You are on page 1of 18

PRIMER

Severe childhood malnutrition


Zulfiqar A. Bhutta1,2, James A. Berkley3–5, Robert H. J. Bandsma1,4,6, Marko Kerac7,
Indi Trehan8–10 and André Briend11
Abstract | The main forms of childhood malnutrition occur predominantly in children <5 years of age
living in low-income and middle-income countries and include stunting, wasting and kwashiorkor,
of which severe wasting and kwashiorkor are commonly referred to as severe acute malnutrition.
Here, we use the term ‘severe malnutrition’ to describe these conditions to better reflect the
contributions of chronic poverty, poor living conditions with pervasive deficits in sanitation and
hygiene, a high prevalence of infectious diseases and environmental insults, food insecurity, poor
maternal and fetal nutritional status and suboptimal nutritional intake in infancy and early childhood.
Children with severe malnutrition have an increased risk of serious illness and death, primarily from
acute infectious diseases. International growth standards are used for the diagnosis of severe
malnutrition and provide therapeutic end points. The early detection of severe wasting and
kwashiorkor and outpatient therapy for these conditions using ready-to-use therapeutic foods form
the cornerstone of modern therapy, and only a small percentage of children require inpatient care.
However, the normalization of physiological and metabolic functions in children with malnutrition
is challenging, and children remain at high risk of relapse and death. Further research is urgently
needed to improve our understanding of the pathophysiology of severe malnutrition, especially the
mechanisms causing kwashiorkor, and to develop new interventions for prevention and treatment.

Tackling malnutrition is a major global health ­priority known as nutritional oedema). In this Primer, we focus
that is relevant to several of the United Nations’ on and use the composite term ‘severe malnutrition’
Sustainable Development Goals (SDGs) and is high­ to refer to severe wasting and kwashiorkor and to better
lighted directly in Goal 2 (‘Zero hunger’), which aims reflect the longstanding nature of the combined infectious
to “end hunger, achieve food security and improved and environmental insults that can occur in such cases.
nutrition and promote sustainable agriculture1”. Several It is important to emphasize the multi­factorial aetiology
forms of malnutrition are recognized, including stunt­ of severe malnutrition and its strong association with
ing, which is characterized by reduced linear growth, mortality2, as well as the frequent co­existence of different
wasting (including moderate wasting and severe wast­ types of malnutrition in the same child over time3 and
ing, also known as marasmus), which is characterized that such concurrence further exacerbates mortality4.
by low body tissue mass and other physiological abnor­ Indeed, most children with severe wasting or kwashior­
malities (FIG. 1), and kwashiorkor, which is characterized kor are also stunted5–9. In addition, we concentrate on the
by diffuse peripheral oedema. The current classification ­largest and most vulnerable group, children <5 years of
of malnutrition is based on body size or the presence of age, reflecting SDGs Goal 2.2, which aims to “…by 2030
oedema, which does not indicate the aetiology or pre­ end all forms of malnutrition, including achieving by 2025
cise nutritional deficits in an individual. Accordingly, the inter­nationally agreed targets on stunting and wasting
this classification can effectively screen and identify in c­ hildren under five years of age…1”. Other populations
Correspondence to Z.A.B.
Centre for Global Child
mal­nutrition, but it does not address each and every can be malnourished, such as pregnant women (which
Health, Hospital for Sick specific nutrient deficiency a child might have or the bio­ has implications for fetal growth)10, elderly individ­
Children, Peter Gilgan Centre logical variability among children. Such differences have uals11,12 and those with disabilities13,14, but discussing these
for Research & Learning, no effect on current empirical management strategies populations is beyond the scope of this Primer.
686 Bay Street, Toronto,
that aim to address the predominant macronutrient and
Ontario, M5G 0A4, Canada.
zulfiqar.bhutta@sickkids.ca micronutrient deficiencies and treat possible infections. Epidemiology
The term ‘severe acute malnutrition’ has replaced Regions with a high prevalence of severe malnutrition
Article number: 17067
doi:10.1038/nrdp.2017.67 ‘protein–­energy malnutrition’, which was used to describe often also have high childhood mortality, with under­lying
Published online 21 Sep 2017 children with severe wasting and kwashiorkor (also malnutrition having a major role in the risk of death.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17067 | 1


