016.1121048

    https://doi.org/10.1080/02684527.2014.998436

    https://doi.org/10.1515/jbbbl-2018-0010

    • Abstract
    • Introduction
      Health security intelligence
      Stolen biological agents
      Dual use research and synthetic biology

      Capability gaps
      Core intelligence processes
      Governance
      Outlook
      Governance
      Closing collection capability gaps
      Analysis
      Notes
      Disclosure statement
      Notes on contributor
      Bibliography

    royalsocietypublishing.org/journal/rstb

    Review
    Cite this article: Polonsky JA et al.

    2

    01

    9

    Outbreak analytics: a developing data science

    for informing the response to emerging

    pathogens. Phil. Trans. R. Soc. B

    3

    7

    4

    :
    201

    8

    027

    6

    .

    http://dx.doi.org/

    10

    .1098/rstb.2018.0276

    Accepted: 4 December 2018

    One contribution of 16 to a theme issue

    ‘Modelling infectious disease outbreaks in

    humans, animals and plants: epidemic

    forecasting and control’.

    Subject Areas:
    health and disease and epidemiology

    Keywords:
    epidemics, infectious, methods, tools,

    pipeline, software

    Author for correspondence:
    Thibaut Jombart

    e-mail: thibautjombart@gmail.com

    †These authors contributed equally to the

    study.

    & 2019 The Authors. Published by the Royal Society under the terms of the Creative Commons Attribution
    License http://creativecommons.org/licenses/by/4.0/, which permits unrestricted use, provided the original
    author and source are credited.

    Outbreak analytics: a developing data
    science for informing the response
    to emerging pathogens

    Jonathan A. Polonsky1,3,†, Amrish Baidjoe4,†, Zhian N. Kamvar4, Anne Cori4,
    Kara Durski2, W. John Edmunds

    5

    ,6, Rosalind M. Eggo5,6, Sebastian Funk5,6,
    Laurent Kaiser3, Patrick Keating5,8, Olivier le Polain de Waroux5,8,9,
    Michael Marks7, Paula Moraga10, Oliver Morgan1, Pierre Nouvellet4,

    11

    ,
    Ruwan Ratnayake5,6, Chrissy H. Roberts7, Jimmy Whitworth5,8

    and Thibaut Jombart4,5,8

    1Department of Health Emergency Information and Risk Assessment, and 2Department of Infectious Hazard
    Management, World Health Organization, Avenue Appia 20, 1211 Geneva, Switzerland
    3Faculty of Medicine, University of Geneva, 1 rue Michel-Servet, 1211 Geneva, Switzerland
    4Department of Infectious Disease Epidemiology, School of Public Health, MRC Centre for Global Infectious Disease
    Analysis, Imperial College London, Medical School Building, St Mary’s Campus, Norfolk Place London W2 1PG, UK
    5Department of Infectious Disease Epidemiology, 6Centre for Mathematical Modelling of Infectious Diseases, and
    7Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and
    Tropical Medicine, Keppel St, London WC1E 7HT, UK
    8UK Public Health Rapid Support Team, London School of Hygiene and Tropical Medicine, Keppel St, London
    WC1E 7HT, UK
    9Public Health England, Wellington House, 133 – 155 Waterloo Road, London SE1 8UG, UK
    10Centre for Health Informatics, Computing and Statistics (CHICAS), Lancaster Medical School,
    Lancaster University, Lancaster LA1 4YW, UK
    11School of Life Sciences, University of Sussex, Sussex House, Brighton BN1 9RH, UK

    JAP, 0000-0002-8634-4255; AB, 0000-0001-5295-5085; ZNK, 0000-0003-1458-7108;
    AC, 0000-0002-8443-9162; SF, 0000-0002-2842-3406; OM, 0000-0002-9543-3778;
    PN, 0000-0002-6094-5722; TJ, 0000-0003-2226-8692

    Despite continued efforts to improve health systems worldwide, emerging

    pathogen epidemics remain a major public health concern. Effective response

    to such outbreaks relies on timely intervention, ideally informed by all available

    sources of data. The collection, visualization and analysis of outbreak data are

    becoming increasingly complex, owing to the diversity in types of data, questions

    and available methods to address them. Recent advances have led to the rise of

    outbreak analytics, an emerging data science focused on the technological and
    methodological aspects of the outbreak data pipeline, from collection to analysis,

    modelling and reporting to inform outbreak response. In this article, we assess

    the current state of the field. After laying out the context of outbreak response,

    we critically review the most common analytics components, their inter-

    dependencies, data requirements and the type of information they can provide

    to inform operations in real time. We discuss some challenges and opportunities

    and conclude on the potential role of outbreak analytics for improving our

    understanding of, and response to outbreaks of emerging pathogens.

    This article is part of the theme issue ‘Modelling infectious disease outbreaks

    in humans, animals and plants: epidemic forecasting and control‘. This theme

    issue is linked with the earlier issue ‘Modelling infectious disease outbreaks in

    humans, animals and plants: approaches and important themes’.

    1. Introduction
    Emerging infectious diseases are a constant threat to public health worldwide.

    In the past decade, several major outbreaks, such as the 2009 influenza pandemic [1],

    http://crossmark.crossref.org/dialog/?doi=10.1098/rstb.2018.0276&domain=pdf&date_stamp=2019-05-20

    http://dx.doi.org/10.1098/rstb/374/1776

    http://dx.doi.org/10.1098/rstb/374/1776

    http://dx.doi.org/10.1098/rstb/374/1776

    mailto:thibautjombart@gmail.com

    http://orcid.org/

    http://orcid.org/0000-0002-8634-4255

    http://orcid.org/0000-0001-5295-5085

    http://orcid.org/0000-0003-1458-7108

    http://orcid.org/0000-0002-8443-9162

    http://orcid.org/0000-0002-2842-3406

    http://orcid.org/0000-0002-9543-3778

    http://orcid.org/0000-0002-6094-5722

    http://orcid.org/0000-0003-2226-8692

    http://creativecommons.org/licenses/by/4.0/

    http://creativecommons.org/licenses/by/4.0/

    http://creativecommons.org/licenses/by/4.0/

    Figure 1. Successive phases of an outbreak response. The histogram along the top represents reported ( yellow) and unreported ( grey) incidence.

    royalsocietypublishing.org/journal/rstb
    Phil.Trans.R.Soc.B

    374:20180276

    2

    the Middle-East Respiratory Syndrome coronavirus (MERS-

    CoV) [2 – 4], the emergence of Zika [5,6] and the West African

    Ebola virus disease (EVD) outbreak [7,8], have been potent

    reminders of the need for robust surveillance systems and

    timely responses to nascent epidemics [9]. The West African

    EVD outbreak, by far the largest such epidemic in recorded his-

    tory, in particular, had a strong impact on global health security

    and public health policy and practice [7,8,10]. It highlighted the

    difficulties of maintaining situational awareness in the absence

    of standards for surveillance, data collection and analysis, as

    well as the challenges of mounting and sustaining a large-scale

    international response [7,8,11,12]. Despite the lessons learnt

    [9,13,14], the recent (2018) EVD outbreaks in Democratic Repub-

    lic of the Congo [15,16] are a stark reminder that a large number

    of these challenges remain.

    An important feature of the modern response to epidemics

    is the increasing focus on exploiting all available data to inform

    the response in real time and allow evidence-based decision

    making [3,4,7,8,13,17]. Using data for improving situational

    awareness is complex, involving a range of inter-connected

    tasks and skills from point-of-care data collection to the gener-

    ation of informative situational reports (sitreps). The science

    underpinning these data pipelines involves a wide range of

    approaches, including database design and mobile technology

    [18,19], frequentist statistics and maximum-likelihood esti-

    mation [7], interactive data visualization [20,21], geostatistics

    [22 – 24], graph theory [20,25,26], Bayesian statistics [8,27,28],

    mathematical modelling [29 – 31], genetic analyses [32 – 36]

    and evidence synthesis approaches [37]. This accretion of

    heterogeneous disciplines, which may be best summarized as

    ‘outbreak analytics’, forms an emerging domain of data

    science: an ‘interdisciplinary field that uses scientific methods,

    processes, algorithms and systems to extract knowledge and

    insights from data in various forms’ [38], dedicated to inform-

    ing outbreak response. Outbreak analytics sits at the crossroads
    of public health planning, field epidemiology, methodological

    development and information technologies, opening up excit-

    ing opportunities for specialists in these fields to work together

    to meet the needs for an epidemic response.

    In this article, we outline this developing research field and

    review the current state of outbreak analytics. In particular, we

    focus on how different analysis components interact within

    functional workflows, and how each component can be used

    to inform different stages of an outbreak response. We discuss

    key challenges and opportunities associated with the deploy-

    ment of efficient, reliable and informative data analysis

    pipelines and their potential impact.

    2. The outbreak response context
    (a) The different phases of an outbreak response
    The focus of the public health response shifts during the

    course of an epidemic or outbreak, and so do the analytics.

    We identify four main stages (figure 1). The detection stage
    starts with the first case and ends with the first intervention

    activities (e.g. patient isolation, contact tracing, vaccination)

    and involves surveillance systems and mostly qualitative

    risk assessments. Next, the early response is the initial part
    of the intervention during which the first simple analytics

    can take place, essentially centred around estimating trans-

    missibility. This blends into the intervention stage, where
    more complex analytics may be involved to inform plann-

    ing (e.g. vaccination strategies), which ends once the last

    reported case has recovered or died. The post-intervention
    stage is for lessons to be learned, for improving prepared-

    ness for the next epidemic and for training and capacity

    building [39].

    (b) Questions during and after the intervention
    During the early response, efforts are dedicated to estimating

    the likely impact of the outbreak and anticipating the nature,

    scale and timing of resources needed [7,13,15]. Theoretically,

    different factors including not only the total number of cases

    and fatalities but also the morbidity and overall impact on qual-

    ity of life, as well as societal and economic impact, should

    ideally be taken into account when attempting to predict

    disease burden [40 – 43]. Generally, as the demographic and

    morbidity data needed by composite measures of health-

    adjusted life years [40] are lacking in outbreak response

    contexts, efforts tend to focus on other proxies of impact: asses-

    sing transmissibility, predicting future case incidence and

    associated mortality and investigating risk factors [1,3,7,15].

    Analytical needs to diversify as the intervention progresses.

    While investigations of transmissibility, mortality and risk

    factors remain key throughout [8], new questions may arise to

    inform the implementation of control and mitigation measures.

    These may focus on predicting the impact of potential measures

    including testing (e.g. ‘Could a rapid test help reduce inci-

    dence?’ [29]), vaccine development (e.g. ‘Could a candidate

    vaccine be evaluated in this outbreak?’ [44,45]), vaccination

    campaigns (e.g. ‘Which is the optimal vaccination strategy?’

    [46,47]) or travel restrictions (e.g. ‘Should international travel

    be restricted?’ [48]), or on estimating the impact of current

    measures such as improvements in access to care (e.g. ‘Has the

    royalsocietypublishing.org/journal/rstb
    Phil.Trans.R.Soc.B
    374:20180276
    3
    delay between symptom onset and hospitalization been
    reduced?’ [14,15]). As case incidence reduces, statistical model-

    ling can also be useful for assessing or predicting the end of an

    outbreak [49 – 51]. At the field operational level, outbreak

    response analytics may be best focused on informing and moni-

    toring core surveillance activities and performance indicators,

    such as contact tracing [11], through the use of tools for contact

    data visualization [52], mapping [53,54] and on analysis pipe-

    lines integrating mobile data collection tools [18,19,55,56]

    with automated reporting systems [57 – 59]. Finally, the post-

    intervention phase lends itself to retrospective studies, which

    can assess further the impact of interventions [60], tease apart

    finer processes driving the epidemic dynamics such as contact

    patterns [12,61], study risk factors [54,62], identify avenues for

    fortifying surveillance [13,36,63] and evaluate, improve and

    develop modelling techniques [28,64,65].

