You are on page 1of 13

Seminar

Measles
William J Moss

Measles is a highly contagious disease that results from infection with measles virus and is still responsible for more Published Online
than 100 000 deaths every year, down from more than 2 million deaths annually before the introduction and June 30, 2017
http://dx.doi.org/10.1016/
widespread use of measles vaccine. Measles virus is transmitted by the respiratory route and illness begins with fever, S0140-6736(17)31463-0
cough, coryza, and conjunctivitis followed by a characteristic rash. Complications of measles affect most organ Department of Epidemiology
systems, with pneumonia accounting for most measles-associated morbidity and mortality. The management of (Prof W J Moss MD),
patients with measles includes provision of vitamin A. Measles is best prevented through vaccination, and the major Department of International
reductions in measles incidence and mortality have renewed interest in regional elimination and global eradication. Health (Prof W J Moss), W Harry
Feinstone Department of
However, urgent efforts are needed to increase stagnating global coverage with two doses of measles vaccine through Molecular Microbiology and
advocacy, education, and the strengthening of routine immunisation systems. Use of combined measles-rubella Immunology (Prof W J Moss),
vaccines provides an opportunity to eliminate rubella and congenital rubella syndrome. Ongoing research efforts, and International Vaccine
including the development of point-of-care diagnostics and microneedle patches, will facilitate progress towards Access Center (Prof W J Moss),
Bloomberg School of Public
measles elimination and eradication. Health, Johns Hopkins
University, Baltimore, MD, USA
Introduction manifestations, diagnosis, management, and prevention Correspondence to:
Measles is a highly contagious, acute febrile illness that of measles, highlighting recent research findings as well Prof William J Moss, Department
of Epidemiology, Bloomberg
results from infection with measles virus. Measles virus as the progress and challenges of measles elimination School of Public Health,
is most closely related genetically to rinderpest virus, a and eradication (panel 1). Johns Hopkins University,
pathogen of cattle that was declared eradicated by the Baltimore, MD 21205, USA
World Organization for Animal Health in May, 2011, and Disease burden wmoss1@jhu.edu

probably evolved as a zoonotic infection in communities Deaths due to measles have declined substantially over
in which humans and cattle lived together.1 Although the past century, first through improvements in nutrition,
historical evidence is lacking, epidemiological evidence socioeconomic status, and health care and subsequently
suggests measles likely became a disease of humans through a major reduction in measles incidence as a
5000–10 000 years ago when early agrarian civilisations in consequence of increasing measles vaccine coverage.7
the fertile crescent achieved sufficient population size to This progress has had the perverse effect of diminishing
maintain virus transmission.2,3 Measles was a leading the perceived public health importance of measles and
global cause of child morbidity and mortality before the the value of measles vaccination. Nevertheless, there is
introduction of measles vaccines in the 1960s, and was no doubt that the burden of measles, including
responsible for more than 2 million deaths annually pneumonia, blindness, chronic neurological conditions,
before the increase in global measles vaccine coverage in and death, has decreased substantially because of
the 1980s as a result of the Expanded Programme on measles vaccination. Precise measurements of measles
Immunization (figure 1). Measles incidence and incidence and mortality are lacking, however, because
mortality have declined substantially over the past most cases and deaths occur in countries with poor vital
two decades due to the increasingly widespread use of registration and disease surveillance systems.
attenuated measles vaccines administered through Consequently, estimates are based on imperfect reporting
routine immunisation programmes and mass and models. Almost all countries use case-based
vaccination campaigns. Despite this enormous progress, surveillance and have access to standardised laboratory
measles remains an important vaccine-preventable cause testing through the WHO Global Measles and Rubella
of morbidity and mortality, responsible for more than Laboratory Network for diagnostic confirmation and
100 000 deaths each year, and serves as an indicator of the
quality of immunisation programmes. The reduction in
measles incidence and mortality, along with progress Search strategy and selection criteria
toward achieving polio eradication, have renewed interest I searched PubMed for publications in English using the terms
in measles regional elimination and global eradication.4 “measles”, “measles virus”, “measles and epidemiology”,
However, increased political will, public support, and “measles and pathophysiology”, “measles and diagnosis”,
financial resources, facilitated by new instruments, “measles and treatment”, and “measles and prevention”. My
technologies, and strategies, such as point-of-care search focused on, but was not restricted to, publications in
diagnostics and microneedle patches, will be needed to the past 4 years. I also searched the Cochrane Database of
achieve regional measles elimination goals and eventual Systematic Reviews using the term “measles” and our own
eradication.5 This primer updates a previous Lancet database of references, as well as those of linked articles in
seminar published in 20126 and summarises current the searched journals. When more than one article illustrated
knowledge of the disease burden, epidemiology, a specific point, the most representative article was chosen.
virology, pathophysiology, immune responses, clinical

www.thelancet.com Published online June 30, 2016 http://dx.doi.org/10.1016/S0140-6736(17)31463-0 1


Seminar

molecular epidemiology,8 consisting of 703 laboratories although these reports vastly underestimate the true
that support surveillance in 191 countries. number of measles cases. Improving measles surveillance
WHO publishes annually the reported number of and reporting will be crucial to achieving regional and
measles cases and estimated number of deaths, as well as global milestones. Most reported measles cases in 2015
estimates of national measles vaccine coverage for both were from the African (40%), Western Pacific (27%), and
the first and second doses (figures 2, 3). The number of South East Asia (12%, with 88% of these from India)
reported measles cases decreased worldwide between Regions.9 Importantly, 11% of reported global measles
2000 to 2015 by 70%, from 853 479 to 254 928 cases,9 cases were from the European region (25 947 cases).
WHO measles mortality estimates are derived from a
model based on the number and age distribution of
Panel 1: Important recent developments in measles reported cases, measles vaccine coverage (routine and
• Disease burden: estimated global measles deaths decreased 79% from 2000 to 2015 supplemental mass vaccination), and age and country
• Epidemiology: measles outbreaks have been reported in populations with immunity specific case fatality ratios.10 Between 2000 and 2015,
gaps despite high overall vaccine coverage, including individuals who received estimated measles deaths decreased by 79% from 651 600
two doses of measles vaccine (95% CI 449 900–1 034 500) to 134 200 (74 400–353 600;
• Virology: only eight of the 24 known measles virus genotypes have been detected figure 2).9 Almost two-thirds (64%) of all measles deaths
since 2009, suggesting many genotypes are no longer circulating were estimated to have occurred in the African Region,
• Pathophysiology: persistence of measles virus RNA for 2–3 months after rash onset although measles mortality declined by 85% in this
could contribute to the life-long immunity and prolonged state of immune region from 2000 to 2015.9 A quarter of global measles
suppression following measles deaths occurred in the South East Asia region, with India
• Immune responses: development of so-called immune amnesia after measles, in which accounting for two-thirds of these deaths.9 Measles
measles virus-specific lymphocytes replace the established memory cell repertoire, is a vaccination was estimated to have prevented 20·3 million
recent hypothesis to explain the immune suppression that follows measles deaths during this period.9 This decline in measles
• Clinical presentation, complications, and outcomes: the incidence of subacute mortality was a key factor in progress toward achieving
sclerosing panencephalitis might be higher than previously estimated, particularly the Millennium Development Goal 4 to reduce child
when measles is acquired in early childhood mortality.11
• Diagnosis: point-of-care diagnostic tests that detect measles virus-specific IgM
antibodies in blood or oral fluid might allow earlier response to outbreaks Epidemiology
• Prevention: Global measles vaccine coverage has stagnated at 85% for almost a The epidemiology of measles is largely determined by
decade but microneedle patches might revolutionise measles vaccine delivery the respiratory mode of transmission, high
• Elimination and eradication: the Region of the Americas was the first WHO region to contagiousness and lifelong immunity that follows
be declared to have eliminated measles in September, 2016, but the 2015 global infection or vaccination. Measles has thus served as a
measles milestones were not met model of an acute, immunising infection for studies of
infectious disease dynamics,12,13 driven by contact

Rhazes Francis Home Henry Koplik Joseph Licensure of Start of the Measles and Measles elimination goal
distinguishes transmits describes spots Goldberger and first attenuated Expanded Rubella for European and Eastern
smallpox and measles on the buccal John Anderson measles vaccine Programme on Initiative Mediterranean regions
measles through blood, mucosa with show measles is Immunization launched
Gavi, the Vaccine Alliance
analogous to measles caused by a
commits support for
variolation filterable virus
measles and rubella
vaccines
Certification of measles
elimination in the Americas
September, 2016;
rubella declared eliminated
in 2015