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Author addresses during which figures were uncertain and HIV was less
prevalent. This influence is especially relevant for Sub-
1
Centre for Global Child Health, Hospital for Sick Children, Saharan Africa, where HIV and severe malnutrition are
Peter Gilgan Centre for Research & Learning, 686 Bay strongly linked22.
Street, Toronto, Ontario, M5G 0A4, Canada. The 2015 Global Burden of Disease Study also
2
Center of Excellence in Women and Child Health,
reported that 174,000 deaths among children <5 years of
Aga Khan University, Karachi, Pakistan.
3
Clinical Research Department, KEMRI/Wellcome Trust age were directly due to protein–energy ­malnutrition25.
Research Programme, Kilifi, Kenya. Indeed, malnutrition, especially severe malnutrition,
4
The Childhood Acute Illness & Nutrition (CHAIN) Network, is associated with a high risk of death. The latest Lancet
Nairobi, Kenya. Nutrition series (published in 2013) estimated that
5
Nuffield Department of Medicine, University of Oxford, 875,000 deaths were attributable to wasting (which was
Oxford, UK. the cause of death of 12.6% of children <5 years of age),
6
Department of Biomedical Sciences, College of Medicine, and 516,000 deaths were related to severe wasting (which
University of Malawi, Blantyre, Malawi. was the cause of death of 7.4% of children <5 years of
7
Department of Population Health, London School of age)2. In ten longitudinal studies including ~54,000
Hygiene & Tropical Medicine, London, UK.
child-years of follow-up and 1,300 deaths in children
8
Lao Friends Hospital for Children, Luang Prabang, Laos.
9
Department of Pediatrics, Washington University in <5 years of age, all degrees of stunting, wasting and
St. Louis, St. Louis, Missouri, USA. underweight were associated with an increased risk of
10
Department of Paediatrics and Child Health, University death4. Mortality is higher in children with both wasting
of Malawi, Blantyre, Malawi. and stunting (who have a mortality hazard ratio (HR)
11
Department of Nutrition, Exercise and Sports, Faculty of of 12.3) than in those who have either wasting or stunt­
Science, University of Copenhagen, Copenhagen, Denmark. ing (a mortality HR of 2.3 for those with wasting only)4.
The causes of death in individuals with mal­nutrition
include infectious diseases (such as diarrhoeal dis­
The global estimates of the prevalence of severe mal­ eases, pneumonia, measles and malaria) and metabolic
nutrition vary between agencies and according to the disturb­ances (such as hypoglycaemia and refeeding syn­
methodology used (BOX 1). drome (see Management))26,27. TABLE 1 summarizes the
Commonly used figures are from the WHO, UNICEF known HRs for deaths due to major childhood infectious
and World Bank Group Interagency estimates, which ­diseases associated with various types of malnutrition2.
provide both levels and trends for child growth and mal­
nutrition15 and are based on standard anthropometric Mechanisms/pathophysiology

indices. An estimated 155 million children <5 years of Determinants of severe malnutrition
age were stunted in 2016, and 52 million were wasted, The loss of muscle and fat tissue that characterizes wast­
of whom 17 million were severely wasted15. The global ing can be caused by inadequate protein and energy
burden of kwashiorkor remains uncertain due to its wide intake resulting from food insecurity, poor diet and
geographical variability and the failure to include assess­ disease. However, severe malnutrition is rarely caused
ments of oedema in most large nutritional surveys16; by a single factor and generally arises from an inter­
however some estimates from regions of southern and play between social, political and economic factors, the
eastern Africa suggest that kwashiorkor accounts for presence of chronic infections and inflammation (both
50–70% of cases of severe malnutrition5. Geographical in the gut and systemically). In some circumstances,
variations in the prevalence of severe malnutrition have gender issues, such as a lack of female empowerment,
been observed (FIG. 2), which probably reflects differ­ are important drivers of malnutrition28. Children with
ences in the distribution, causes and effects of multiple severe malnutrition are common in conditions of
risk factors for malnutrition. These risk factors include popu­lation displacement, conflict and food shortage29,
social and environmental factors (such as poverty, poor which worsens the effects of many of the risk factors
education, limited health care access and contaminated for malnutrition and are associated with ineffective
environments)17–20 and living in areas with a high b
­ urden remedial strategies.
of infectious diseases, such as respiratory tract infec­
tions, diarrhoeal diseases21, HIV and turberculosis19,22. Underlying mechanisms
Other risks include dietary factors, such as acute and Since the initial descriptions of severe malnutrition, stud­
chronic food insecurity23, as occurs during famines, ies aiming to understand the mechanisms and organ-spe­
as well as suboptimal breastfeeding and suboptimal cific and metabolic pathophysiology of severe weight
­complementary feeding practices24. deficits and oedema have been carried out. Historical
The other major global metrics initiative, the comparisons are difficult given the changing definitions
Global Burden of Disease study, estimates the n ­ umber of malnutrition over time30 and the wide range of clinical
of deaths and disabilities directly related to protein– manifestations that reflects different pathologies.
energy malnutrition. The 2015 Global Burden of
Disease Study reported a global decline in the preva­ Wasting. Our knowledge of the mechanisms and meta­
lence of severe malnutrition from 25.4 million in 1990 bolic changes associated with wasting comes mainly
to 22.4 million in 2015 (REF. 25). However, 1990 might from the literature on long-term starvation and cach­
have represented a peak compared with previous years, exia (that is, wasting induced by a chronic illness)31.