    (c) What are outbreak data?
    The term ‘outbreak data’ encompasses different types of
    information, of which we first distinguish ‘case data’ from ‘back-
    ground data’. Case data include the description of reported cases
    gathered in linelists, i.e. flat files where each row is a case and
    each column a recorded variable (e.g. dates of onset and admis-

    sion, gender, age, location), thereby fulfilling the definition of

    ‘tidy data’ in the data science community [66]. Case data also

    include exposure and contact tracing data, either stored within

    a linelist or in separate files, pathogen whole genome sequencing

    (WGS) and data pertaining to outbreak investigations (e.g. case –

    control and cohort study data). Background data document the
    underlying characteristics of the affected populations. This

    includes demographic information (e.g. maps of population den-

    sities, age stratification, mixing patterns), movement data (e.g.

    borders, traveller flows, migration), health infrastructure

    (e.g. healthcare facilities, drug stockpiles) and epidemiological

    data themselves (e.g. levels of pre-existing immunity). A final

    type of data we consider here is ‘intervention data’, which refers
    to information on decisions made and efforts deployed as part

    of the intervention, such as vaccination coverage, the extent of

    active case finding or potential changes in the epidemiologi-

    cal case definition. An in-depth discussion of data needs in

    outbreaks can be found in Cori et al. [13].

    3. Outbreak analytics
    (a) An overview of the outbreak analytics toolbox
    We use the term ‘outbreak analytics’ to refer to the variety of
    tools and methods used to collect, curate, visualize, analyse,

    model and report on outbreak data. These tools and their

    inter-dependencies are summarized in an exemplary workflow

    represented in figure 2, derived from analyses pipelines used

    during recent epidemics of pandemic influenza [1], MERS-

    CoV [4] and EVD [7,8,17]. Note that workflows may vary

    substantially in other epidemic contexts. For instance, analyses

    of food-borne outbreaks may focus on traceback data [67 – 69],

    while vector-borne disease analysis may focus heavily on

    modelling the vector’s ecological niche [70,71].

    (b) Tools for the collection of epidemiological data
    Tools for data capture have become a focus of much discussion

    in recent years as those involved in outbreak response seek

    to make use of important technological advances including

    mobile data collection, cloud computing and built-in automated

    data analyses and reporting. In resource-limited settings, in par-

    ticular, epidemiological data are still often collected with pen

    and paper, the advantages of which are familiarity, simplicity,

    low cost and reliability where access to Internet and power

    sources may be limited. However, there are some downsides

    to using paper as a data management tool, becoming increas-

    ingly important with larger outbreaks, as any system for

    the printing and distribution, collection and storage and digitiz-

    ation of forms becomes overwhelmed. Additionally, two-stage

    processes involving transcription of data from forms typically

    introduces additional data entry errors [72 – 75] and substantial

    delays from data capture to analysis [72].

    Electronic data collection (EDC) is becoming increasingly

    popular [18,19,55,56]. These tools make use of widely avail-

    able, low-cost hardware (e.g. smartphones and tablets) [76]

    that can, when appropriately configured, consume little

    power and collect data offline, making them suitable for use

    in resource-poor settings. Some of those may be part of existing

    surveillance systems or be deployed instead for specific

    enhanced surveillance and response activities during an out-

    break. EDC platforms can also enhance data quality through

    the use of restriction rules and logical checks, and enforce

    reporting (even when there are zero cases) and entry of essen-

    tial variables [72,76]. EDC can decrease the delay between data

    collection, centralization and analysis, which is critical for

    data-driven responses. Time can be saved through ‘form

    logic’ (e.g. automatically skipping sections of a survey not

    relevant to a participant), while real-time, automated centrali-

    zation, data analysis and reporting can be directly built into

    the platform. In addition, mobile-based EDC enables the collec-

    tion of other types of data including GPS coordinates,

    photographs, barcode (useful to link case data and clinical

    specimens) and even aiding diagnostics by directly interfacing

    with point-of-care diagnostic devices [77 – 79].

    Maintaining confidentiality and privacy is a legitimate con-

    cern whenever data concerning human subjects are collected.

    While EDC systems provide opportunities for unauthorized

    interception and access to such information, many systems

    support end-to-end encryption during data transfer [80],

    although few provide additional security through encryption

    at the level of data entry.

    (c) Descriptive analyses
    The first, and arguably one of the most important steps in

    data analysis is exploration, where visualization plays a

    central role, completed with informative summary statistics

    [81,82]. The first type of graphics needed for rapid assessment

    of ongoing dynamics is the epidemic curve (epicurve), which

    shows case incidence time series as a histogram of new onset

    dates for a given time interval [83 – 85]. Cumulative case

    counts, sometimes used in the absence of a raw linelist, are

    best avoided in epicurves, as they tend to obscure ongoing

    dynamics and create statistical dependencies in data points

    that will result in biases and lead to under-estimating

    uncertainty in downstream modelling [86].

    Maps have been at the core of infectious disease epide-

    miology from a very early stage [87]. Nowadays, they are

    typically used to visualize the distribution of disease [88], for

    representing the ‘ecological niche’ of infectious diseases at

    large scales [23,24,89] and for assessing the spatial dynamics

    of an outbreak and strategizing interventions [7,8]. Providers

    of free and crowd-sourced [90] geographical data like the

    Figure 2. Example of outbreak analytics workflow. This schematic represents eight general analyses that can be performed from outbreak data. Outputs containing
    actionable information for the operations are represented as hexagons. Data needed for each analysis are represented as a different colour in the center, using plain
    and light shading for mandatory and optional data, respectively. (Online version in colour.)

    royalsocietypublishing.org/journal/rstb
    Phil.Trans.R.Soc.B
    374:20180276
    4

    Humanitarian Open Street Maps Team (Humanitarian

    OpenStreetMap Team Home; see https://www.hotosm.org/

    (accessed 26 September 2018)), the Missing Maps project (Mis-

    singMaps; see https://www.missingmaps.org/ (accessed 26

    September 2018)), healthsites.io (see https://healthsites.io/

    (accessed 26 September 2018)) and the Radiant Earth Foun-

    dation (Radiant Earth Foundation – Earth imagery for

    impact; see https://www.radiant.earth (accessed 18 November

    2018)) provide layers of spatial data that include information on

    the location of households and health facilities, among other

    determinants. Several tools including SaTScan and ClusterSeer

    are routinely applied to surveillance system data for automated

    outbreak detection and the evaluation of clustering of disease

    by time and space [91]. Other examples of freely available map-

    ping tools that can help track the spread of infectious diseases

    include the Spatial Epidemiology of Viral Haemorrhagic

    Fevers (VHF) disease visualization (see http://www.health-

    data.org/datavisualization/spatial-epidemiology-viralhemor-

    rhagic-fevers; accessed 19 September 2018), which maps risks of

    emergence and spread of VHF diseases, Nextstrain [92] and

    Microreact [93], which focus on mapping pathogen evolution

    and epidemic spread, and HealthMap [94], which provides

    resources for the rapid detection of outbreaks. Geographical

    locations of reported cases can also be useful for informing

    more complex modelling approaches [95].

    https://www.hotosm.org/

    https://www.missingmaps.org/

    https://healthsites.io/

    https://www.radiant.earth

    http://www.healthdata.org/datavisualization/spatial-epidemiology-viralhemorrhagic-fevers

    http://www.healthdata.org/datavisualization/spatial-epidemiology-viralhemorrhagic-fevers

    http://www.healthdata.org/datavisualization/spatial-epidemiology-viralhemorrhagic-fevers

    royalsocietypublishing.org/journal/rstb
    Phil.Trans.R.Soc.B
    374:20180276
    5
    In epidemics driven by person-to-person transmission, a
    last essential source of data is contact data [20], which includes

    data on case exposure [12] as well as contact tracing, where
    appropriate [11,63]. Exposure data document transmission

    pairs, which can yield precious insights into ‘paired delays’

    (figure 2) including the serial interval (time between onsets

    of a case and their infector) or the generation time (time

    between the dates of infections of a case and their infector)

    [7,8], which are in turn useful for estimating transmissibility

    [27,28,96,97]. Exposure data can also be used to investigate

    the occurrence and determinants of super-spreading events

    [12] and help identify introduction events in the case of zoono-

    tic diseases [98]. Contact tracing, through the early detection of

    new cases and their subsequent isolation and treatment, plays a

    central role in reducing onward transmission and therefore

    containing outbreaks [11,63,99], while additionally providing

    potential information on risk factors [7,11].

    Summary statistics are a useful complement to data visual-

    ization in the exploratory phase of data analysis. Some metrics,

    such as transmissibility, require the use of statistical or math-

    ematical models in order to be estimated (see §3d below) and

    are therefore not readily available as descriptive tools. Other

    useful statistics can be readily computed from linelists, includ-

    ing different demographic indicators of the reported cases

    (e.g. gender, age, occupation [7,100,101]), case fatality ratios

    (the proportion of cases who died of the infection) or case

    delays such as the times to hospitalization, recovery or death,

    reported as a whole [1,7,8] or stratified by groups [100,101].

    The incubation period (time from infection to symptom onset)

    is another important delay for informing the intervention (e.g.

    to define the duration of contact tracing or declare the end of

    an outbreak), but can be harder to derive as it requires data on

    case exposure as well. Note that in the case of delays, these are

    best analysed by characterising the full distribution (e.g. by fit-

    ting to an appropriate probability distribution such as

    discretized Gamma [7]) rather than reported as a single central

    value [7,8,102,103].

    (d) Quantifying transmissibility
    The ‘transmissibility’ of a disease is here used to refer to the

    rate at which new cases arise in the population, resulting

    either in epidemic growth or decline [1,3,27,28]. Rather than

    an intrinsic property of a specific disease, transmissibility

    thus defined quantifies the propagation of a pathogen in a

    given epidemic setting and is impacted by multiple factors

    including population demographics, mixing and levels pre-

    existing immunity. Importantly, estimates of transmissibility

    reported in the literature will typically be biased towards

    higher values, as subcritical outbreaks are by definition less

    likely to be detected. Several metrics of transmissibility can

    be used depending on the type of data available and can be

    estimated using different approaches.

    A first measure of transmissibility is the growth rate (r),
    which is estimated from a simple model where case incidence

    is either exponentially growing (r . 0) or declining (r , 0).
    Typically, r is estimated directly from epicurves (figure 2)
    using a log-linear model, where r is defined as the slope of a
    linear regression on log-transformed incidence [104,105].

    Besides its simplicity and its computational efficiency, this

    approach has the benefits of being embedded in the linear

    modelling framework, thereby allowing one to measure the

    uncertainty associated with a given estimate of r, to test for

    differences in growth rates, e.g. between different locations,

    and to derive short-term incidence predictions. Moreover, the

    growth rate can also be used to estimate the doubling and halv-

    ing times of the epidemic, i.e. the time during which incidence

    doubles (respectively is halved), as alternative metrics of trans-

    missibility [103]. Unfortunately, the log-linear model can only

    fit exponentially growing or decaying outbreaks, which may

    not always be appropriate in the presence of complex spatial

    or age structure, or owing to changes in reporting, transmissi-

    bility or proportion of susceptible individuals over time.