9th century 1676 1757 1846 1896 1905 1911 1954 1963 1968 1974 1987 2001 2012 2015 2020

Peter Panum
investigates Maurice
measles Hilleman
outbreak on further
Thomas Faroe Islands Ludvig Hektoen attenuates Cuba conducts
Sydenham and describes transmits measles first mass Measles Measles
provides first incubation measles John Enders vaccine; rubella measles elimination elimination
detailed clinical period and experimentally and Thomas vaccine vaccination goal for the goal for Africa
description of lifelong and studies Peebles isolate introduced in catch-up Western Pacific and South East
measles immunity effects measles virus 1969 campaign region Asia Regions

Figure 1: Measles timeline

2 www.thelancet.com Published online June 30, 2016 http://dx.doi.org/10.1016/S0140-6736(17)31463-0


Seminar

A MCV1 coverage in infants, 2015

<50% (four countries or 2%) ≥90% (119 countries or 61%)


50–79% (38 countries or 20%) Not available
80–89% (33 countries or 17%)

B Global measles incidence by WHO region, 2000–15 C Estimated annual number of global measles deaths, 2000–15
1000·0 Goal EMR SEAR 1·5 Estimated number of measles deaths
Measles incidence per million population (log scale)

AFR EUR WPR 95% upper confidence limit


Annual number of measles deaths (millions)

AMR 95% lower confidence limit

100·0
1·0

10·0
2015 measles incidence goal
5·0
0·5
1·0

0·1 0
06

08
09

06

08
09
04

04
00

00
02
03

05

02
03

05
14
07

14
07
01

10

01

10
12
13

15

12
13

15
11

11
20

20
20
20

20

20
20

20
20

20

20

20
20

20

20

20
20
20

20

20
20

20
20

20
20

20
20

20
20

20

20
20

Year Year

Figure 2: Progress toward achieving global measles milestones for measles vaccine coverage (A), measles incidence (B), and measles mortality (C)
(A) Milestone 1: increase routine coverage with the first dose of measles-containing vaccine (MCV1) for children aged 1 year to ≥90% nationally and ≥80% in every
district. Progress: The number of countries with ≥90% MCV1 coverage increased from 84 (44%) in 2000 to 119 (61%) in 2015.9 Among countries with ≥90% MCV1
coverage nationally, the percentage with ≥80% coverage in every district was only 39% of 119 countries in 2015. (B) Milestone 2: reduce global measles incidence to
less than 5 cases per 1 million population. Progress: reported global annual measles incidence decreased 75% from 2000 to 2015, but only the Region of the Americas
achieved the milestone of less than 5 cases per 1 million population.9 (C) Milestone 3: reduce global measles mortality by 95% from the 2000 estimate. Progress: the
number of estimated global annual measles deaths decreased 79% from 2000 to 2015.9 EMR=Eastern Mediterranean Region. SEAR=South-East Asia Region.
AFR=African Region. EUR=European Region. WPR=Western Pacific Region. AMR=Region of the Americas. Reproduced from WHO,9 with permission from the WHO
and the US Centers for Disease Control and Prevention.

patterns between susceptible and infectious individuals from 11·8 days to 13·2 days.18 The longest reported
and affected by birth rates (introducing new susceptible incubation period for measles was 23 days.19 The
individuals), heterogeneities in vaccine coverage, and infectious period begins several days before and lasts
human mobility.14 Measles virus is most often for several days after the onset of rash, coinciding with
transmitted by respiratory droplets over short distances, peak levels of viraemia and when cough and coryza are
but also by small particle aerosols that remain most intense, facilitating transmission. However,
suspended in the air for up to 2 h (figure 4).15,16 The precise measurements of the duration of infectiousness
incubation period for measles is about 10 days from the are difficult and require detailed contact histories.
time of infection to the onset of fever and 14 days to the Measles virus RNA can be detected for several months
onset of rash, although these frequently cited estimates in blood, urine, and nasopharyngeal specimens after
represent a log-normal distribution of incubation rash onset.20 Although the infectious period is unlikely
periods.17 A systematic review based on 55 observations to be this long, measles cases are reported with no
from eight observational studies estimated the median known source despite intense contact investigation,21
incubation period from infection to the onset of signs raising the possibility of rare, prolonged infectious
and symptoms to be 12·5 days, with a 95% CI extending periods.

www.thelancet.com Published online June 30, 2016 http://dx.doi.org/10.1016/S0140-6736(17)31463-0 3


Seminar

100
patterns facilitating transmission (eg, congregation of
MCV1 coverage
MCV2 coverage
children at school) and environmental factors favouring
90
the viability and transmission of measles virus.26 Measles
80
outbreaks in the tropics have more variable seasonal
70
patterns and, in regions with high birth rates, highly
MCV coverage* (%)

60
irregular, large measles outbreaks can occur.27
50 Passively acquired, maternal anti-measles virus IgG
40 antibodies protect young infants against measles in the
30 first months of life but can also interfere with vaccine
20 responses by neutralising vaccine virus.28 Because
10 antibody levels are generally higher in women with
0 naturally acquired immunity, infants born to women
with vaccine-induced immunity become susceptible to
80

82

84

86

88

90

92

94

96

98

06

08
00

02

04

10

12

14
20
20

20
20
19

19

20

20
19

19

20

20
19

19
19

19

19

19

measles at a younger age than those born to women with


Year
a history of wildtype measles virus infection.29,30 The
Figure 3: Global measles vaccine coverage for the first (MCV1) and second (MCV2) doses average age of measles cases is a function of the rate of
Reproduced from WHO, by permission of WHO. MCV=measles-containing vaccine. *Coverage as estimated by decline of protective maternal antibodies, the age at
WHO and UNICEF.
which children acquire protective immunity from
The high contagiousness of measles virus is expressed vaccination, and the rate of contact between susceptible
by the basic reproductive number (R0), which is the and infectious individuals. In densely populated urban
average number of secondary cases resulting from the settings with low measles vaccine coverage, the average
introduction of an infectious individual into a completely age of infection is low and measles is a disease of
susceptible population.22 A function of pathogen infants and young children. As measles vaccine coverage
transmission characteristics, population density, and increases, or the rate of contact between susceptible and
social contact patterns, the basic reproductive number of infectious individuals decreases, the age distribution
measles virus has been estimated to be 9–18 in different shifts toward older children. With increasing vaccination
settings (figure 4).23 Measles has one of the highest basic coverage and levels of population immunity, the age
reproductive numbers for a directly transmitted pathogen, distribution of measles is further shifted into adolescence
significantly higher than that for smallpox (R0=5–7) or and adulthood. Following recent progress in increasing
influenza (R0=2–3) viruses. This epidemiological measles vaccine coverage, many countries now face a
characteristic of measles is the major obstacle to changing epidemiological profile in which a higher
elimination as the virus spreads rapidly in susceptible proportion of measles cases occur in adolescents and
populations and requires high levels of population adults, albeit with a lower number of measles cases.31
immunity to interrupt transmission. A simple analytical In addition to the changing age distribution of measles
estimate, assuming random mixing of individuals, is that cases, measles outbreaks have been increasingly
levels of population immunity as high as 89–94% are recognised within populations with immunity gaps
required to achieve measles elimination. However, these despite high overall vaccine coverage, including cases in
estimates do not account for spatial heterogeneities in individuals who received two doses of measles vaccine.32,33
susceptibility and non-random contact patterns.24 For example, during a measles outbreak at a high school
Measles virus can only be maintained in human in Quebec, Canada, in 2011, measles cases were identified
populations by unbroken chains of transmission. Measles among students who received two doses of measles
does not result in known latent or persistent infectious vaccine, with those who received the first dose before
states and no animal reservoirs maintain virus 15 months of age at greatest risk of infection.34,35
transmission, features that make eradication possible.5
Non-human primates can be infected with measles virus,25 Virology
but their population size is well below the critical The measles virus is a non-segmented, negative-sense
community size of up to 300 000 to 500 000 individuals to RNA virus and a member of the Morbillivirus genus in
sustain virus transmission.2 the family of Paramyxoviridae. The genome of about
Endemic measles virus transmission has a typical 16 000 nucleotides encodes six structural proteins, the
temporal pattern characterised by annual seasonal nucleoprotein, phosphoprotein, matrix, fusion, haemag­
epidemics superimposed upon longer epidemic cycles of glutinin, and large protein, and two non-structural
2–5 years, resulting from the accumulation of susceptible proteins V and C encoded within the phosphoprotein
people over successive birth cohorts and the subsequent gene. The haemagglutinin protein is one of two trans­
decline in the number of susceptible individuals following membrane glycoproteins on the surface of the virion and
an outbreak (figure 4).26 Annual measles outbreaks binds to cellular receptors, including the signalling
typically occur in the late winter and early spring in lymphocyte activation molecule (SLAM or CD150) on
temperate climates, determined in part by social contact lymphocytes, monocytes, macrophages, and dendritic