2 | ARTICLE NUMBER 17067 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

During short-term starvation (that is, up to several days Kwashiorkor. Despite longstanding knowledge of
of fasting), free fatty acids (FFAs) and ketone bodies are kwashiorkor, the underlying pathophysiological mech­
primar­ily oxidized using available fat stores from adipose anisms are poorly understood. In her earliest report,
tissue, and myofibrillar proteins can be broken down Cecily Williams documented that children with kwash­
into amino acids, which can be converted into glucose iorkor in Ghana were fed mostly a monotonous corn diet
(through gluconeogenesis). After several days of starv­ that was deficient in essential amino acids such as lysine
ation (when body fat has been depleted), myo­fibrillar and tryptophan35. However, few studies have identified
proteins are extensively broken down to maintain essen­ any specific nutritional deficiencies associated with the
tial metabolic processes. The short-term regulation of development of kwashiorkor, and studies typically have
macronutrient oxidation and synthesis depends on insu­ not revealed major differences in food group intake
lin and glucagon, whereas the long-term regulation of between children who developed kwashiorkor compared
these processes is mediated by other hormones, such as with those who did not or with those who developed
growth hormone, thyroid hormones, catecholamines wasting36,37. Despite numerous hypotheses, the aetio­
and corticosteroids. logy of oedema, which is the hallmark of ­kwashiorkor,
In addition, cytokine release in cachexia, especially remains undefined. In animal models, some features
the release of tumour necrosis factor (TNF), IL-1 and of kwashiorkor, such as hypoalbuminaemia, can be
IL-6, can negatively influence body composition through induced by a diet low in protein and high in mono­
a reduction in appetite and food intake, and direct cata­ saccharides and disaccharides38,39, but oedema is rarely
bolic effects on skeletal muscle and adipose tissue32,33. observed. In addition, the degree of hypoalbuminaemia
Increased activation of the ubiquitin–­proteasome path­ and recovery upon nutritional management in children
way is the main process that degrades myo­fibrillar pro­ with kwashiorkor correlates poorly with the degree of
teins in cachectic conditions31,34. Autophagy has also oedema or the speed of its resolution40. Thus, controv­
been implicated in muscle wasting31 and on­going auto­ ersy remains regarding the contribution of factors other
phagy can be detrimental for muscle cells by removing than hypoalbuminaemia to the development of oedema
cellular components important for muscle metabolism in individuals with kwashiorkor38,41.
and contraction, such as mitochondria. However, the
specific roles of inflammation, proteasomes and auto­ Infections. Children with severe malnutrition are highly
phagy in severe wasting in children in low-income susceptible to life-threatening infections2,42, which is a
and middle-income countries have not been studied consequence of a secondary immunodeficiency. Indeed,
in detail. the presentation of children with severe malnutrition to
medical services might be due to a serious infection
rather than the presence of malnutrition alone. Several
• Lethargy and irritability • Impaired thyroid function potential mechanisms of immune dysfunction exist in
• Long-term effects on • Impaired cortisol and growth individuals with malnutrition43,44.
development? hormone responses
• Decreased appetite
Skin, respiratory and gastrointestinal mucosal ­barrier
integrity are often impaired in children with  mal­
nutrition, and the effects of this impairment are com­
Respiratory tract pounded by chronic subclinical enteric dysfunction in
infections close associ­ation with alterations to gut microbiota45,46.
Impaired cardiac function?
Children with severe malnutrition have increased levels
of markers of systemic immune activation, such as the
Impaired hepatic Altered immune pro-inflammatory cytokines TNF, IL-1, IL-6 and IL-12,
synthetic function and function which alter the growth hormone–insulin-like growth
hepatobiliary function factor 1 (IGF1) axis and result in wasting and linear
growth failure. Alterations in this axis are also observed
• Impaired macronutrient in other childhood inflammatory conditions, such as
• Reduced β cell absorption juvenile arthritis and inflammatory bowel disease47.
function • Intestinal infections
• Impaired pancreatic
Systemic immune activation in severe malnutrition can
• Changes to microbiota be caused by acute and chronic infection, inflammatory
exocrine function • Chronic inflammation?
enteropathy, translocation of microbial components
from the gut lumen or mucosa into the circulation48,49
Impaired glomerular and dysregulated immune responses50.
and tubular function? Loss of skin integrity Other alterations in the immune system in severe
malnutrition include T cell dysfunction and reductions
in neutrophil microbicidal activity51, the number of
dendritic cells, antigen priming and presentation52 and
protein levels in the complement cascade44. Thymic
atrophy, T cell hyporesponsiveness and impaired T cell
Figure 1 | Organ system involvement in severe malnutrition. Severe malnutrition
Nature Reviews | Disease Primers prolifer­ation could result from chronic immune activ­
can affect several organ systems. The functional impairments in these systems have ation and/or the metabolic demands of T cells for glu­
been characterized, but the underlying mechanisms have not been fully elucidated. cose, amino acids and nutritionally mediated regulatory