    Besides, it cannot readily accommodate time periods with no

    cases, so that its applicability may in practice be restricted.

    While r quantifies the speed at which a disease spreads, it does
    not contain information on the level of the intervention that is
    necessary to control a disease [106]. This is better characterized

    by the reproduction number (here generically noted ‘R’), which
    measures the average number of secondary cases caused by

    each primary case. Researchers typically distinguish the basic

    reproduction number (R0 [104]), which applies in a large, fully
    susceptible population, without any control measures, from

    the effective reproduction number (Reff ), which is the number
    of secondary cases after accounting for behavioural changes,

    interventions and declines in susceptibility [96]. The current

    reproduction number determines the dynamics of the epidemic

    in the near future, with values greater than 1 predicting an

    increase in cases, and values less than 1 predicting control

    [104]. The value of R can also be used to calculate the fraction
    of the population that needs to be immunized (typically through

    vaccination) in order to contain an outbreak [104].

    Different methodological approaches have been developed

    to estimate the reproduction number. R can be approximated
    using estimates of the growth rate r combined with knowledge
    of the generation time distribution [97]. R can also be derived
    from compartmental models [104,107]. The formula will

    depend on the type of model used, but such estimation

    will usually require that different rates (e.g. rates of infection,

    recovery, death) are either known or estimated by fitting the

    model to data [104,107]. Real-world complexities can be incor-

    porated into this approach; however, fitting such models can be

    challenging and may require computationally intensive algor-

    ithms such as data augmentation, approximate bayesian

    computation, or particle filters [108]. Compartmental models

    also require assumptions about the total population size and

    the proportion of the population at risk, which may be difficult

    to estimate in an outbreak. As an alternative, branching process

    models can be used to estimate R directly from incidence data
    [27,28,96,109]. This requires a pre-specified distribution of the

    generation time, or of the serial interval, although recent devel-

    opments suggest that in some cases, the generation time

    distribution itself can also be simultaneously estimated [4].

    Branching process models are usually much simpler to fit to

    data than their compartmental counterparts, which facilitates

    their use in real time [27].

    Beyond the mere estimation of transmissibility, it is often

    essential to forecast future incidence for advocacy and plan-

    ning purposes, e.g. to compare different interventions and

    epidemic scenarios [7,8,15,30]. A variety of mathematical and

    statistical models, including those reviewed here for estimating

    transmissibility, can also be used for short-term incidence fore-

    casting [65]. Despite the growing body of research focusing on

    predicting incidence during epidemics [65,110], there are cur-

    rently no gold standards and the relative performances of

    forecasting methods largely remain to be assessed. Methods

    royalsocietypublishing.org/journal/rstb
    Phil.Trans.R.Soc.

    6
    that have been developed and applied in other fields to rigor-
    ously assess not just the accuracy of forecasts but also how

    well models quantify the inherent uncertainty in making

    predictions, are only rarely applied in infectious disease epide-

    miology [111,112]. Whether it is to estimate R or predict future
    incidence, the most appropriate method ultimately depends on

    the particular epidemiological setting, existing knowledge of

    the transmission dynamics and data availability. Branching

    process models, for example, can be used for a quick estimate

    of the current value of R from the recent trend in case numbers
    and, by extrapolating this forward, of expected case numbers in

    the near future [27,28,96]. Mechanistic or simulation models,

    on the other hand, aim to include a more explicit representation

    of the different factors that might influence transmission. They

    can be a more natural choice for assessing the expected impact

    of possible interventions, but they usually require careful para-

    metrization and often intensive computation [29,30,45,113],

    both of which can be challenging early in an outbreak when

    data are scarce and rapid turnaround crucial.

    B
    374:20180276

    (e) Analytical epidemiological techniques
    Analytical epidemiological studies use data to better describe

    outbreaks and populations at risk and inform real-time and

    subsequent response efforts. Typically, these are conducted

    during the intervention and post-intervention phases of an out-

    break response (figure 1). They include observational designs

    such as retrospective cohort and case – control studies to ident-

    ify risk factors and quantify associations between potential

    causes and their outcomes (typically, the disease in question),

    and experimental designs, such as randomized-control studies

    used to estimate the impact of interventions such as vaccination

    and treatments [114]. These studies reside outside of the

    normal scope of outbreak response activities, being inserted

    ad hoc as functions that are not necessarily routine response
    activities such as strengthening surveillance. In the case of

    observational epidemiological studies, data on exposures and

    outcomes are required, permitting estimations of the increased

    risk of disease among people exposed to risk factors of interest

    [54,62,115,116]. In the case of experimental epidemiology, data

    on outcomes of interest are collected to permit estimations of

    heterogeneity among groups (e.g. in the presence/absence of

    intervention).

    The usefulness of such studies in informing outbreak

    response is highly context-dependent. Observational studies

    may be undertaken early on in the intervention phase to

    help identify ongoing infection sources of environmental,

    food-borne or water-borne nature [117] and to stop the out-

    break at its source. In longer-running outbreaks, they can

    provide insights into opportunities for control [53,115,118]

    and inform global policy decisions that relate to outbreak

    response [119]. However, the time and expertise needed to

    prepare and implement these studies may preclude their

    application in the midst of an ongoing outbreak, so that the

    cost and benefits of such an undertaking need to be carefully

    weighed in emergency settings.

    ( f ) Genetic analyses
    Whole genome sequencing of pathogens is increasingly afford-

    able and reliable, and therefore more frequent in outbreak

    investigations [1,120 – 126]. As technology is making real-time

    sequencing in the field a developing standard in the coming

    years [127,128], genetic analysis will likely carve out its own

    space in the outbreak analytics toolkit.

    Different methods can be used to extract information from

    pathogen WGS. In bacterial genomics, molecular epidemiol-

    ogy methods have been used extensively for defining strains

    of related isolates [32,129], which can be used to infer various

    features of the pathogens sampled such as their origins, antimi-

    crobial resistance profiles, virulence or antigenic characteristics

    [130 – 132]. These methods usually exploit only a fraction of the

    information contained within pathogens’ genomes, as they rely

    on genetic variation in a limited number of housekeeping

    genes [32,129]. While these methods will likely remain useful

    in years to come, substantially more information can be

    extracted by using WGS to reconstruct phylogenetic trees,

    which represent the evolutionary history of the sampled iso-

    lates, assuming the absence of selection or horizontal gene

    transfers [133]. Different types of phylogenetic reconstruction

    methods can be used, including fast, scalable distance-based

    methods [134] or more computer-intensive approaches using

    a maximum-likelihood [135,136] or the Bayesian framework

    [33,137]. Phylogenies can be used to assess the origins of a

    set of pathogens [138], patterns of geographical spread [125],

    host species jumps [139,140], past fluctuations in the pathogen

    population sizes [141] and even, in some cases, the reproduc-

    tion number [1]. Importantly, there is a growing tendency to

    analyse phylogenetic trees in the broader context of other epi-

    demiological data (mainly geographical locations until now),

    which is facilitated by user-friendly Web applications [92,93].

    A further step towards integrating WGS alongside epide-

    miological data is the reconstruction of transmission trees

    (who infects whom) using evidence synthesis approaches.

    This methodological field has been growing fast over the past

    decade [25,142 – 148], but most applications of these methods

    remain within academia and their usefulness in the field in

    an outbreak response context needs to be critically assessed.

    A potential benefit of accurately reconstructing transmission

    trees lies in the identification of multiple introductions, the

    quantification of the proportion of unreported cases and the

    detection of heterogeneities in individual transmissibility

    [145]. Unfortunately, the reconstruction of transmission trees

    is a difficult and computationally intensive problem. First,

    most diseases do not accumulate sufficient genetic diversity

    during the course of an outbreak to allow the accurate recon-

    struction of transmission chains, so that multiple data sources

    need to be used [35], making these methods more data-

    demanding than most other approaches in outbreak analytics

    (figure 2). In addition, the complex nature of the problem

    requires the use of Bayesian methods for model fitting,

    making these approaches difficult to interpret by non-experts

    [145,146,148].

    4. Discussion
    In this article, we reviewed methodological and technological

    resources forming the basis of outbreak analytics, an emerg-

    ing data science for informing outbreak response. Outbreak

    analytics is embedded within a broader public health infor-

    mation context that starts with disease surveillance systems,

    followed by risk assessment and management, the epidemiolo-

    gical response itself, and finishes with the production of

    actionable information for decision making. Part of the chal-

    lenge that this new field will face in the coming years

    royalsocietypublishing.org/journal/rstb
    Phil.Trans.R.Soc.B
    374:20180276
    7
    pertains to the seamless integration of data analytics pipelines
    within existing workflows. As responders can allocate only

    limited time to data analysis, analytics resources should

    produce simple, interpretable results, highlighting the most

    pressing issues that need addressing and monitoring all

    relevant indicators to inform the response.

    Outbreak analytics and resulting outputs are central to the

    surveillance pillar of any outbreak response, yet resources and

    capacities to ensure data availability and quality are often lim-

    ited owing to operational constraints [16]. Priorities in terms of

    data needs should be defined by what actionable information it

    may give access to through the available analytics pipelines

    [13]. In this respect, we foresee that typical linelist data such

    as dates of events (e.g. onset, reporting, hospitalization, dis-

    charge), age, gender, disease outcome, geographical locations

    and exposure data will fulfil most needs, while other data

    such as WGS may only be useful for specific diseases and con-

    texts [34,35]. Intervention data are rarely collected but should

    be given more consideration, as they are key to assessing the

    impact and effectiveness of control measures, both during

    and after the operations. Similarly, data on the fraction of

    cases reported (and its variations through time), as well as be-

    havioural changes (e.g. care-seeking behaviour) in the affected

    populations, can be very important sources of information for

    modelling [149].

    Fortunately, what we called ‘background data’ in this

    article can be gathered and shared outside of the epidemic con-

    text. Besides maps, population census, sero-surveys or genetic

    databanks, data on the natural histories of diseases derived

    from past epidemics, such as key delay distributions and trans-

    missibility, can form a useful substitute to real-time estimates,

    especially in the early stages of outbreaks when such data may

    be lacking. While crowd-sourced initiatives are promising and

    have been used successfully in low resource settings [90], more

    efforts are needed to collate and curate open data sources,

    assess their quality and make them widely available to the

    community. We argue that international public health agencies

    and non-governmental agencies should play a central role in

    orchestrating such background data preparedness.

    Outbreak analytics is a developing field, and as such, there

    remain many gaps in terms of data collection, analysis and

    reporting tools. Some methodological challenges persist, such

    as better characterising forecasting methods [28,64,65], includ-

    ing spatial information and population flows into existing

    transmission models [95], and improving the integration of

    different types of data for reconstructing transmission trees

    [35]. In order to ensure transparent methods and availability

    to analysts in any setting, the implementation must be as

    freely available, open-source software. Among other popular

    programming languages, such as Python, Java, or Julia, the R

    software [150] arguably offers the largest collection of free

    tools for data analysis and reporting, and an increasing

    number of packages for infectious disease epidemiology

    [20,21,27,84,145] may form a solid starting point for the devel-

    opment of a comprehensive, robust and transparent toolkit for

    the analysis of epidemic data [151]. Importantly, the use of a

    common platform for the development and use of outbreak

    analytics tools will also likely contribute to standardizing

    data practices, including collection, sharing and analysis.