4 www.thelancet.com Published online June 30, 2016 http://dx.doi.org/10.1016/S0140-6736(17)31463-0


Seminar

A Epidemiology
40 Measles can have regular temporal patterns, driven by
35 the accumulation and decline of susceptible individuals,
Number of measles cases
notified (in thousands)

and cluster spatially among susceptible populations


30
25
20
15
10
5
0
1950 1952 1954 1956 1958 1960 1962 1964 1966 1968 1970 1972 1974 1976 1978

B Transmission
Measles virus is transmitted A single person with Measles virus spreads first
by respiratory droplets and measles infects 9–18 to local lymphoid tissue
aerosolised particles other people on average and is then disseminated
throughout the blood
stream through infected
lymphocytes, infecting cells
F protein in almost all organ systems
H protein

The incubation period for


measles is 12·5 days on
average (95% CI 11·8–13·2
days), with a range up to
23 days

C Disease course D Complications


Fever
Temperature (°C)

40
39 Neurological:
Keratoconjunctivitis
ADEM, MIBE, SSPE
38 (blindness)
37 Otitis media
Stomatitis
Laryngitis (croup)
Rash
Pneumonia

Koplik’s spots

Conjunctivitis Adverse pregnancy


outcomes Diarrhoea

Coryza

Death

Cough

1 2 3 4 5 6 7 8 9 10

Incubation Prodromal Rash phase Convalescent


phase phase phase

Figure 4: Measles epidemiology (A), transmission (B),disease course (C), and complications (D)
Part A adapted from Fine PE and Clarkson JA.26 ADEM=acute demyelinating encephalomyelitis. MIBE=measles inclusion body encephalitis. SSPE=subacute sclerosing
panencephalitis.

cells,36 and nectin-4, a component of epithelial cell membrane, enabling entry of viral ribonucleoproteins
adherens junctions.37 The distribution of these receptors into the cell cytoplasm.39
determines the broad cell types and tissues infected with Measles virus can be genetically characterised by
measles virus. The lifelong immunity that follows sequencing a stretch of 450 basepairs that code for a
measles is due to neutralising IgG antibodies to the variable region of the nucleoprotein gene. 24 genotype
haemagglutinin protein that block binding to host cell reference strains are recognised by WHO.40 Genetic
receptors.38 The fusion protein, the second viral characterisation of circulating wildtype measles virus is
glycoprotein exposed on the viral surface, is responsible important in documenting transmission pathways,
for fusion of the viral envelope with the host cell distinguishing endemic from imported strains, and

www.thelancet.com Published online June 30, 2016 http://dx.doi.org/10.1016/S0140-6736(17)31463-0 5


Seminar

verifying measles elimination by demonstrating the Measles virus RNA remained detectable in lymphoid
absence of endemic viral strains.41 Genotyping can also tissue after it was no longer detectable in blood.55
differentiate vaccine from wildtype measles virus, which is
important in assessing vaccine-associated adverse events.42 Immune responses
Only 13 of the 24 known genotypes were detected between Immune responses to measles virus are crucial for viral
2005 and 2014 and only eight since 2009, suggesting that clearance and the establishment of protective immunity
many genotypes are no longer circulating.43 Recent but also are the pathological basis of the clinical signs
evidence suggests detailed measles virus transmission and symptoms that contribute to measles morbidity
networks can be identified by sequencing larger segments and mortality. The rash of measles, for example, is
of the measles virus genome, such as the haemagglutinin characterised histologically by perivascular, lymphocytic
and phosphoprotein genes, or sequencing the whole viral infiltrates.56 The earliest, innate immune responses occur
genome through Sanger or next generation sequencing during the prodromal phase before the onset of rash. Two
techniques.44,45 non-structural viral proteins (V and C) suppress host
An important characteristic of measles virus is that it interferon production, facilitating virus replication.57 The
is an antigenically monotypic virus, despite its genotypic adaptive immune response follows and consists of cellular
diversity and the fact that RNA viruses have high and humoral responses, which are essential for recovery
mutation rates.46 Consequently, attenuated measles and the establishment of long-term, protective immunity,
vaccines derived from a single measles virus genotype respectively. The initial humoral response consists of IgM
isolated in the 1950s,47 including the Schwarz and antibodies that arise at the time of the rash and persist for
Moraten measles vaccine strains, remain protective 6–8 weeks. Detection of IgM antibodies by enzyme
worldwide. Thus, new measles vaccines do not need to immunoassay is the most commonly used laboratory
be developed to counter evolving measles virus strains method of confirming measles virus infection, but IgM
because the neutralising epitopes on the haemagglutinin antibodies might not be detectable early in the disease
protein that confer protection are highly conserved, course shortly after rash onset.58 Subsequently, IgG
probably because of functional constraints on the amino antibodies are produced, the most abundant of which are
acid sequence and tertiary structure of the surface against the nucleoprotein. The efficacy of antibodies alone
proteins.48 in preventing measles is shown by the protection
conferred by passively acquired maternal antibodies and
Pathophysiology post-exposure administration of antimeasles virus
Measles virus acquired through respiratory droplets immune globulin.59 Cellular immune responses to
or aerosolised particles initially infects lymphocytes, measles virus are important for viral clearance and
dendritic cells, and alveolar macrophages in the respiratory recovery, as shown by the fact that children with agamma­
tract.49,50 The virus replicates and spreads during the globulinaemia recover from measles, but children with
incubation period, first to local lymphoid tissue and is T-cell deficiencies develop severe or fatal disease.60 The
then disseminated throughout the blood stream by importance of cellular immune responses for viral
infected lymphocytes, infecting epithelial and endothelial clearance were further shown in macaque models.61,62
cells primarily through direct transmission across cells51 Plasma interferon-γ levels, consistent with a predominant
in almost all organ systems.52 Infected dendritic cells and Th1 immune response, are increased during the
lymphocytes transfer measles virus to epithelial cells of acute phase of infection.63 During convalescence, a Th2
the respiratory tract using the nectin-4 receptor.53 Measles response promotes the development of protective measles
virus buds from the apical surface of respiratory epithelial virus-specific antibodies and is characterised by high
cells or is shed through damaged epithelium, enabling concentrations of interleukin 4, interleukin 10, and
respiratory transmission to susceptible hosts.54 interleukin 13.64
Although the infectious period for measles extends Measles was the first immunosuppressive infection to
from several days before to several days after start of the be described.65 Deficiencies of both innate and adaptive
rash, measles virus RNA can be detected in clinical immune responses can render individuals with measles
samples for at least 3 months after rash onset.20 Recent more susceptible to secondary bacterial and viral
studies of measles pathogenesis further challenge the infections.66 Transient lymphopenia occurs in the blood
traditional view that measles is an acute infection of during measles67 but is likely due to the redistribution of
2–3 weeks duration. For instance, measles virus lymphocytes from peripheral blood to lymphatic tissues.68
nucleoprotein RNA was detected in peripheral blood Functional abnormalities of immune cells have been
mononuclear cells for up to 67 days in an experimental described, including decreased lymphocyte proliferative
macaque model, with clearance of measles virus RNA responses and impaired dendritic cell function ex vivo,69
occurring in three phases.55 Measles virus RNA declined but it is not clear that these mechanisms are responsible
rapidly as infectious virus was cleared, followed by a for immune suppression in vivo.70 The Th2 response
rebound in measles virus RNA by as much as ten-fold during convalescence might inhibit Th1 responses,
that slowly declined to undetectable levels over 10 weeks.55 increasing susceptibility to intracellular pathogens.