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17067 | 3


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

hormones, such as leptin50,53,54. Although some abnor­ study did not show elevated lipolysis in children with
malities in the immune system can resolve with nutri­ severe malnutrition at hospital admission compared
tional rehabilitation55, the reversal of susceptibility to with those in a state of nutritional recovery, although
infection and reduction in immune activation have not reduced FFA (palmitate) flux and oxidation were noted
been well characterized. in children with kwashiorkor compared with children
with severe wasting63. One caveat of this study is that
Altered metabolism. Severe malnutrition can also lipolysis was assessed when children were in the semi-
affect many aspects of macronutrient metabolism and fasted state, when insulin levels were likely low and
endocrine function56,57 (FIG. 3). In contrast to severe lipolysis was stimulated. Thus, how lipolysis is altered
wast­ing, which is associated with a starvation-induced in the postprandial state is unknown. A recent study
response of subcutaneous fat loss and muscle wasting, identified high acylcarnitine levels in the early period of
kwashiorkor has been speculated to be associated with hospital admission for severe malnutrition regardless
a maladaptive metabolic response; a high-carbohydrate, of the presence of oedema, suggesting preferential fatty
low-protein diet is thought to lead to a continuous glyco­ acid oxidation64.
lytic response and protein catabolism that is inadequate Glucose homeostasis is also disrupted in children
to meet the amino acid requirements necessary to main­ with severe malnutrition; hypoglycaemia is common,
tain essential protein synthesis pathways58. Indeed, pro­ although frequent or continuous glucose measurement
tein breakdown has been shown to be lower in children studies have not yet been carried out to characterize
with kwashiorkor than in children with severe wasting this fully56,65. Kwashiorkor is associated with reduced
and in those in a recovered state59,60. In addition, reduced endo­genous glucose production compared with prod­
concentrations of essential and some nonessential amino uction in children with severe wasting or in healthy
acids have been reported, and these deficits were even children, likely contributing to the development of hypo­
more pronounced in children with kwashiorkor61. glycaemia66. This finding is consistent with the hypoth­
Lipid metabolism is also differentially affected in esis that kwashiorkor is associated with a maladaptive
kwashiorkor and severe wasting. Adipocyte lipolysis is metabolic response58. In addition, glucose clearance
a tightly regulated process, with a central role for insu­ from the blood and uptake by tissues, which is regulated
lin in inhibiting hormone-sensitive lipase. Lipolysis is by insulin, are affected; several studies have reported
stimu­lated during starvation (that is, when insulin levels impaired glucose clearance in both kwashiorkor and
are low)62,63, and lipolysis has been shown to be increased severe wasting67–69. A striking feature that seems to con­
in a small cohort of children with severe malnutrition tribute to impaired glucose clearance is the blunted pan­
compared with a control cohort62. However, another creatic endocrine response, with insulin sensitivity likely
unaffected67. However, despite the reports of impaired
glucose clearance in metabolic studies, hyperglycae­
Box 1 | Estimating the epidemiology of malnutrition mia has not been specifically reported in children with
Global data regarding the epidemiology of malnutrition must be understood with severe malnutrition (likely due to glucose levels usually
reference to methodological challenges and contextual factors. For example, wasting being checked only when patients are symptomatic for
is often highly seasonal, and the number of cases peaks around the time of pre-harvest ­hypoglycaemia), and its prevalence is unknown.
rains, when food is scarce and the burden of infectious diseases increases (for example, The mechanisms of impaired pancreatic function
a higher prevalence of malaria and diarrhoeal diseases). In many instances, the interface in children with severe malnutrition are poorly under­