    A final point relates to the use and dissemination of these

    new resources: how can outbreak analytics best help improve

    public health? As noted by Bausch & Clougherty [39], health
    science should not be an entity unto itself, but a means to an end.
    Insofar as it can help field epidemiologists collect, visualize

    and analyse data, and subsequently provide decision-makers

    with actionable information, outbreak analytics will likely

    occupy an increasing space in field epidemiology over the

    years to come. We foresee that the dissemination of free train-

    ing resources [152], the modernization of field epidemiology

    training programmes and the deployment of applied data

    scientists to the field with a sustained capacity building in

    resource-poor and vulnerable countries will be instrumental

    in shaping the future of this emerging field of health science.

    Data accessibility. This article has no additional data.
    Authors’ contributions. T.J. drafted the outline of the review and revised
    the manuscript. J.A.P., A.B., T.J. wrote the first draft of the manu-
    script. Z.N.K. produced the figures. A.C., W.J.E., R.M.E., S.F., L.K.,
    P.K., M.M., P.M., P.N., O.P.W., R.R., J.W. contributed the content.

    Competing interests. We declare we have no competing interests.
    Funding. This paper was supported with funding from the Global Chal-
    lenges Research Fund (GCRF) for the project ‘RECAP—research
    capacity building and knowledge generation to support preparedness
    and response to humanitarian crises and epidemics’ managed through
    RCUK and ESRC (ES/P010873/1). P.K., O.L.P., J.W. and T.J. receive
    support from the UK Public Health Rapid Support Team, which is
    funded by the United Kingdom Department of Health and Social
    Care. We acknowledge the National Institute for Health Research—
    Health Protection Research Unit for Modelling Methodology (T.J.)
    for funding. M.M., C.H.R., receive funding through the National Insti-
    tute for Health Research (PR-OD-1017-20001). R.M.E. acknowledges
    funding from an HDR UK Innovation Fellowship (grant no. MR/
    S003975/1). A.C. thanks the Medical Research Council for funding.
    S.F. was supported by the Wellcome Trust (210758/Z/18/Z). The
    authors alone are responsible for the views expressed in this article
    and they do not necessarily represent the views, decisions or policies
    of the institutions with which they are affiliated.

    Acknowledgements. We would like to thank Annick Lenglet and Isidro
    Carrion-Martin, Epidemiologists at Medecins Sans Frontieres (MSF,
    Operational Centre Amsterdam) for their additional reflections. The
    views expressed in this publication are those of the authors and
    not necessarily those of the National Health Service, the National
    Institute for Health Research or the Department of Health and
    Social Care. The authors alone are responsible for the views
    expressed in this article and they do not necessarily represent the
    views, decisions or policies of the institutions with which they are
    affiliated.

    References

    1. Fraser C et al. 2009 Pandemic potential of a strain
    of influenza A (H1N1): early findings. Science 324,
    1557 – 1561. (doi:10.1126/science.1176062)

    2. Assiri A et al. 2013 Hospital outbreak of Middle
    East respiratory syndrome coronavirus.
    N. Engl. J. Med. 369, 407 – 416. (doi:10.1056/
    NEJMoa1306742)

    3. Cauchemez S, Fraser C, Van Kerkhove MD, Donnelly
    CA, Riley S, Rambaut A, Enouf V, van der Werf S,
    Ferguson NM. 2014 Middle East respiratory
    syndrome coronavirus: quantification of the extent
    of the epidemic, surveillance biases, and
    transmissibility. Lancet Infect. Dis. 14, 50 – 56.
    (doi:10.1016/S1473-3099(13)70304-9)

    4. Cauchemez S et al. 2016 Unraveling the drivers
    of MERS-CoV transmission. Proc. Natl Acad.
    Sci. USA 113, 9081 – 9086. (doi:10.1073/pnas.
    1519235113)

    5. Campos GS, Bandeira AC, Sardi SI. 2015 Zika virus
    outbreak, Bahia, Brazil. Emerg. Infect. Dis. 21,
    1885 – 1886. (doi:10.3201/eid2110.150847)

    http://dx.doi.org/10.1126/science.1176062

    http://dx.doi.org/10.1056/NEJMoa1306742

    http://dx.doi.org/10.1056/NEJMoa1306742

    http://dx.doi.org/10.1016/S1473-3099(13)70304-9

    http://dx.doi.org/10.1073/pnas.1519235113

    http://dx.doi.org/10.1073/pnas.1519235113

    http://dx.doi.org/10.3201/eid2110.150847

    royalsocietypublishing.org/journal/rstb
    Phil.Trans.R.Soc.B
    374:20180276
    8
    6. European Centre for Disease Prevention and Control.
    2015 Zika virus epidemic in the Americas: potential
    association with microcephaly and Guillain-Barré
    syndrome ( first update), 21 January 2016. (See
    https://ecdc.europa.eu/sites/portal/files/media/en/
    publications/Publications/zika-virus-americas-
    association-with-microcephaly-rapid-risk-
    assessment ).

    7. WHO Ebola Response Team. 2014 Ebola virus
    disease in West Africa – the first 9 months
    of the epidemic and forward projections.
    N. Engl. J. Med. 371, 1481 – 1495. (doi:10.1056/
    NEJMoa1411100)

    8. WHO Ebola Response Team et al. 2015 West African
    Ebola epidemic after one year – slowing but not
    yet under control. N. Engl. J. Med. 372, 584 – 587.
    (doi:10.1056/NEJMc1414992)

    9. Moon S et al. 2015 Will Ebola change the game?
    Ten essential reforms before the next pandemic. The
    report of the Harvard-LSHTM independent panel on
    the global response to Ebola. Lancet 386,
    2204 – 2221. (doi:10.1016/S0140-6736(15)00946-0)

    10. Van Kerkhove MD, Bento AI, Mills HL, Ferguson NM,
    Donnelly CA. 2015 A review of epidemiological
    parameters from Ebola outbreaks to inform early
    public health decision-making. Sci Data 2, 150019.
    (doi:10.1038/sdata.2015.19)

    11. Senga M et al. 2017 Contact tracing performance
    during the Ebola virus disease outbreak in Kenema
    district, Sierra Leone. Phil. Trans. R. Soc. B 372,
    20160300. (doi:10.1098/rstb.2016.0300)

    12. International Ebola Response Team. 2016 Exposure
    patterns driving Ebola transmission in West Africa: a
    retrospective observational study. PLoS Med. 13,
    e1002170. (doi:10.1371/journal.pmed.1002170)

    13. Cori A et al. 2017 Key data for outbreak evaluation:
    building on the Ebola experience. Phil. Trans. R. Soc. B
    372, 20160371. (doi:10.1098/rstb.2016.0371)

    14. Lewnard JA. 2018 Ebola virus disease: 11 323
    deaths later, how far have we come? Lancet 392,
    189 – 190. (doi:10.1016/S0140-6736(18)31443-0)

    15. Ebola Outbreak Epidemiology Team. 2018 Outbreak
    of Ebola virus disease in the Democratic Republic of
    the Congo, April – May, 2018: an epidemiological
    study. Lancet 392, 213 – 221. (doi:10.1016/S0140-
    6736(18)31387-4)

    16. Polonsky J et al. 2019 Lessons learnt from Ebola
    virus disease surveillance in Équateur Province,
    May – July 2018. Weekly Epidemiological Record 94,
    23 – 27.

    17. 2017 WHO j Ebola outbreak Democratic Republic of
    the Congo 2017. See https://www.who.int/
    emergencies/ebola-DRC-2017/en/.

    18. Hartung C, Lerer A, Anokwa Y, Tseng C, Brunette W,
    Borriello G. 2010 Open Data Kit: Tools to build
    information services for developing regions. In
    Proceedings of the 4th ACM/IEEE International
    Conference on Information and Communication
    Technologies and Development, pp. 18:1 – 18:12.
    New York, NY: ACM.

    19. Brunette W, Sundt M, Dell N, Chaudhri R, Breit N,
    Borriello G. 2013 Open Data Kit 2.0: Expanding and
    refining information services for developing regions.

    In Proceedings of the 14th Workshop on Mobile
    Computing Systems and Applications, pp.
    10:1 – 10:6. New York, NY: ACM.

    20. Nagraj VP, Randhawa N, Campbell F, Crellen T,
    Sudre B, Jombart T. 2018 epicontacts: Handling,
    visualisation and analysis of epidemiological
    contacts. F1000Res. 7, 566. (doi:10.12688/
    f1000research.14492.1)

    21. Moraga P, Dorigatti I, Kamvar ZN, Piatkowski P,
    Toikkanen SE, Nagraj VP, Donnelly CA, Jombart T.
    2018 epiflows: an R package for risk assessment of
    travel-related spread of disease. F1000Res. 7, 1374.
    (doi:10.12688/f1000research.16032.1)

    22. Pigott DM et al. 2017 Local, national, and regional
    viral haemorrhagic fever pandemic potential in
    Africa: a multistage analysis. Lancet 390,
    2662 – 2672. (doi:10.1016/S0140-6736(17)32092-5)

    23. Messina JP et al. 2016 Mapping global
    environmental suitability for Zika virus. Elife 5,
    e15272. (doi:10.7554/eLife.15272)

    24. Pigott DM et al. 2014 Mapping the zoonotic niche
    of Ebola virus disease in Africa. Elife 3, e04395.
    (doi:10.7554/eLife.04395)

    25. Jombart T, Eggo RM, Dodd PJ, Balloux F. 2011
    Reconstructing disease outbreaks from genetic data:
    a graph approach. Heredity 106, 383 – 390. (doi:10.
    1038/hdy.2010.78)

    26. Famulare M, Hu H. 2015 Extracting transmission
    networks from phylogeographic data for epidemic
    and endemic diseases: Ebola virus in Sierra Leone,
    2009 H1N1 pandemic influenza and polio in
    Nigeria. Int. Health 7, 130 – 138. (doi:10.1093/
    inthealth/ihv012)

    27. Cori A, Ferguson NM, Fraser C, Cauchemez S. 2013
    A new framework and software to estimate time-
    varying reproduction numbers during epidemics.
    Am. J. Epidemiol. 178, 1505 – 1512. (doi:10.1093/
    aje/kwt133)

    28. Nouvellet P et al. 2017 A simple approach to
    measure transmissibility and forecast incidence.
    Epidemics 22, 29 – 35. (doi:10.1016/j.epidem.2017.
    02.012)

    29. Nouvellet P et al. 2015 The role of rapid diagnostics
    in managing Ebola epidemics. Nature 528,
    S109 – S116. (doi:10.1038/nature16041)

    30. Finger F, Funk S, White K, Siddiqui R, John
    Edmunds W, Kucharski AJ. 2018 Real-time analysis
    of the diphtheria outbreak in forcibly displaced
    Myanmar nationals in Bangladesh. bioRxiv. 388645.
    (doi:10.1101/388645)

    31. Bausch DG, Edmunds J. 2018 Real-time modeling
    should be routinely integrated into outbreak
    response. Am. J. Trop. Med. Hyg. 98, 1214 – 1215.
    (doi:10.4269/ajtmh.18-0150)

    32. Feil EJ, Li BC, Aanensen DM, Hanage WP, Spratt BG.
    2004 eBURST: inferring patterns of evolutionary
    descent among clusters of related bacterial
    genotypes from multilocus sequence typing data.
    J. Bacteriol. 186, 1518 – 1530. (doi:10.1128/JB.186.
    5.1518-1530.2004)

    33. Bouckaert R, Heled J, Kühnert D, Vaughan T, Wu C-
    H, Xie D, Suchard MA, Rambaut A, Drummond AJ.
    2014 BEAST 2: a software platform for Bayesian

    evolutionary analysis. PLoS Comput. Biol. 10,
    e1003537. (doi:10.1371/journal.pcbi.1003537)

    34. Holmes EC, Rambaut A, Andersen KG. 2018
    Pandemics: spend on surveillance, not prediction.
    Nature 558, 180 – 182. (doi:10.1038/d41586-018-
    05373-w)

    35. Campbell F, Strang C, Ferguson N, Cori A, Jombart T.
    2018 When are pathogen genome sequences
    informative of transmission events? PLoS
    Pathog. 14, e1006885. (doi:10.1371/journal.
    ppat.1006885)

    36. Carroll D, Daszak P, Wolfe ND, Gao GF, Morel CM,
    Morzaria S, Pablos-Méndez A, Tomori O, Mazet JAK.
    2018 The Global Virome Project. Science 359,
    872 – 874. (doi:10.1126/science.aap7463)

    37. Birrell PJ, De Angelis D, Presanis AM. 2018 Evidence
    synthesis for stochastic epidemic models. Stat. Sci.
    33, 34 – 43. (doi:10.1214/17-STS631)

    38. Wikipedia contributors. 2018 Data science.
    Wikipedia, The Free Encyclopedia. See https://en.
    wikipedia.org/w/index.php?title=Data_
    science&oldid=868658447 (accessed on 16
    November 2018).