6 www.thelancet.com Published online June 30, 2016 http://dx.doi.org/10.1016/S0140-6736(17)31463-0


Seminar

Plasma interleukin 10 concentrations are increased for Complications of measles can affect most organ
weeks in children with measles and also might contribute systems and are most common in young infants, adults
to immune suppression.64 A more recent hypothesis is older than 20 years, pregnant women, and those who are
that measles virus infection results in a proliferation of immunocompromised or undernourished, particularly
measles virus-specific lymphocytes that replace the children with vitamin A deficiency (figure 4).82 The
established memory cell repertoire, resulting in so-called respiratory tract is a frequent site of complication, with
immune amnesia and susceptibility to previously pneumonia accounting for most measles-associated
encountered antigens, including vaccine antigens.70 morbidity and mortality.83 Pneumonia is most often
Immune suppression and the associated increased risk caused by secondary viral or bacterial pathogens but can
of secondary infections are thought to last several weeks be due to measles virus itself resulting in Hecht’s giant
to months following measles.71,72 However, a recent cell pneumonia.84 Bacterial and viral pathogens associated
intriguing study suggested this state of increased risk with pneumonia in children with measles are not well
could extend for as long as 2–3 years after measles.73 characterised, particularly in children vaccinated against
Stimulated by research on immune amnesia and the pneumococcus and Haemophilus influenzae type b. Other
potential loss of immunological memory to previously complications of the respiratory tract include laryngo­
encountered antigens, this study using population-level tracheobronchitis (croup) and otitis media. Diarrhoea
data from high-income countries found that non-measles can result in considerable morbidity and mortality, and
infectious disease mortality lagged measles incidence by is often due to secondary infections with bacteria
2–3 years, a finding that is consistent with, but does not or protozoa. Keratoconjunctivitis, another serious
in itself prove, the immune amnesia hypothesis.73 complication of measles, was a frequent cause of
Another intriguing but controversial observation is the blindness before the widespread use of measles vaccine
potential for non-specific beneficial effects of measles and vitamin A supplementation.85 Measles in pregnancy
vaccination, such as reduced mortality from other is associated with an increased risk of low birthweight,
infectious diseases.74,75 This phenomenon is part of a larger spontaneous abortion, intrauterine fetal death, and
debate on potential non-specific clinical and immunological maternal death.86
effects of vaccines, both positive and negative.76,77 Three rare but serious CNS complications of measles
were a major motivating factor to prevent infection
Clinical presentation, complications, through vaccination in countries where case fatality was
and outcomes low (figure 4). First, acute disseminated encephalomyelitis
Measles is an acute febrile illness associated with a (ADEM) is a demyelinating autoimmune disease that is
characteristic erythematous, maculopapular rash triggered by measles virus and occurs within days to weeks
(figure 4). The illness begins with fever and typically at in approximately one in 1000 cases. ADEM is characterised
least one of the three “Cs”: cough, coryza, and by fever, seizures, and other neurological deficits.87 Second,
conjunctivitis. Koplik’s spots appear on the buccal mucosa measles inclusion body encephalitis (MIBE) is a
as small white papules and provide an opportunity to progressive measles virus infection of the brain that results
clinically diagnose measles a day or two before the rash. in neurological deterioration and death in individuals with
The rash appears 3–4 days after the onset of fever, first on impaired cellular immunity within months of the acute
the face and behind the ears, and then spreads to the illness.87 MIBE has been described in children who are
trunk and extremities, coinciding with development of the immunosuppressed following organ transplants and in
adaptive immune response. The fever and catarrhal HIV-infected persons.88 Third, subacute sclerosing
symptoms typically peak with the rash, which persists for panencephalitis (SSPE) is a delayed complication of
3–4 days. The rash might be minimal in children with measles that occurs in about 1:10 000 to 1:100 000 cases
vaccine-modified measles who have previous immunity 5–10 years after the acute illness, caused by the host
following vaccination, and these children might not have response to production of mutated virions with defective
cough, coryza, or conjunctivitis.78 Malnourished children assembly and budding.87 SSPE most often occurs in people
can develop a deeply pigmented rash that desquamates infected with measles virus before 2 years of age and is
during recovery.79 As the rash represents a perivascular characterised by seizures, progressive deterioration of
lymphocytic infiltration, children with impaired cellular cognitive and motor function, and death.87 A recent report
immunity, such as those infected with HIV, might not of SSPE in the USA identified a much higher incidence
develop the characteristic rash or the rash might be than previously described, including an incidence of
delayed.80 Recovery typically occurs within 1 week of rash 1:1367 cases in children who acquired measles younger
onset in people with uncomplicated measles. The measles than 5 years of age and 1:609 cases in children with measles
case definition, consisting of a generalised maculopapular before 1 year of age.89 Measles vaccination reduces the
rash, fever (≥38·3oC) and either cough, coryza, or incidence of SSPE.90
conjunctivitis, has high sensitivity (75–90%) but a low Measles case fatality ratios range from less than one in
positive predictive value when measles incidence is low, 1000 cases to 5% in endemic areas in sub-Saharan Africa
highlighting the need for serological confirmation.81 and Asia, to as high as 20–30% in refugees and internally

www.thelancet.com Published online June 30, 2016 http://dx.doi.org/10.1016/S0140-6736(17)31463-0 7


Seminar

displaced populations.91 This variation is determined by confirmed by detection of viral RNA through RT-PCR
the average age of infection, nutritional and immunological using throat, nasal, nasopharyngeal, and urine samples
status of the population, measles vaccine coverage, and before measles virus-specific IgM antibodies are
access to health care. detectable.
Recent advances in diagnostic technologies could
Diagnosis facilitate rapid outbreak detection and response. IgM and
Measles is readily recognised by clinicians familiar IgG antibodies to measles virus, as well as measles virus
with the disease in people presenting with fever and RNA, can be detected in oral fluid or in serum eluted
generalised rash, particularly during outbreaks or in from dried blood spots. The use of oral fluid samples
patients with a history of travel to endemic areas. Other might increase participation in serological surveys in
acute viral infections that might be confused with some communities, although this approach comes with
measles include infection with rubella virus, human loss of sensitivity.95 Dried blood spots facilitate
herpes virus type 6, parvovirus B19, and dengue viruses. uncomplicated specimen transport and storage.96 Point-
The medical history and physical examination should of-care diagnostic tests that detect measles virus-specific
focus on the clinical features of measles as well as IgM and IgG antibodies in blood and oral fluid have been
potential complications, including pneumonia, otitis developed and validated, but are not yet widely
media, keratoconjunctivitis, and diarrhoea. Assessment available.97,98
of nutritional and immune status, most importantly
vitamin A deficiency and HIV infection, will identify Management
individuals at highest risk of mortality. Health-care The management of patients with measles consists of
personnel should take appropriate measures, including supportive therapy to correct or prevent dehydration
prompt isolation of infectious cases using airborne and nutritional deficiencies, prompt recognition and
precautions, to prevent transmission within health-care treatment of secondary bacterial infections, and provision
settings.92 The clinical diagnosis of measles is more of vitamin A. WHO recommends administration of once
challenging to clinicians unfamiliar with the disease, daily doses of 200 000 IU of vitamin A for 2 consecutive
before the onset of rash, in immunocompromised and days to all children with measles older than 1 year of age.99
undernourished children in whom the rash might be In younger children, lower doses are recommended,
absent or altered, and in individuals with pre-existing specifically 100 000 IU per day for children 6–12 months
antibodies from maternal immunity, immune globulin, of age and 50  000 IU per day for children younger
or previous vaccination who can have a longer incubation than 6 months.99 For children with clinical evidence of
period, milder prodromal illness, and a less apparent vitamin A deficiency, a third dose is recommended
rash than typical cases. 2–4 weeks later.99 Two doses of vitamin A, but not a single
The most common laboratory method for confirming dose, has been associated with a reduction in the risk of
measles virus infection is detection of measles virus- mortality in children younger than 2 years (risk ratio 0·18
specific IgM antibodies in serum or plasma. However, [95% CI 0·03–0·61]) and a reduction in the risk of
measles virus-specific IgM antibodies might be low or pneumonia-specific mortality (0·33 [0·08–0·92]).100 No
undetectable until 4 days or more after rash onset, specific antiviral therapies exist for measles, although
resulting in false negative results if samples are collected ribavirin, interferon alfa, and other antiviral drugs have
early.58 About 75% of people with measles will have been used to treat severe measles.101 Evidence supporting
detectable measles virus-specific IgM antibodies within the use of prophylactic antibiotics for children with
the first 72 h after rash and almost all people with measles measles is limited and such use is not recommended,102
will have detectable measles virus-specific IgM antibodies but antibiotics are indicated for people with measles who
after 4 days.93 Measles virus-specific IgM antibodies peak have clinical evidence of bacterial infection, including
within 1–3 weeks after the onset of rash and decline to pneumonia and otitis media.
undetectable levels within 4–8 weeks. Acute infection
also can be confirmed serologically by measuring a four- Prevention
fold or greater increase in measles virus-specific IgG Measles is best prevented through measles vaccination.
antibody levels between acute and convalescent sera. Currently licensed measles vaccines are attenuated viral
Commercially available enzyme immunoassays are most vaccines that replicate within the host to induce
often used to detect antibodies to measles virus, although protective immunity.99 Measles vaccines can be
the gold standard test with the highest sensitivity is the administered as combined vaccines with those for
plaque reduction neutralisation assay.94 Individuals who rubella (MR), mumps (MMR), or varicella (MMR-V).
are seronegative by enzyme immunoassay might have Use of combined measles–rubella vaccines provides an
detectable antibodies to measles virus by neutralisation opportunity to eliminate rubella and congenital rubella
assay. The presence of IgG antibodies to measles virus in syndrome, and are increasingly used throughout the
a single serum specimen is evidence of previous infection world through the support of Gavi, the Vaccine Alliance.
or immunisation. Measles virus infection also can be Attenuation is achieved by passaging wildtype measles