    39. Bausch DG, Clougherty MM. 2015 Ebola virus:
    sensationalism, science, and human rights.
    J. Infect. Dis. 212(Suppl. 2), S79 – S83. (doi:10.
    1093/infdis/jiv359)

    40. Kwong JC et al. 2012 The impact of infection on
    population health: results of the Ontario burden of
    infectious diseases study. PLoS ONE 7, e44103.
    (doi:10.1371/journal.pone.0044103)

    41. Vos T et al. 2012 Years lived with disability (YLDs)
    for 1160 sequelae of 289 diseases and injuries
    1990 – 2010: a systematic analysis for the Global
    Burden of Disease Study 2010. Lancet 380,
    2163 – 2196. (doi:10.1016/S0140-6736(12)61729-2)

    42. Global Burden of Disease Study 2013 Collaborators.
    2015 Global, regional, and national incidence,
    prevalence, and years lived with disability for 301
    acute and chronic diseases and injuries in 188
    countries, 1990 – 2013: a systematic analysis for the
    Global Burden of Disease Study 2013. Lancet 386,
    743 – 800. (doi:10.1016/S0140-6736(15)60692-4)

    43. Prüss-Üstün A et al. 2003 Introduction and methods:
    assessing the environmental burden of disease at
    national and local levels. WHO Environmental
    Burden of Disease Series, No. 1. Geneva,
    Switzerland: World Health Organization.

    44. Camacho A, Eggo RM, Funk S, Watson CH, Kucharski
    AJ, Edmunds WJ. 2015 Estimating the probability of
    demonstrating vaccine efficacy in the declining
    Ebola epidemic: a Bayesian modelling approach.
    BMJ Open 5, e009346. (doi:10.1136/bmjopen-2015-
    009346)

    45. Camacho A et al. 2017 Real-time dynamic
    modelling for the design of a cluster-randomized
    phase 3 Ebola vaccine trial in Sierra Leone. Vaccine
    35, 544 – 551. (doi:10.1016/j.vaccine.2016.12.019)

    46. Garske T, Van Kerkhove MD, Yactayo S, Ronveaux O,
    Lewis RF, Staples JE, Perea W, Ferguson NM, Yellow
    Fever Expert Committee. 2014 Yellow fever in
    Africa: estimating the burden of disease and impact
    of mass vaccination from outbreak and serological

    https://ecdc.europa.eu/sites/portal/files/media/en/publications/Publications/zika-virus-americas-association-with-microcephaly-rapid-risk-assessment

    https://ecdc.europa.eu/sites/portal/files/media/en/publications/Publications/zika-virus-americas-association-with-microcephaly-rapid-risk-assessment

    https://ecdc.europa.eu/sites/portal/files/media/en/publications/Publications/zika-virus-americas-association-with-microcephaly-rapid-risk-assessment

    https://ecdc.europa.eu/sites/portal/files/media/en/publications/Publications/zika-virus-americas-association-with-microcephaly-rapid-risk-assessment

    http://dx.doi.org/10.1056/NEJMoa1411100

    http://dx.doi.org/10.1056/NEJMoa1411100

    http://dx.doi.org/10.1056/NEJMc1414992

    http://dx.doi.org/10.1016/S0140-6736(15)00946-0

    http://dx.doi.org/10.1038/sdata.2015.19

    http://dx.doi.org/10.1098/rstb.2016.0300

    http://dx.doi.org/10.1371/journal.pmed.1002170

    http://dx.doi.org/10.1098/rstb.2016.0371

    http://dx.doi.org/10.1016/S0140-6736(18)31443-0

    http://dx.doi.org/10.1016/S0140-6736(18)31387-4

    http://dx.doi.org/10.1016/S0140-6736(18)31387-4

    https://www.who.int/emergencies/ebola-DRC-2017/en/

    https://www.who.int/emergencies/ebola-DRC-2017/en/

    http://dx.doi.org/10.12688/f1000research.14492.1

    http://dx.doi.org/10.12688/f1000research.14492.1

    http://dx.doi.org/10.12688/f1000research.16032.1

    http://dx.doi.org/10.1016/S0140-6736(17)32092-5

    http://dx.doi.org/10.7554/eLife.15272

    http://dx.doi.org/10.7554/eLife.04395

    http://dx.doi.org/10.1038/hdy.2010.78

    http://dx.doi.org/10.1038/hdy.2010.78

    http://dx.doi.org/10.1093/inthealth/ihv012

    http://dx.doi.org/10.1093/inthealth/ihv012

    http://dx.doi.org/10.1093/aje/kwt133

    http://dx.doi.org/10.1093/aje/kwt133

    http://dx.doi.org/10.1016/j.epidem.2017.02.012

    http://dx.doi.org/10.1016/j.epidem.2017.02.012

    http://dx.doi.org/10.1038/nature16041

    http://dx.doi.org/10.4269/ajtmh.18-0150

    http://dx.doi.org/10.1128/JB.186.5.1518-1530.2004

    http://dx.doi.org/10.1128/JB.186.5.1518-1530.2004

    http://dx.doi.org/10.1371/journal.pcbi.1003537

    http://dx.doi.org/10.1038/d41586-018-05373-w

    http://dx.doi.org/10.1038/d41586-018-05373-w

    http://dx.doi.org/10.1371/journal.ppat.1006885

    http://dx.doi.org/10.1371/journal.ppat.1006885

    http://dx.doi.org/10.1126/science.aap7463

    http://dx.doi.org/10.1214/17-STS631

    https://en.wikipedia.org/w/index.php?title=Data_science&oldid=868658447

    https://en.wikipedia.org/w/index.php?title=Data_science&oldid=868658447

    https://en.wikipedia.org/w/index.php?title=Data_science&oldid=868658447

    https://en.wikipedia.org/w/index.php?title=Data_science&oldid=868658447

    http://dx.doi.org/10.1093/infdis/jiv359

    http://dx.doi.org/10.1093/infdis/jiv359

    http://dx.doi.org/10.1371/journal.pone.0044103

    http://dx.doi.org/10.1016/S0140-6736(12)61729-2

    http://dx.doi.org/10.1016/S0140-6736(15)60692-4

    http://dx.doi.org/10.1136/bmjopen-2015-009346

    http://dx.doi.org/10.1136/bmjopen-2015-009346

    http://dx.doi.org/10.1016/j.vaccine.2016.12.019

    royalsocietypublishing.org/journal/rstb
    Phil.Trans.R.Soc.B
    374:20180276
    9
    data. PLoS Med. 11, e1001638. (doi:10.1371/
    journal.pmed.1001638)

    47. Kraemer MUG et al. 2017 Spread of yellow fever
    virus outbreak in Angola and the Democratic
    Republic of the Congo 2015 – 16: a modelling study.
    Lancet Infect. Dis. 17, 330 – 338. (doi:10.1016/
    S1473-3099(16)30513-8)

    48. Dorigatti I, Hamlet A, Aguas R, Cattarino L, Cori A,
    Donnelly CA, Garske T, Imai N, Ferguson NM. 2017
    International risk of yellow fever spread from the
    ongoing outbreak in Brazil, December 2016 to May
    2017. Euro Surveill. 22, 30572. (doi:10.2807/1560-
    7917.ES.2017.22.28.30572)

    49. Brookmeyer R, You X. 2006 A hypothesis test for the
    end of a common source outbreak. Biometrics 62,
    61 – 65. (doi:10.1111/j.1541-0420.2005.00421.x)

    50. Nishiura H, Miyamatsu Y, Chowell G, Saitoh M. 2015
    Assessing the risk of observing multiple generations
    of Middle East respiratory syndrome (MERS) cases
    given an imported case. Euro Surveill. 20, 21181.
    (doi:10.2807/1560-7917.ES2015.20.27.21181)

    51. Nishiura H, Miyamatsu Y, Mizumoto K. 2016
    Objective determination of end of MERS outbreak,
    South Korea, 2015. Emerg. Infect. Dis. 22, 146 – 148.
    (doi:10.3201/eid2201.151383)

    52. Fähnrich C et al. 2015 Surveillance and outbreak
    response management system (SORMAS) to support
    the control of the Ebola virus disease outbreak in
    West Africa. Euro Surveill. 20, 21071. (doi:10.2807/
    1560-7917.ES2015.20.12.21071)

    53. Polonsky JA, Martı́nez-Pino I, Nackers F, Chonzi P,
    Manangazira P, Van Herp M, Maes P, Porten K,
    Luquero FJ. 2014 Descriptive epidemiology of
    typhoid fever during an epidemic in Harare,
    Zimbabwe, 2012. PLoS One 9, e114702. (doi:10.
    1371/journal.pone.0114702)

    54. Page A-L et al. 2015 Geographic distribution and
    mortality risk factors during the cholera outbreak in
    a rural region of Haiti, 2010 – 2011. PLoS Neglect.
    Trop. Dis. 9, e0003605. (doi:10.1371/journal.pntd.
    0003605)

    55. Aanensen DM, Huntley DM, Feil EJ, al-Own F, Spratt
    BG. 2009 EpiCollect: linking smartphones to web
    applications for epidemiology, ecology and
    community data collection. PLoS One 4, e6968.
    (doi:10.1371/journal.pone.0006968)

    56. Tom-Aba D et al. 2015 Innovative technological
    approach to Ebola virus disease outbreak response
    in Nigeria using the open data kit and form hub
    technology. PLoS One 10, e0131000. (doi:10.1371/
    journal.pone.0131000)

    57. Xie Y, Allaire JJ, Grolemund G. 2018 R markdown:
    The definitive guide. Boca Raton, FL: Chapman and
    Hall/CRC. https://bookdown.org/yihui/rmarkdown/.