8 www.thelancet.com Published online June 30, 2016 http://dx.doi.org/10.1016/S0140-6736(17)31463-0


Seminar

viruses through repeated culture in non-human cells. the routine immunisation system at 15–18 months of
The first measles vaccine (Edmonston B strain) was age with a minimum of 4 weeks between doses. In
licensed in the USA in 1963, but caused mild measles countries with low level measles virus transmission
with fever and rash, and was administered with gamma where MCV1 is administered at 12 months of age or
globulin (figure 1). Further attenuated measles vaccine older, MCV2 can be administered at 15–18 months of age
strains were licensed in the late 1960s, including the or at school entry.
Schwarz and Moraten vaccine viruses that are still widely Estimated global coverage with the first dose of MCV1
used. Two previously licensed measles vaccines were increased from 72% to 85% from 2000 to 2010 but
subsequently withdrawn after they were shown to have plateaued at 85% in 2015 (figure 3).9 Vaccinating these
serious adverse effects. The first was a formalin- remaining 15% of children to raise global MCV1
inactivated measles vaccine, also licensed in the early coverage above 85% should be the primary goal of
1960s, that led to the formation of immune complexes103 measles control and elimination programmes. Among
and atypical measles, including pneumonitis and a the 20·8 million children who did not receive MCV1 in
maculopapular or petechial rash that started on the 2015, 53% lived in six countries: India, Nigeria, Pakistan,
wrists and ankles, upon exposure to wildtype virus.104 Indonesia, Ethiopia, and the Democratic Republic of
The second was a high-titre measles vaccine Congo.9 An increasing number of countries provide
recommended for use by WHO in 1989 that contained a MCV2 through routine immunisation services but
higher concentration of measles virus to overcome the estimated global MCV2 coverage remains suboptimal, at
inhibitory effect of maternal antibodies but resulted in only 61% in 2015.9 Measles SIAs continue to be used to
unexplained delayed mortality in girls.105 achieve high levels of population immunity and
WHO recommends that the first dose of measles- 184 million people received MCV through SIAs in 2015.9
containing vaccine (MCV1) be administered at 9 months However, SIAs are expensive and resource-intensive,
of age in settings with endemic measles but as early as require careful planning, monitoring, and evaluation,
6 months of age in some circumstances, including and coverage is often lower than planned.
during outbreaks, for internally displaced populations
and refugees, for HIV-infected and exposed children, Measles elimination and eradication
and children at high risk of contracting measles, but The World Health Assembly established three global
allows flexibility based on local epidemiology.99 The milestones for measles control to be achieved by 2015
proportions of children who develop protective levels of (figure 2).43 The first addressed routine measles
antibody after measles vaccination are about 85% at vaccination coverage, with the goal of 90% or higher
9 months of age and 95% at 12 months of age.106 When coverage nationally and 80% or higher in every district
measles vaccine is given to children younger than
9 months of age, a lower proportion develop protective
immunity because of the inhibitory effect of maternal Panel 2: Definitions109
antibodies and immaturity of the immune system.107 • Measles eradication: worldwide interruption of measles
Administering the first dose of measles vaccine at virus transmission in the presence of a surveillance system
12–15 months results in a higher proportion of protected that has been verified to be performing well
children, but can only be done in settings where the risk • Measles elimination: the absence of endemic measles
of measles is low. Children who are infected with HIV virus transmission in a defined geographical area for more
should be revaccinated against measles following than 12 months in the presence of a well performing
immune reconstitution with highly active antiretroviral surveillance system
therapy because of failure to maintain protective • Endemic measles transmission: the existence of
antibody levels.108 continuous transmission of indigenous or imported
The high levels of population immunity necessary to measles virus that persists for more than 12 months in
interrupt measles virus transmission cannot be achieved any defined geographical area
with a single dose schedule. A second dose of measles • Re-establishment of endemic transmission: occurs when
vaccine should be provided to immunise those children epidemiological and laboratory evidence indicates the
who failed to respond to the first dose. MCV1 is usually presence of a chain of transmission of a virus strain that
given through routine immunisation services but two continues uninterrupted for more than 12 months in a
strategies have been used to administer additional doses defined geographical area where measles had previously
of measles vaccine, the first through routine been eliminated
immunisation services (MCV2) and the second • Measles outbreak in countries with an elimination goal:
through mass vaccination campaigns (sometimes when two or more confirmed cases are temporally related
called supplemental immunisation activities [SIAs]) that (with dates of rash onset occurring between 7 and 21 days
typically target children from 9 months to 5 years or apart) and are epidemiologically or virologically linked,
15 years of age. In countries where MCV1 is administered or both
at 9 months of age, MCV2 can be administered through

www.thelancet.com Published online June 30, 2016 http://dx.doi.org/10.1016/S0140-6736(17)31463-0 9


Seminar

meets many of the biological criteria for disease


Panel 3: Research needs eradication, including the absence of a non-human
Research will be crucial to achieving regional measles elimination goals and global reservoir, accurate diagnosis, and the availability of a
eradication. Research is needed on how best to strengthen routine immunisation services highly effective vaccine.5
to improve access and increase demand for vaccination; how best to increase measles
vaccine coverage and identify immunity gaps to achieve high, equitable coverage; and The future of measles
how best to improve measles surveillance to rapidly identify and respond to outbreaks Great progress has been made in reducing measles
and better measure progress towards elimination. incidence, morbidity, and mortality through the
widespread use of measles vaccines (figure 2). Mortality
Strengthening routine immunisation services110 has been reduced from more than 2 million deaths year
• Develop better methods to collect valid and timely vaccine coverage data111 before the widespread use of measles vaccines to slightly
• Develop strategies to address vaccine hesitancy112,113 more than 100 000 in 2015. Global measles vaccine
• Build upon lessons learned from the Global Polio Eradication Initiative114 coverage is high and more countries are introducing
Achieving and sustaining high measles vaccine coverage a second dose of measles vaccine through routine
• Develop and evaluate better measles vaccines that do not require a cold chain and can immunisation services. With the support of Gavi, many
be delivered without needles, such as microneedle patches115,116 countries are introducing combined measles–rubella
• Evaluate the optimal use of serological surveys to guide immunisation programmes117 vaccines, providing the opportunity to eliminate rubella
and congenital rubella syndrome. New technologies,
Improve measles surveillance such as microneedle patches, could revolutionise
• Develop more accurate and comprehensive surveillance systems to track progress measles vaccination strategies. Yet all is not well. MCV1
toward measles mortality reduction and elimination31 coverage has stagnated at about 85% for almost a decade.
• Develop and evaluate point-of-care measles and rubella IgM antibody tests, with the Anti-vaccine sentiments are increasingly voiced, in part
capacity to perform viral genotyping97 because of successful measles control. Estimates of
• Develop better analytical approaches to predicting measles outbreaks through human measles incidence, deaths, and vaccine coverage are
mobility patterns and spatial heterogeneities in susceptibility118,119 often based on poor quality data, hindering the ability to
track progress and target interventions. The Measles and
Rubella Global Strategic Plan 2012–2020 Mid-Term
(although this coverage level is insufficient for Review identified deficiencies in implementing current
elimination). The second addressed measles incidence, strategies, largely because of inadequate country
with the goal of fewer than five cases per 1 million ownership, global political will, and resources.121 Urgent
people. And the third addressed measles deaths, with efforts are needed to improve measles surveillance and
the goal to reduce measles mortality by 95% or higher vaccine coverage estimates and increase global coverage
from 2000 estimates. Subsequently, the Global Vaccine with two doses of measles vaccine through advocacy,
Action Plan for 2012–20 established targets to eliminate education, and the strengthening of routine
measles and rubella in five WHO regions by 2020 and, immunisation systems. Building on the polio legacy, the
as of September, 2013, all six WHO regions adopted political will and financial resources must be garnered to
measles elimination goals by 2020 or earlier achieve the regional elimination goals and eventual
(figure 1; panel 2).43 The Region of the Americas was the global measles eradication.
first WHO region to be declared to have eliminated Declaration of interests
measles after a rigorous verification process by the WJM is a member of WHO’s Strategic Advisory Group of Experts on
International Expert Committee for Documenting and Immunization Working Group on Measles and Rubella. However, the
views expressed in this Seminar reflect those of the author and are not
Verifying Measles, Rubella, and Congenital Rubella necessarily those of WHO or the Strategic Advisory Group of Experts on
Syndrome in September, 2016.9 However, in view of the Immunization Working Group on Measles and Rubella.
slow progress, the WHO Strategic Advisory Group of Acknowledgments
Experts on Immunization concluded that neither the The author is grateful to the anonymous reviewers for their thoughtful
2015 global measles milestones nor the measles comments and suggestions.
elimination goals were achieved.108 Identifying, References
prioritising, funding, and answering critical research 1 Nambulli S, Sharp CR, Acciardo AS, Drexler JF, Duprex WP.
Mapping the evolutionary trajectories of morbilliviruses: what,
questions will be necessary to achieve these goals where and whither. Curr Opin Virol 2016; 16: 95–105.
(panel 3). 2 Black FL. Measles endemicity in insular populations: critical
Countries or regions that have eliminated measles are community size and its evolutionary implication. J Theor Biol 1966;
11: 207–11.
at continual risk of imported measles as long as measles
3 Keeling MJ, Grenfell BT. Disease extinction and community size:
virus is circulating in other parts of the world, a risk that modeling the persistence of measles. Science 1997; 275: 65–67.
is increased by global travel. Outbreaks result in high 4 Meeting of the International Task Force for Disease Eradication,
costs related to case-based investigations, outbreak November 2015. Weekly Epidemiol Rec 2016; 91: 61–71.
5 Moss WJ, Strebel P. Biological feasibility of measles eradication.
responses, and provision of health care.120 This risk will J Infect Dis 2011; 204: S47–53.
only be mitigated if measles is eradicated. Measles virus

10 www.thelancet.com Published online June 30, 2016 http://dx.doi.org/10.1016/S0140-6736(17)31463-0


Seminar

6 Moss WJ, Griffin DE. Measles. Lancet 2012; 379: 153–64. 32 Sugerman DE, Barskey AE, Delea MG, et al. Measles outbreak in a
7 Shanks GD, Waller M, Briem H, Gottfredsson M. Age-specific highly vaccinated population, San Diego, 2008: role of the
measles mortality during the late 19th-early 20th centuries. intentionally undervaccinated. Pediatrics 2010; 125: 747–55.
Epidemiol Infect 2015; 143: 3434–41. 33 Phadke VK, Bednarczyk RA, Salmon DA, Omer SB.
8 Mulders MN, Rota PA, Icenogle JP, et al. Global Measles and Association between vaccine refusal and vaccine-preventable
Rubella Laboratory Network Support for Elimination Goals, diseases in the United States: A review of measles and pertussis.
2010–2015. MMWR Morb Mortal Wkly Rep 2016; 65: 438–42. JAMA 2016; 315: 1149–58.
9 Patel MK, Gacic-Dobo M, Strebel PM, et al. Progress toward 34 De Serres G, Boulianne N, Defay F, et al. Higher risk of measles