    58. Xie Y. 2016 Bookdown: authoring books and
    technical documents with R markdown. Boca Raton,
    FL: CRC Press.

    59. Karo B, Haskew C, Khan AS, Polonsky JA, Mazhar
    MKA, Buddha N. 2018 World Health Organization
    early warning, alert, and response system in the
    Rohingya Crisis, Bangladesh, 2017 – 2018. Emerg.
    Infect. Dis. 24, 2074 – 2076. (doi:10.3201/
    eid2411.181237)

    60. Cauchemez S, Valleron A-J, Boëlle P-Y, Flahault A,
    Ferguson NM. 2008 Estimating the impact of school
    closure on influenza transmission from Sentinel
    data. Nature 452, 750 – 754. (doi:10.1038/
    nature06732)

    61. Cauchemez S, Bhattarai A, Marchbanks TL, Fagan
    RP, Ostroff S, Ferguson NM, Swerdlow D,
    Pennsylvania H1N1 working group. 2011 Role of
    social networks in shaping disease transmission
    during a community outbreak of 2009 H1N1
    pandemic influenza. Proc. Natl Acad. Sci. USA 108,
    2825 – 2830. (doi:10.1073/pnas.1008895108)

    62. Gignoux E, Polonsky J, Ciglenecki I, Bichet M,
    Coldiron M, Thuambe Lwiyo E, Akonda I, Serafini M,
    Porten K. 2018 Risk factors for measles mortality
    and the importance of decentralized case
    management during an unusually large measles
    epidemic in eastern Democratic Republic of Congo
    in 2013. PLoS One 13, e0194276. (doi:10.1371/
    journal.pone.0194276)

    63. Saurabh S, Prateek S. 2017 Role of contact tracing in
    containing the 2014 Ebola outbreak: a review. Afr.
    Health Sci. 17, 225 – 236. (doi:10.4314/ahs.v17i1.28)

    64. Funk S, Camacho A, Kucharski AJ, Eggo RM,
    Edmunds WJ. 2018 Real-time forecasting of
    infectious disease dynamics with a stochastic semi-
    mechanistic model. Epidemics 22, 56 – 61. (doi:10.
    1016/j.epidem.2016.11.003)

    65. Viboud C et al. 2018 The RAPIDD Ebola forecasting
    challenge: synthesis and lessons learnt. Epidemics
    22, 13 – 21. (doi:10.1016/j.epidem.2017.08.002)

    66. Wickham H. 2014 Tidy data. J. Stat. Softw. 59,
    1 – 23. (doi:10.18637/jss.v059.i10)

    67. Dallman T et al. 2016 Phylogenetic structure of
    European Salmonella enteritidis outbreak correlates
    with national and international egg distribution
    network. Microb Genom 2, e000070. (doi:10.1099/
    mgen.0.000070)

    68. Jenkins C et al. 2015 Public health investigation of
    two outbreaks of shiga toxin-producing Escherichia
    coli O157 associated with consumption of
    watercress. Appl. Environ. Microbiol. 81,
    3946 – 3952. (doi:10.1128/AEM.04188-14)

    69. Inns T et al. 2015 A multi-country Salmonella
    enteritidis phage type 14b outbreak associated with
    eggs from a German producer: ‘near real-time’
    application of whole genome sequencing and food
    chain investigations, United Kingdom, May to
    September 2014. Eurosurveillance 20, 21098.
    (doi:10.2807/1560-7917.ES2015.20.16.21098)

    70. Bousema T et al. 2016 The impact of hotspot-
    targeted interventions on malaria transmission in
    Rachuonyo South District in the Western Kenyan
    highlands: a cluster-randomized controlled trial.
    PLoS Med. 13, e1001993. (doi:10.1371/journal.
    pmed.1001993)

    71. Baidjoe AY et al. 2016 Factors associated with high
    heterogeneity of malaria at fine spatial scale in the
    Western Kenyan highlands. Malar. J. 15, 307.
    (doi:10.1186/s12936-016-1362-y)

    72. Ahmed R, Robinson R, Elsony A, Thomson R, Squire
    SB, Malmborg R, Burney P, Mortimer K. 2018 A
    comparison of smartphone and paper data-

    collection tools in the Burden of Obstructive Lung
    Disease (BOLD) study in Gezira state, Sudan. PLoS
    One 13, e0193917. (doi:10.1371/journal.pone.
    0193917)

    73. Solomon AW et al. 2018 Quality assurance and
    quality control in the global trachoma mapping
    project. Am. J. Trop. Med. Hyg. 99, 858 – 863.
    (doi:10.4269/ajtmh.18-0082)

    74. King JD et al. 2013 A novel electronic data collection
    system for large-scale surveys of neglected tropical
    diseases. PLoS One 8, e74570. (doi:10.1371/journal.
    pone.0074570)

    75. Njuguna HN et al. 2014 A comparison of
    smartphones to paper-based questionnaires for
    routine influenza sentinel surveillance, Kenya,
    2011 – 2012. BMC Med. Inform. Decis. Mak. 14, 107.
    (doi:10.1186/s12911-014-0107-5)

    76. Poushter J. 2016 Smartphone ownership and
    internet usage continues to climb in emerging
    economies. Pew Res. Center 22, 1 – 44.

    77. Bogoch II, Koydemir HC, Tseng D, Ephraim RKD,
    Duah E, Tee J, Andrews JR, Ozcan A. 2017
    Evaluation of a mobile phone-based microscope for
    screening of Schistosoma haematobium infection in
    rural Ghana. Am. J. Trop. Med. Hyg. 96,
    1468 – 1471. (doi:10.4269/ajtmh.16-0912)

    78. Kühnemund M et al. 2017 Targeted DNA
    sequencing and in situ mutation analysis using
    mobile phone microscopy. Nat. Commun. 8, 13913.
    (doi:10.1038/ncomms13913)

    79. Quesada-González D, Merkoçi A. 2017 Mobile
    phone-based biosensing: an emerging ‘diagnostic
    and communication’ technology. Biosens.
    Bioelectron. 92, 549 – 562. (doi:10.1016/j.bios.
    2016.10.062)

    80. Macharia P, Dunbar MD, Sambai B, Abuna F,
    Betz B, Njoroge A, Bukusi D, Cherutich P,
    Farquhar C. 2015 Enhancing data security in open
    data kit as an mHealth application. In 2015
    International Conference on Computing,
    Communication and Security (ICCCS),
    Pamplemousses, Mauritius, 4 – 5 December 2015.
    (doi:10.1109/cccs.2015.7374205)

    81. Crawley MJ. 2012 The R book. Hoboken, NJ: John
    Wiley & Sons.

    82. Wickham H. 2016 Ggplot2: elegant graphics for data
    analysis. Berlin, Germany: Springer.

    83. Höhle M. 2007 surveillance: An R package for the
    monitoring of infectious diseases. Comput. Stat. 22,
    571 – 582. (doi:10.1007/s00180-007-0074-8)

    84. Jombart T et al. 2014 OutbreakTools: a new
    platform for disease outbreak analysis using the R
    software. Epidemics 7, 28 – 34. (doi:10.1016/j.
    epidem.2014.04.003)

    85. Jombart T, Kamvar ZN, FitzJohn R. 2018 Incidence:
    compute, handle, plot and model incidence of
    dated events. R package version 1.5.4. https://
    CRAN.R-project.org/package¼incidence.

    86. King AA, Domenech de Cellès M, Magpantay FMG,
    Rohani P. 2015 Avoidable errors in the modelling of
    outbreaks of emerging pathogens, with special
    reference to Ebola. Proc. R. Soc. B 282, 20150347.
    (doi:10.1098/rspb.2015.0347)

    http://dx.doi.org/10.1371/journal.pmed.1001638

    http://dx.doi.org/10.1371/journal.pmed.1001638

    http://dx.doi.org/10.1016/S1473-3099(16)30513-8

    http://dx.doi.org/10.1016/S1473-3099(16)30513-8

    http://dx.doi.org/10.2807/1560-7917.ES.2017.22.28.30572

    http://dx.doi.org/10.2807/1560-7917.ES.2017.22.28.30572

    http://dx.doi.org/10.1111/j.1541-0420.2005.00421.x

    http://dx.doi.org/10.2807/1560-7917.ES2015.20.27.21181

    http://dx.doi.org/10.3201/eid2201.151383

    http://dx.doi.org/10.2807/1560-7917.ES2015.20.12.21071

    http://dx.doi.org/10.2807/1560-7917.ES2015.20.12.21071

    http://dx.doi.org/10.1371/journal.pone.0114702

    http://dx.doi.org/10.1371/journal.pone.0114702

    http://dx.doi.org/10.1371/journal.pntd.0003605

    http://dx.doi.org/10.1371/journal.pntd.0003605

    http://dx.doi.org/10.1371/journal.pone.0006968

    http://dx.doi.org/10.1371/journal.pone.0131000

    http://dx.doi.org/10.1371/journal.pone.0131000

    https://bookdown.org/yihui/rmarkdown/

    http://dx.doi.org/10.3201/eid2411.181237

    http://dx.doi.org/10.3201/eid2411.181237

    http://dx.doi.org/10.1038/nature06732

    http://dx.doi.org/10.1038/nature06732

    http://dx.doi.org/10.1073/pnas.1008895108

    http://dx.doi.org/10.1371/journal.pone.0194276

    http://dx.doi.org/10.1371/journal.pone.0194276

    http://dx.doi.org/10.4314/ahs.v17i1.28

    http://dx.doi.org/10.1016/j.epidem.2016.11.003

    http://dx.doi.org/10.1016/j.epidem.2016.11.003

    http://dx.doi.org/10.1016/j.epidem.2017.08.002

    http://dx.doi.org/10.18637/jss.v059.i10

    http://dx.doi.org/10.1099/mgen.0.000070

    http://dx.doi.org/10.1099/mgen.0.000070

    http://dx.doi.org/10.1128/AEM.04188-14

    http://dx.doi.org/10.2807/1560-7917.ES2015.20.16.21098

    http://dx.doi.org/10.1371/journal.pmed.1001993

    http://dx.doi.org/10.1371/journal.pmed.1001993

    http://dx.doi.org/10.1186/s12936-016-1362-y

    http://dx.doi.org/10.1371/journal.pone.0193917

    http://dx.doi.org/10.1371/journal.pone.0193917

    http://dx.doi.org/10.4269/ajtmh.18-0082

    http://dx.doi.org/10.1371/journal.pone.0074570

    http://dx.doi.org/10.1371/journal.pone.0074570

    http://dx.doi.org/10.1186/s12911-014-0107-5

    http://dx.doi.org/10.4269/ajtmh.16-0912

    http://dx.doi.org/10.1038/ncomms13913

    http://dx.doi.org/10.1016/j.bios.2016.10.062

    http://dx.doi.org/10.1016/j.bios.2016.10.062

    http://dx.doi.org/10.1109/cccs.2015.7374205

    http://dx.doi.org/10.1007/s00180-007-0074-8

    http://dx.doi.org/10.1016/j.epidem.2014.04.003

    http://dx.doi.org/10.1016/j.epidem.2014.04.003

    https://CRAN.R-project.org/package=incidence

    https://CRAN.R-project.org/package=incidence

    https://CRAN.R-project.org/package=incidence

    https://CRAN.R-project.org/package=incidence

    http://dx.doi.org/10.1098/rspb.2015.0347

    royalsocietypublishing.org/journal/rstb
    Phil.Trans.R.Soc.B
    374:20180276
    10
    87. Snow J. 1855 On the mode of communication of
    cholera. London, UK: John Churchill.

    88. Wertheim HFL, Horby P, Woodall JP. 2012 Atlas of
    human infectious diseases. Hoboken, NJ: John Wiley
    & Sons.

    89. Nunes MRT et al. 2015 Emergence and potential for
    spread of Chikungunya virus in Brazil. BMC Med. 13,
    102. (doi:10.1186/s12916-015-0348-x)

    90. In press. Radiant Earth Foundation – Earth imagery
    for impact. See https://www.radiant.earth (accessed
    on 18 November 2018).

    91. In press. Spatial epidemiology of Viral Hemorrhagic
    Fevers. See http://www.healthdata.org/data-
    visualization/spatial-epidemiology-viral-
    hemorrhagic-fevers (accessed on 19 September
    2018).

    92. Hadfield J, Megill C, Bell SM, Huddleston J, Potter
    B, Callender C, Sagulenko P, Bedford T, Neher RA.
    2018 Nextstrain: real-time tracking of pathogen
    evolution. Bioinformatics 34, 4121 – 4123. (doi:10.
    1093/bioinformatics/bty407)

    93. Argimón S et al. 2016 Microreact: visualizing and
    sharing data for genomic epidemiology and
    phylogeography. Microb Genom 2, e000093. (doi:10.
    1099/mgen.0.000093)

    94. Freifeld CC, Mandl KD, Reis BY, Brownstein JS.
    2008 HealthMap: global infectious disease
    monitoring through automated classification and
    visualization of Internet media reports. J. Am. Med.
    Inform. Assoc. 15, 150 – 157. (doi:10.1197/jamia.
    M2544)

    95. Backer JA, Wallinga J. 2016 Spatiotemporal analysis
    of the 2014 Ebola epidemic in West Africa. PLoS
    Comput. Biol. 12, e1005210. (doi:10.1371/journal.
    pcbi.1005210)

    96. Wallinga J, Teunis P. 2004 Different epidemic curves
    for severe acute respiratory syndrome reveal similar
    impacts of control measures. Am. J. Epidemiol. 160,
    509 – 516. (doi:10.1093/aje/kwh255)

    97. Wallinga J, Lipsitch M. 2007 How generation
    intervals shape the relationship between growth
    rates and reproductive numbers. Proc. R. Soc. B 274,
    599 – 604. (doi:10.1098/rspb.2006.3754)

    98. Cauchemez S, Van Kerkhove MD, Riley S, Donnelly
    CA, Fraser C, Ferguson NM. 2013 Transmission
    scenarios for Middle East Respiratory Syndrome
    Coronavirus (MERS-CoV) and how to tell them
    apart. Euro Surveill. 18, 20503.

    99. Shrivastava SR, Shrivastava PS, Ramasamy J. 2014
    Utility of contact tracing in reducing the magnitude
    of Ebola disease. Germs 4, 97 – 99. (doi:10.11599/
    germs.2014.1063)

    100. WHO Ebola Response Team et al. 2015 Ebola virus
    disease among children in West Africa.
    N. Engl. J. Med. 372, 1274 – 1277. (doi:10.1056/
    NEJMc1415318)

    101. WHO Ebola Response Team. 2016 Ebola virus
    disease among male and female persons in West
    Africa. N. Engl. J. Med. 374, 96 – 98. (doi:10.1056/
    NEJMc1510305)

    102. Donnelly CA et al. 2003 Epidemiological
    determinants of spread of causal agent of severe
    acute respiratory syndrome in Hong Kong. Lancet

    361, 1761 – 1766. (doi:10.1016/S0140-
    6736(03)13410-1)

    103. Anderson RM, Fraser C, Ghani AC, Donnelly CA, Riley
    S, Ferguson NM, Leung GM, Lam TH, Hedley AJ.
    2004 Epidemiology, transmission dynamics and
    control of SARS: the 2002 – 2003 epidemic. Phil.
    Trans. R. Soc. Lond. B 359, 1091 – 1105. (doi:10.
    1098/rstb.2004.1490)

    104. Anderson RM, May RM. 1991 Infectious diseases of
    humans, vol. 1. Oxford, UK: Oxford University Press.

    105. Farrington CP, Andrews NJ, Beale AD, Catchpole MA.
    1996 A statistical algorithm for the early detection
    of outbreaks of infectious disease. J. R. Stat. Soc.
    Ser. A Stat. Soc. 159, 547 – 563. (doi:10.2307/
    2983331)

    106. Park SW, Champredon D, Weitz J, Dushoff J. 2018 A
    practical generation interval-based approach to
    inferring the strength of epidemics from their
    speed. bioRxiv. 312397. (doi:10.1101/312397)

    107. Keeling M, Rohani P. 2008 Modeling infectious
    diseases in humans and animals. Clin. Infect. Dis.
    47, 864 – 866. (doi:10.1086/591197)

    108. McKinley T, Cook AR, Deardon R. 2009 Inference in
    epidemic models without likelihoods. Int. J. Biostat.
    5(1): Article 24. (doi:10.2202/1557-4679.1171)

    109. Obadia T, Haneef R, Boëlle P-Y. 2012 The R0
    package: a toolbox to estimate reproduction
    numbers for epidemic outbreaks. BMC Med.
    Inform. Decis. Mak. 12, 147. (doi:10.1186/1472-
    6947-12-147)

    110. Chowell G, Viboud C, Simonsen L, Merler S,
    Vespignani A. 2017 Perspectives on model forecasts
    of the 2014 – 2015 Ebola epidemic in West Africa:
    lessons and the way forward. BMC Med. 15, 42.
    (doi:10.1186/s12916-017-0811-y)

    111. Held L, Meyer S, Bracher J. 2017 Probabilistic
    forecasting in infectious disease epidemiology: the
    13th Armitage lecture. Stat. Med. 36, 3443 – 3460.
    (doi:10.1002/sim.7363)

    112. Funk S, Camacho A, Kucharski AJ, Lowe R, Eggo RM,
    Edmunds WJ. 2017 Assessing the performance of
    real-time epidemic forecasts. bioRxiv. 177451.
    (doi:10.1101/177451)

    113. Kucharski AJ, Camacho A, Flasche S, Glover RE,
    Edmunds WJ, Funk S. 2015 Measuring the impact of
    Ebola control measures in Sierra Leone. Proc. Natl
    Acad. Sci. USA 112, 14 366 – 14 371. (doi:10.1073/
    pnas.1508814112)

    114. Buring JE. 1987 Epidemiology in medicine.
    Philadelphia, PA: Lippincott Williams & Wilkins.

    115. Grandesso F et al. 2014 Risk factors for cholera
    transmission in Haiti during inter-peak periods:
    insights to improve current control strategies from
    two case-control studies. Epidemiol. Infect. 142,
    1625 – 1635. (doi:10.1017/S0950268813002562)

    116. Gross M. 1976 Oswego County revisited. Public
    Health Rep. 91, 168 – 170.

    117. Buchholz U et al. 2011 German outbreak of
    Escherichia coli O104:H4 associated with sprouts.
    N. Engl. J. Med. 365, 1763 – 1770. (doi:10.1056/
    NEJMoa1106482)

    118. Ebola ça Suffit Ring Vaccination Trial Consortium.
    2015 The ring vaccination trial: a novel cluster

    randomised controlled trial design to evaluate
    vaccine efficacy and effectiveness during outbreaks,
    with special reference to Ebola. BMJ 351, h3740.
    (doi:10.1136/bmj.h3740)

    119. Grais RF, Conlan AJK, Ferrari MJ, Djibo A, Le Menach
    A, Bjørnstad ON, Grenfell BT. 2008 Time is of the
    essence: exploring a measles outbreak response
    vaccination in Niamey, Niger. J. R. Soc. Interface 5,
    67 – 74. (doi:10.1098/rsif.2007.1038)

    120. Harris SR et al. 2013 Whole-genome sequencing for
    analysis of an outbreak of meticillin-resistant
    Staphylococcus aureus: a descriptive study. Lancet
    Infect. Dis. 13, 130 – 136. (doi:10.1016/S1473-
    3099(12)70268-2)

    121. Gire SK et al. 2014 Genomic surveillance elucidates
    Ebola virus origin and transmission during the 2014
    outbreak. Science 345, 1369 – 1372. (doi:10.1126/
    science.1259657)

    122. Cotten M et al. 2013 Transmission and evolution of
    the Middle East respiratory syndrome coronavirus in
    Saudi Arabia: a descriptive genomic study. Lancet
    382, 1993 – 2002. (doi:10.1016/S0140-6736(13)
    61887-5)

    123. Robinson ER, Walker TM, Pallen MJ. 2013 Genomics
    and outbreak investigation: from sequence to
    consequence. Genome Med. 5, 36. (doi:10.1186/
    gm440)

    124. Hatherell H-A, Didelot X, Pollock SL, Tang P, Crisan
    A, Johnston JC, Colijn C, Gardy JL. 2016 Declaring a
    tuberculosis outbreak over with genomic
    epidemiology. Microb. Genomics 2, e000060.
    (doi:10.1099/mgen.0.000060)

    125. Dudas G et al. 2017 Virus genomes reveal
    factors that spread and sustained the Ebola
    epidemic. Nature 544, 309 – 315. (doi:10.1038/
    nature22040)

    126. Faria NR et al. 2017 Establishment and cryptic
    transmission of Zika virus in Brazil and the
    Americas. Nature 546, 406 – 410. (doi:10.1038/
    nature22401)

    127. Quick J et al. 2016 Real-time, portable genome
    sequencing for Ebola surveillance. Nature 530,
    228 – 232. (doi:10.1038/nature16996)

    128. Pallen MJ, Loman NJ, Penn CW. 2010 High-
    throughput sequencing and clinical microbiology:
    progress, opportunities and challenges. Curr. Opin.
    Microbiol. 13, 625 – 631. (doi:10.1016/j.mib.2010.
    08.003)

    129. Spratt BG, Hanage WP, Li B, Aanensen DM, Feil EJ.
    2004 Displaying the relatedness among isolates of
    bacterial species—the eBURST approach. FEMS
    Microbiol. Lett. 241, 129 – 134. (doi:10.1016/j.
    femsle.2004.11.015)

    130. Enright MC, Robinson DA, Randle G, Feil EJ,
    Grundmann H, Spratt BG. 2002 The evolutionary
    history of methicillin-resistant Staphylococcus aureus
    (MRSA). Proc. Natl Acad. Sci. USA 99, 7687 – 7692.
    (doi:10.1073/pnas.122108599)