regional measles elimination—worldwide, 2000–2015. MMWR when the first dose of a 2-dose schedule of measles vaccine is given
Morb Mortal Wkly Rep 2016; 65: 1228–33. at 12-14 months versus 15 months of age. Clin Infect Dis 2012;
10 Simons E, Ferrari M, Fricks J, et al. Assessment of the 2010 global 55: 394–402.
measles mortality reduction goal: results from a model of 35 Defay F, De Serres G, Skowronski DM, et al. Measles in children
surveillance data. Lancet 2012; 379: 2173–78. vaccinated with 2 doses of MMR. Pediatrics 2013; 132: e1126–33.
11 Liu L, Oza S, Hogan D, et al. Global, regional, and national causes 36 Tatsuo H, Ono N, Tanaka K, Yanagi Y. SLAM (CDw150) is a cellular
of child mortality in 2000–13, with projections to inform post-2015 receptor for measles virus. Nature 2000; 406: 893–97.
priorities: an updated systematic analysis. Lancet 2015; 385: 430–40. 37 Muhlebach MD, Mateo M, Sinn PL, et al. Adherens junction protein
12 McLean AR, Anderson RM. Measles in developing countries. nectin-4 is the epithelial receptor for measles virus. Nature 2011;
Part I. Epidemiological parameters and patterns. Epidemiol Infect 480: 530–53.
1988; 100: 111–33. 38 Tahara M, Ohno S, Sakai K, et al. The receptor-binding site of the
13 McLean AR, Anderson RM. Measles in developing countries. measles virus hemagglutinin protein itself constitutes a conserved
Part II. The predicted impact of mass vaccination. Epidemiol Infect neutralizing epitope. J Virol 2013; 87: 3583–86.
1988; 100: 419–42. 39 Plattet P, Alves L, Herren M, Aguilar HC. Measles virus fusion
14 Ferrari MJ, Grenfell BT, Strebel PM. Think globally, act locally: protein: structure, function and inhibition. Viruses 2016; 8: 112.
the role of local demographics and vaccination coverage in the 40 WHO. Measles virus nomenclature update: 2012.
dynamic response of measles infection to control. Wkly Epidemiol Rec 2012; 87: 73–81.
Phill Trans Royal Soc London B 2013; 368: 20120141. 41 Mulders MN, Truong AT, Muller CP. Monitoring of measles
15 Remington PL, Hall WN, Davis IH, Herald A, Gunn RA. elimination using molecular epidemiology. Vaccine 2001;
Airborne transmission of measles in a physician’s office. JAMA 19: 2245–49.
1985; 253: 1574–77. 42 Moss WJ, Scott S, Ndhlovu Z, et al. Suppression of human
16 Hope K, Boyd R, Conaty S, Maywood P. Measles transmission in immunodeficiency virus type 1 viral load during acute measles.
health care waiting rooms: implications for public health Pediatr Infect Dis J 2009; 28: 63–65.
response. West Pacific Surv Resp J 2012; 3: 33–38. 43 Perry RT, Murray JS, Gacic-Dobo M, et al. Progress toward regional
17 Sartwell PE. The incubation period and the dynamics of infectious measles elimination—worldwide, 2000–2014. MMWR Morb Mortal
disease. Am J Epidemiol 1966; 83: 204–06. Wkly Rep 2015; 64: 1246–51.
18 Lessler J, Reich NG, Brookmeyer R, Perl TM, Nelson KE, 44 Penedos AR, Myers R, Hadef B, Aladin F, Brown KE.
Cummings DA. Incubation periods of acute respiratory viral Assessment of the utility of whole genome sequencing of measles
infections: a systematic review. Lancet Infect Dis 2009; 9: 291–300. virus in the characterisation of outbreaks. PloS One 2015;
19 Fitzgerald TL, Durrheim DN, Merritt TD, Birch C, Tran T. 10: e0143081.
Measles with a possible 23 day incubation period. 45 Gardy JL, Naus M, Amlani A, et al. Whole-genome sequencing of
Commun Dis Intel Quart Report 2012; 36: E277–80. measles virus genotypes H1 and D8 during outbreaks of infection
20 Riddell MA, Moss WJ, Hauer D, Monze M, Griffin DE. following the 2010 Olympic Winter Games reveals viral
Slow clearance of measles virus RNA after acute infection. transmission routes. J Infect Dis 2015; 212: 1574–78.
J Clin Virol 2007; 39: 312–17. 46 Beaty SM, Lee B. Constraints on the genetic and antigenic
21 Fill MM, Sweat D, Morrow H, et al. Notes from the field: measles variability of measles virus. Viruses 2016; 8: 109.
outbreak of unknown source - Shelby County, Tennessee, 47 Enders JF, Peebles TC. Propagation in tissue cultures of
April–May 2016. MMWR Morb Mortal Wkly Rep 2016; 65: 1039–40. cytopathogenic agents from patients with measles.
22 Dietz K. The estimation of the basic reproduction number for Proc Soc Exp Biol Med 1954; 86: 277–86.
infectious diseases. Stat Method Med Res 1993; 2: 23–41. 48 Fulton BO, Sachs D, Beaty SM, et al. Mutational analysis of measles
23 Anderson R MR. Infectious Diseases of Humans. Oxford: virus suggests constraints on antigenic variation of the
Oxford University Press, 1991. glycoproteins. Cell Reports 2015; 11: 1331–38.
24 Wallinga J, Heijne JC, Kretzschmar M. A measles epidemic 49 de Swart RL, Ludlow M, de Witte L, et al. Predominant infection of
threshold in a highly vaccinated population. PLoS Med 2005; CD150+ lymphocytes and dendritic cells during measles virus
2: e316. infection of macaques. PLoS Pathog 2007; 3: e178.
25 Jones-Engel L, Engel GA, Schillaci MA, et al. 50 Ludlow M, de Vries RD, Lemon K, et al. Infection of lymphoid tissues
Considering human-primate transmission of measles virus in the macaque upper respiratory tract contributes to the emergence
through the prism of risk analysis. Am J Primatol 2006; 68: 868–79. of transmissible measles virus. J Gen Virol 2013; 94: 1933–44.
26 Fine PE, Clarkson JA. Measles in England and Wales—I: 51 Singh BK, Li N, Mark AC, Mateo M, Cattaneo R, Sinn PL.
An analysis of factors underlying seasonal patterns. Int J Epidemiol Cell-to-cell contact and nectin-4 govern spread of measles virus
1982; 11: 5–14. from primary human myeloid cells to primary human airway
27 Ferrari MJ, Grais RF, Bharti N, et al. The dynamics of measles in epithelial cells. J Virol 2016; 90: 6808–17.
sub-Saharan Africa. Nature 2008; 451: 679–84. 52 Ludlow M, McQuaid S, Milner D, de Swart RL, Duprex WP.
28 Albrecht P, Ennis FA, Saltzman EJ, Krugman S. Persistence of Pathological consequences of systemic measles virus infection.
maternal antibody in infants beyond 12 months: mechanism of J Pathol 2015; 235: 253–65.
measles vaccine failure. J Pediatr 1977; 91: 715–18. 53 Leonard VH, Sinn PL, Hodge G, et al. Measles virus blind to its
29 Leuridan E, Hens N, Hutse V, Ieven M, Aerts M, Van Damme P. epithelial cell receptor remains virulent in rhesus monkeys but
Early waning of maternal measles antibodies in era of measles cannot cross the airway epithelium and is not shed. J Clin Invest
elimination: longitudinal study. BMJ 2010; 340: 1626. 2008; 118: 2448–58.
30 Waaijenborg S, Hahne SJ, Mollema L, et al. Waning of maternal 54 de Vries RD, Mesman AW, Geijtenbeek TB, Duprex WP, de Swart RL.
antibodies against measles, mumps, rubella, and varicella in The pathogenesis of measles. Curr Opin Virol 2012; 2: 248–55.
communities with contrasting vaccination coverage. J Infect Dis 55 Lin WH, Kouyos RD, Adams RJ, Grenfell BT, Griffin DE.
2013; 208: 10–16. Prolonged persistence of measles virus RNA is characteristic of
31 Durrheim DN, Crowcroft NS, Strebel PM. Measles—the epidemiology primary infection dynamics. Proc Natl Acad Sci USA 2012;
of elimination. Vaccine 2014; 32: 6880–83. 109: 14989–94.