    131. King SJ, Leigh JA, Heath PJ, Luque I, Tarradas C,
    Dowson CG, Whatmore AM. 2002 Development of a
    multilocus sequence typing scheme for the pig
    pathogen Streptococcus suis: identification of
    virulent clones and potential capsular serotype

    http://dx.doi.org/10.1186/s12916-015-0348-x

    https://www.radiant.earth

    https://www.radiant.earth

    http://www.healthdata.org/data-visualization/spatial-epidemiology-viral-hemorrhagic-fevers

    http://www.healthdata.org/data-visualization/spatial-epidemiology-viral-hemorrhagic-fevers

    http://www.healthdata.org/data-visualization/spatial-epidemiology-viral-hemorrhagic-fevers

    http://www.healthdata.org/data-visualization/spatial-epidemiology-viral-hemorrhagic-fevers

    http://dx.doi.org/10.1093/bioinformatics/bty407

    http://dx.doi.org/10.1093/bioinformatics/bty407

    http://dx.doi.org/10.1099/mgen.0.000093

    http://dx.doi.org/10.1099/mgen.0.000093

    http://dx.doi.org/10.1197/jamia.M2544

    http://dx.doi.org/10.1197/jamia.M2544

    http://dx.doi.org/10.1371/journal.pcbi.1005210

    http://dx.doi.org/10.1371/journal.pcbi.1005210

    http://dx.doi.org/10.1093/aje/kwh255

    http://dx.doi.org/10.1098/rspb.2006.3754

    http://dx.doi.org/10.11599/germs.2014.1063

    http://dx.doi.org/10.11599/germs.2014.1063

    http://dx.doi.org/10.1056/NEJMc1415318

    http://dx.doi.org/10.1056/NEJMc1415318

    http://dx.doi.org/10.1056/NEJMc1510305

    http://dx.doi.org/10.1056/NEJMc1510305

    http://dx.doi.org/10.1016/S0140-6736(03)13410-1

    http://dx.doi.org/10.1016/S0140-6736(03)13410-1

    http://dx.doi.org/10.1098/rstb.2004.1490

    http://dx.doi.org/10.1098/rstb.2004.1490

    http://dx.doi.org/10.2307/2983331

    http://dx.doi.org/10.2307/2983331

    http://dx.doi.org/10.1086/591197

    http://dx.doi.org/10.2202/1557-4679.1171

    http://dx.doi.org/10.1186/1472-6947-12-147

    http://dx.doi.org/10.1186/1472-6947-12-147

    http://dx.doi.org/10.1186/s12916-017-0811-y

    http://dx.doi.org/10.1002/sim.7363

    http://dx.doi.org/10.1101/177451

    http://dx.doi.org/10.1073/pnas.1508814112

    http://dx.doi.org/10.1073/pnas.1508814112

    http://dx.doi.org/10.1017/S0950268813002562

    http://dx.doi.org/10.1056/NEJMoa1106482

    http://dx.doi.org/10.1056/NEJMoa1106482

    http://dx.doi.org/10.1136/bmj.h3740

    http://dx.doi.org/10.1098/rsif.2007.1038

    http://dx.doi.org/10.1016/S1473-3099(12)70268-2

    http://dx.doi.org/10.1016/S1473-3099(12)70268-2

    http://dx.doi.org/10.1126/science.1259657

    http://dx.doi.org/10.1126/science.1259657

    http://dx.doi.org/10.1016/S0140-6736(13)61887-5

    http://dx.doi.org/10.1016/S0140-6736(13)61887-5

    http://dx.doi.org/10.1186/gm440

    http://dx.doi.org/10.1186/gm440

    http://dx.doi.org/10.1099/mgen.0.000060

    http://dx.doi.org/10.1038/nature22040

    http://dx.doi.org/10.1038/nature22040

    http://dx.doi.org/10.1038/nature22401

    http://dx.doi.org/10.1038/nature22401

    http://dx.doi.org/10.1038/nature16996

    http://dx.doi.org/10.1016/j.mib.2010.08.003

    http://dx.doi.org/10.1016/j.mib.2010.08.003

    http://dx.doi.org/10.1016/j.femsle.2004.11.015

    http://dx.doi.org/10.1016/j.femsle.2004.11.015

    http://dx.doi.org/10.1073/pnas.122108599

    royalsocietypublishing.org/journal/rstb
    Phil.Trans.R.Soc.B
    374:20180276
    11
    exchange. J. Clin. Microbiol. 40, 3671 – 3680.
    (doi:10.1128/JCM.40.10.3671-3680.2002)

    132. Urwin R, Maiden MCJ. 2003 Multi-locus sequence
    typing: a tool for global epidemiology. Trends Microbiol.
    11, 479 – 487. (doi:10.1016/j.tim.2003.08.006)

    133. Felsenstein J. 2004 Inferring phylogenies.
    Sunderland, MA: Sinauer Associates Sunderland.

    134. Popescu A-A, Huber KT, Paradis E. 2012 ape 3.0:
    New tools for distance-based phylogenetics and
    evolutionary analysis in R. Bioinformatics 28,
    1536 – 1537. (doi:10.1093/bioinformatics/bts184)

    135. Felsenstein J. 1981 Evolutionary trees from DNA
    sequences: a maximum likelihood approach. J. Mol.
    Evol. 17, 368 – 376. (doi:10.1007/BF01734359)

    136. Schliep KP. 2011 phangorn: phylogenetic analysis in
    R. Bioinformatics 27, 592 – 593. (doi:10.1093/
    bioinformatics/btq706)

    137. Ronquist F, Huelsenbeck JP. 2003 MrBayes 3:
    Bayesian phylogenetic inference under mixed
    models. Bioinformatics 19, 1572 – 1574. (doi:10.
    1093/bioinformatics/btg180)

    138. Grubaugh ND, Faria NR, Andersen KG, Pybus OG.
    2018 Genomic insights into zika virus emergence
    and spread. Cell 172, 1160 – 1162. (doi:10.1016/j.
    cell.2018.02.027)

    139. Smith GJD et al. 2009 Origins and evolutionary
    genomics of the 2009 swine-origin H1N1 influenza
    A epidemic. Nature 459, 1122 – 1125. (doi:10.1038/
    nature08182)

    140. Siddle KJ et al. 2018 Genomic analysis of lassa virus
    during an increase in cases in Nigeria in 2018.
    N. Engl. J. Med. 379, 1745 – 1753. (doi:10.1056/
    NEJMoa1804498)

    141. Grenfell BT, Pybus OG, Gog JR, Wood JLN, Daly JM,
    Mumford JA, Holmes EC. 2004 Unifying the
    epidemiological and evolutionary dynamics of
    pathogens. Science 303, 327 – 332. (doi:10.1126/
    science.1090727)

    142. Cottam EM, Thébaud G, Wadsworth J, Gloster J,
    Mansley L, Paton DJ, King DP, Haydon DT. 2008
    Integrating genetic and epidemiological data to
    determine transmission pathways of foot-and-
    mouth disease virus. Proc. R. Soc. B 275, 887 – 895.
    (doi:10.1098/rspb.2007.1442)

    143. Ypma RJF, Bataille AMA, Stegeman A, Koch G,
    Wallinga J, van Ballegooijen WM. 2012 Unravelling
    transmission trees of infectious diseases by
    combining genetic and epidemiological data. Proc.
    R. Soc. B 279, 444 – 450. (doi:10.1098/rspb.2011.
    0913)

    144. Ypma RJF, van Ballegooijen WM, Wallinga J.
    2013 Relating phylogenetic trees to transmission
    trees of infectious disease outbreaks. Genetics
    195, 1055 – 1062. (doi:10.1534/genetics.
    113.154856)

    145. Jombart T, Cori A, Didelot X, Cauchemez S, Fraser C,
    Ferguson N. 2014 Bayesian reconstruction of disease
    outbreaks by combining epidemiologic and genomic

    data. PLoS Comput. Biol. 10, e1003457. (doi:10.
    1371/journal.pcbi.1003457)

    146. Klinkenberg D, Backer JA, Didelot X, Colijn C,
    Wallinga J. 2017 Simultaneous inference of
    phylogenetic and transmission trees in infectious
    disease outbreaks. PLoS Comput. Biol. 13, e1005495.
    (doi:10.1371/journal.pcbi.1005495)

    147. Didelot X, Gardy J, Colijn C. 2014 Bayesian inference
    of infectious disease transmission from whole-
    genome sequence data. Mol. Biol. Evol. 31,
    1869 – 1879. (doi:10.1093/molbev/msu121)

    148. De Maio N, Wu C-H, Wilson DJ. 2016 SCOTTI:
    efficient reconstruction of transmission within
    outbreaks with the structured coalescent. PLoS
    Comput. Biol. 12, e1005130. (doi:10.1371/journal.
    pcbi.1005130)

    149. Springborn M, Chowell G, MacLachlan M, Fenichel
    EP. 2015 Accounting for behavioral responses during
    a flu epidemic using home television viewing. BMC
    Infect. Dis. 15, 21. (doi:10.1186/s12879-014-0691-0)

    150. R Core Team. 2018 R: a language and environment
    for statistical computing. Vienna, Austria: R
    Foundation for Statistical Computing. https://www.
    R-project.org/.

    151. RECON-R Epidemics Consortium. 2018 R epidemics
    consortium. See https://www.repidemicsconsortium.
    org/ (accessed on 26 September 2018).

    152. 2018 RECON learn. See https://www.reconlearn.org
    (accessed on 26 September 2018).

    http://dx.doi.org/10.1128/JCM.40.10.3671-3680.2002

    http://dx.doi.org/10.1016/j.tim.2003.08.006

    http://dx.doi.org/10.1093/bioinformatics/bts184

    http://dx.doi.org/10.1007/BF01734359

    http://dx.doi.org/10.1093/bioinformatics/btq706

    http://dx.doi.org/10.1093/bioinformatics/btq706

    http://dx.doi.org/10.1093/bioinformatics/btg180

    http://dx.doi.org/10.1093/bioinformatics/btg180

    http://dx.doi.org/10.1016/j.cell.2018.02.027

    http://dx.doi.org/10.1016/j.cell.2018.02.027

    http://dx.doi.org/10.1038/nature08182

    http://dx.doi.org/10.1038/nature08182

    http://dx.doi.org/10.1056/NEJMoa1804498

    http://dx.doi.org/10.1056/NEJMoa1804498

    http://dx.doi.org/10.1126/science.1090727

    http://dx.doi.org/10.1126/science.1090727

    http://dx.doi.org/10.1098/rspb.2007.1442

    http://dx.doi.org/10.1098/rspb.2011.0913

    http://dx.doi.org/10.1098/rspb.2011.0913

    http://dx.doi.org/10.1534/genetics.113.154856

    http://dx.doi.org/10.1534/genetics.113.154856

    http://dx.doi.org/10.1371/journal.pcbi.1003457

    http://dx.doi.org/10.1371/journal.pcbi.1003457

    http://dx.doi.org/10.1371/journal.pcbi.1005495

    http://dx.doi.org/10.1093/molbev/msu121

    http://dx.doi.org/10.1371/journal.pcbi.1005130

    http://dx.doi.org/10.1371/journal.pcbi.1005130

    http://dx.doi.org/10.1186/s12879-014-0691-0

    https://www.R-project.org/

    https://www.R-project.org/

    https://www.repidemicsconsortium.org/

    https://www.repidemicsconsortium.org/

    https://www.repidemicsconsortium.org/

    https://www.reconlearn.org

    https://www.reconlearn.org

    • Outbreak analytics: a developing data science for informing the response to emerging pathogens
    • Introduction
      The outbreak response context
      The different phases of an outbreak response
      Questions during and after the intervention
      What are outbreak data?
      Outbreak analytics
      An overview of the outbreak analytics toolbox
      Tools for the collection of epidemiological data
      Descriptive analyses
      Quantifying transmissibility
      Analytical epidemiological techniques
      Genetic analyses
      Discussion
      Data accessibility
      Authors’ contributions
      Competing interests
      Funding
      Acknowledgements
      References

    Calculate your order
    Pages (275 words)
    Standard price: $0.00
    Client Reviews
    4.9
    Sitejabber
    4.6
    Trustpilot
    4.8
    Our Guarantees
    100% Confidentiality
    Information about customers is confidential and never disclosed to third parties.
    Original Writing
    We complete all papers from scratch. You can get a plagiarism report.
    Timely Delivery
    No missed deadlines – 97% of assignments are completed in time.
    Money Back
    If you're confident that a writer didn't follow your order details, ask for a refund.

    Calculate the price of your order

    You will get a personal manager and a discount.
    We'll send you the first draft for approval by at
    Total price:
    $0.00
    Power up Your Academic Success with the
    Team of Professionals. We’ve Got Your Back.
    Power up Your Study Success with Experts We’ve Got Your Back.

    Order your essay today and save 30% with the discount code ESSAYHELP