www.thelancet.com Published online June 30, 2016 http://dx.doi.org/10.1016/S0140-6736(17)31463-0 11


Seminar

56 Kimura A, Tosaka K, Nakao T. Measles rash. I. Light and electron 81 Hutchins SS, Papania MJ, Amler R, et al. Evaluation of the measles
microscopic study of skin eruptions. Arch Virol 1975; 47: 295–307. clinical case definition. J Infect Dis 2004; 189: S153–59.
57 Nakatsu Y, Takeda M, Ohno S, Shirogane Y, Iwasaki M, Yanagi Y. 82 Stevens GA, Bennett JE, Hennocq Q, et al. Trends and mortality
Measles virus circumvents the host interferon response by different effects of vitamin A deficiency in children in 138 low-income and
actions of the C and V proteins. J Virol 2008; 82: 8296–306. middle-income countries between 1991 and 2013: a pooled analysis
58 Bellini WJ, Helfand RF. The challenges and strategies for laboratory of population-based surveys. Lancet Glob Health 2015; 3: e528–36.
diagnosis of measles in an international setting. J Infect Dis 2003; 83 Perry RT, Halsey NA. The clinical significance of measles: a review.
187: S283–90. J Infect Dis 2004; 189: S4–16.
59 Young MK, Nimmo GR, Cripps AW, Jones MA. Post-exposure 84 Enders JF, Mc CK, Mitus A, Cheatham WJ. Isolation of measles
passive immunisation for preventing measles. virus at autopsy in cases of giant-cell pneumonia without rash.
Cochrane Database Syst Rev 2014: 4: Cd010056. N Engl J Med 1959; 261: 875–81.
60 Good RA, Zak SJ. Disturbances in gamma globulin synthesis as 85 Foster A, Sommer A. Childhood blindness from corneal ulceration
experiments of nature. Pediatrics 1956; 18: 109–49. in Africa: causes, prevention, and treatment.
61 Permar SR, Klumpp SA, Mansfield KG, et al. Role of CD8(+) Bull World Health Organ 1986; 64: 619–23.
lymphocytes in control and clearance of measles virus infection of 86 Ogbuanu IU, Zeko S, Chu SY, et al. Maternal, fetal, and neonatal
rhesus monkeys. J Virol 2003; 77: 4396–400. outcomes associated with measles during pregnancy: Namibia,
62 Permar SR, Klumpp SA, Mansfield KG, et al. Limited contribution 2009–2010. Clin Infect Dis 2014; 58: 1086–92.
of humoral immunity to the clearance of measles viremia in rhesus 87 Griffin DE. Measles virus and the nervous system. Handb Clin Neurol
monkeys. J Infect Dis 2004; 190: 998–1005. 2014; 123: 577–90.
63 Griffin DE, Ward BJ, Jauregui E, Johnson RT, Vaisberg A. 88 Hardie DR, Albertyn C, Heckmann JM, Smuts HE.
Immune activation during measles: interferon-gamma and Molecular characterisation of virus in the brains of patients with
neopterin in plasma and cerebrospinal fluid in complicated and measles inclusion body encephalitis (MIBE). Virol J 2013; 10: 283.
uncomplicated disease. J Infect Dis 1990; 161: 449–53. 89 Wendorf KA, Winter, K, Zipprich J, et al. Subacute sclerosing
64 Moss WJ, Ryon JJ, Monze M, Griffin DE. Differential regulation of panencephalitis: the devastating measles complication is more
interleukin (IL)-4, IL-5, and IL-10 during measles in Zambian common than we think. Clin Infect Dis 2017; published online
children. J Infect Dis 2002; 186: 879–87. April 6. DOI:10.1093/cid/cix302.
65 von Pirquet C. Das Verhalten der kutanen Tuberkulin-reaktion 90 Campbell H, Andrews N, Brown KE, Miller E. Review of the effect
während der Masern. Dtsch Med Wochenschr 1908; 34: 1297–300. of measles vaccination on the epidemiology of SSPE. Int J Epidemiol
66 Griffin DE. Measles virus-induced suppression of immune 2007; 36: 1334–48.
responses. Immunol Rev 2010; 236: 176–89. 91 Wolfson LJ, Grais RF, Luquero FJ, Birmingham ME, Strebel PM.
67 Ryon JJ, Moss WJ, Monze M, Griffin DE. Functional and Estimates of measles case fatality ratios: a comprehensive review of
phenotypic changes in circulating lymphocytes from hospitalized community-based studies. Int J Epidemiol 2009; 38: 192–205.
Zambian children with measles. Clin Diagn Lab Immunol 2002; 92 Maltezou HC, Wicker S. Measles in health-care settings.
9: 994–1003. Am J Infect Cont 2013; 41: 661–63.
68 de Vries RD, McQuaid S, van Amerongen G, et al. Measles 93 Helfand RF, Heath JL, Anderson LJ, Maes EF, Guris D, Bellini WJ.
immune suppression: lessons from the macaque model. Diagnosis of measles with an IgM capture EIA: the optimal timing of
PLoS Pathog 2012; 8: e1002885. specimen collection after rash onset. J Infect Dis 1997; 175: 195–99.
69 Abt M, Gassert E, Schneider-Schaulies S. Measles virus modulates 94 Cohen BJ, Doblas D, Andrews N. Comparison of plaque reduction
chemokine release and chemotactic responses of dendritic cells. neutralisation test (PRNT) and measles virus-specific IgG ELISA for
J Gen Virol 2009; 90: 909–14. assessing immunogenicity of measles vaccination. Vaccine 2008;
70 de Vries RD, de Swart RL. Measles immune suppression: 26: 6392–97.
functional impairment or numbers game? PLoS Pathog 2014; 95 Hayford KT, Al-Emran HM, Moss WJ, Shomik MS, Bishai D,
10: e1004482. Levine OS. Validation of an anti-measles virus-specific IgG assay
71 Tamashiro VG, Perez HH, Griffin DE. Prospective study of the with oral fluid samples for immunization surveillance in
magnitude and duration of changes in tuberculin reactivity during Bangladesh. J Virol Meth 2013; 193: 512–18.
uncomplicated and complicated measles. Pediatr Infect Dis J 1987; 96 Uzicanin A, Lubega I, Nanuynja M, et al. Dried blood spots on filter
6: 451–54. paper as an alternative specimen for measles diagnostics: detection
72 Akramuzzaman SM, Cutts FT, Wheeler JG, Hossain MJ. of measles immunoglobulin M antibody by a commercial enzyme
Increased childhood morbidity after measles is short-term in urban immunoassay. J Infect Dis 2011; 204: S564–69.
Bangladesh. Am J Epidemiol 2000; 151: 723–35. 97 Warrener L, Slibinskas R, Chua KB, et al. A point-of-care test for
73 Mina MJ, Metcalf CJ, de Swart RL, Osterhaus AD, Grenfell BT. measles diagnosis: detection of measles-specific IgM antibodies and
Long-term measles-induced immunomodulation increases overall viral nucleic acid. Bull World Health Organ 2011; 89: 675–82.
childhood infectious disease mortality. Science 2015; 348: 694–99. 98 Shonhai A, Warrener L, Mangwanya D, et al. Investigation of a
74 Aaby P. Assumptions and contradictions in measles and measles measles outbreak in Zimbabwe, 2010: potential of a point of care
immunization research: is measles good for something? test to replace laboratory confirmation of suspected cases.
Soc Sci Med 1995; 41: 673–86. Epidemiol Infect 2015; 143: 3442–50.
75 Do VA, Biering-Sorensen S, Fisker AB, et al. Effect of an early dose 99 WHO. Measles vaccines: WHO position paper—April 2017. Wkly
of measles vaccine on morbidity between 18 weeks and 9 months Epidemiol Rec 2017; 92: 205–27.
of age: a randomized, controlled trial in Guinea-Bissau. J Infect Dis 100 Huiming Y, Chaomin W, Meng M. Vitamin A for treating measles
2017; published online Jan 10. DOI:10.1093/infdis/jiw512. in children. Cochrane Database Syst Rev 2005; 4: Cd001479.
76 Higgins JP, Soares-Weiser K, Lopez-Lopez JA, et al. Association of 101 Barnard DL. Inhibitors of measles virus. Antiv Chem Chemother
BCG, DTP, and measles containing vaccines with childhood 2004; 15: 111–19.
mortality: systematic review. BMJ 2016; 355: i5170. 102 Kabra SK, Lodha R. Antibiotics for preventing complications in
77 Kandasamy R, Voysey M, McQuaid F, et al. Non-specific children with measles. Cochrane Database Syst Rev 2013;
immunological effects of selected routine childhood 8: Cd001477.
immunisations: systematic review. BMJ 2016; 355: i5225. 103 Polack FP, Hoffman SJ, Crujeiras G, Griffin DE. A role for
78 Choe YJ, Hu JK, Song KM, et al. Evaluation of an expanded case nonprotective complement-fixing antibodies with low avidity for
definition for vaccine-modified measles in a school outbreak in measles virus in atypical measles. Nat Med 2003; 9: 1209–13.
South Korea in 2010. Jpn J Infect Dis 2012; 65: 371–75. 104 Fulginiti VA, Eller JJ, Downie AW, Kempe CH. Altered reactivity to
79 Morley D. Severe measles in the tropics. I. BMJ 1969; 1: 297–300. measles virus. Atypical measles in children previously immunized
80 Moss WJ, Cutts F, Griffin DE. Implications of the human with inactivated measles virus vaccines. JAMA 1967; 202: 1075–80.
immunodeficiency virus epidemic for control and eradication of 105 WHO. Expanded programme on immunization (EPI). Safety of
measles. Clin Infect Dis 1999; 29: 106–12. high titre measles vaccines. Wkly Epidemiol Rev 1992; 67: 357–61.

12 www.thelancet.com Published online June 30, 2016 http://dx.doi.org/10.1016/S0140-6736(17)31463-0


Seminar

106 Moss WJ, Scott S. The immunological basis for immunization 114 Cochi SL, Freeman A, Guirguis S, Jafari H, Aylward B. Global polio
serie. Module 7: Measles—Update 2009. Geneva: World Health eradication initiative: lessons learned and legacy. J Infect Dis 2014;
Organization, 2009. 210: S540–46.
107 Gans HA, Arvin AM, Galinus J, Logan L, DeHovitz R, Maldonado Y. 115 Edens C, Collins ML, Ayers J, Rota PA, Prausnitz MR.
Deficiency of the humoral immune response to measles vaccine in Measles vaccination using a microneedle patch. Vaccine 2013;
infants immunized at age 6 months. JAMA 1998; 280: 527–32. 31: 3403–09.
108 WHO. Meeting of the Strategic Advisory Group of Experts on 116 Edens C, Collins ML, Goodson JL, Rota PA, Prausnitz MR.
immunization, October 2016—conclusions and recommendations. A microneedle patch containing measles vaccine is immunogenic
Wkly Epidemiol Rec 2016; 91: 561–68. in non-human primates. Vaccine 2015; 33: 4712–18.
109 WHO. Framework for verifying elimination of measles and rubella. 117 Metcalf CJ, Farrar J, Cutts FT, et al. Use of serological surveys to
Wkly Epidemiol Rev 2013; 88: 89–99. generate key insights into the changing global landscape of
110 Orenstein WA, Seib K. Beyond vertical and horizontal programs: infectious disease. Lancet 2016; 388: 728–30
a diagonal approach to building national immunization programs 118 Bogoch, II, Brady OJ, Kraemer MU, et al. Anticipating the
through measles elimination. Expert Rev Vaccine 2016; 15: 791–93. international spread of Zika virus from Brazil. Lancet 2016;
111 Cutts FT, Izurieta HS, Rhoda DA. Measuring coverage in MNCH: 387: 335–36.
design, implementation, and interpretation challenges associated 119 Takahashi S, Metcalf CJ, Ferrari MJ, et al. Reduced vaccination and
with tracking vaccination coverage using household surveys. the risk of measles and other childhood infections post-Ebola.
PLoS Med 2013; 10: e1001404. Science 2015; 347: 1240–42.
112 Schuster M, Eskola J, Duclos P. Review of vaccine hesitancy: 120 Ortega-Sanchez IR, Vijayaraghavan M, Barskey AE, Wallace GS.
Rationale, remit and methods. Vaccine 2015; 33: 4157–60. The economic burden of sixteen measles outbreaks on United States
113 Jarrett C, Wilson R, O’Leary M, Eckersberger E, Larson HJ. public health departments in 2011. Vaccine 2014; 32: 1311–17.
Strategies for addressing vaccine hesitancy—a systematic review. 121 Orenstein WA HA, Nkowane B, Olive JM, Reingold A. Measles and
Vaccine 2015; 33: 4180–90. Rubella Global Strategic Plan 2012–2020. Mid-term review, 2016.

www.thelancet.com Published online June 30, 2016 http://dx.doi.org/10.1016/S0140-6736(17)31463-0 13

You might also like