of food insecurity (that is, a lack of access to a sufficient quantity of affordable, stood, but studies in preclinical models have impli­
nutritious food), population displacement and drought also produce the conditions
cated a more-polarized membrane potential, changes
leading to famine.
In addition, although prevalence estimates from cross-sectional surveys are suitable
in cAMP-dependent protein kinase catalytic subunit-α
for slowly changing conditions such as stunting, they are not suitable for capturing the protein levels and a reduced ability to increase intra­
rapidly changing nature of severe and moderate malnutrition, which have a fairly acute cellular Ca2+ levels in response to glucose70–72. In addi­
onset and rapid resolution. Malnutrition can also recur, which can be unrecorded or tion, no clear data are available regarding the pancreatic
without contact with health services at each episode. Accordingly, incidence is the most glucagon response (which is responsible for stimulat­
informative epidemiological measure of acute malnutrition220, but it is rarely well ing glucose production) in children with severe mal­
documented owing to difficulties in ascertainment221. Addressing this limitation, nutrition. Glucagon concentrations have been reported
prevalence-to-incidence conversion factors are sometimes used to determine the to be either mildly reduced during the acute phase of
incidence of malnutrition, especially to estimate caseloads to plan treatment services222. malnutrition compared with levels at recovery or ele­
Efforts are underway to improve surveillance systems for severe malnutrition.
vated in ­children with severe wasting compared with
The problems with the measurements used to diagnose malnutrition in surveys can also
affect epidemiological estimates. For example, mid-upper arm circumference (MUAC)
controls66,73, but stimulation tests or systematic glucagon
and the presence of oedema, which are both case-defining criteria, are not measured response measurements during hypoglycaemia have not
in many field surveys, resulting in underestimates of prevalence as these criteria do not been carried out.
necessarily overlap with those of wasting, which is defined on the basis of low weight- In general, cortisol levels in children with severe mal­
for-length measures223,224. Even when included, the presence of oedema might be missed nutrition are similar to levels in children without
in rapid surveys undertaken by poorly trained surveyors16. malnutri­tion or are increased, which is likely related to
Finally, infants <6 months of age with malnutrition might be missed in stress and indicates that the hypothalamic–­pituitary–
epidemiological surveys, either by design or because MUAC standards for this age adrenal axis is preserved in children with severe
group have not been established. Whether surveys of malnutrition in children <5 years malnutrition and is probably not responsible for the
of age include infants <6 months of age is not always clear.
development of hypoglycaemia64,74,75. Most studies have

4 | ARTICLE NUMBER 17067 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Prevalence (%)
<5
5–9.9
10–14.9
≥15
No data

Numbers of
individuals affected
(thousands)
0–9.9
10–47
53–253
260–25,980
No data

Figure 2 | Prevalence of wasting. The prevalence of wasting as a percentage of the population (part a) and the number
of individuals (thousands; part b). Data from the Global Targets Tracking tool, version 3 (http://www.who.int/nutrition/
Nature Reviews | Disease Primers
trackingtool/en/; May 2017).

shown a reduction in thyroid function in children with intake of antioxidants likely contributes to the lower
severe malnutrition, but the clinical relevance of this levels observed in children with malnutrition, this
remains unclear76,77. In addition, leptin concentrations is also likely, in part, related to a reduced synthesis of
are low in children with severe malnutrition, reflecting certain antioxidants, such as glutathione59. An imbal­
the extent of adipose tissue loss. Leptin has a direct role ance between levels of reactive oxygen species and
in immune function, metabolism and appetite regulation, anti­oxidants has been postulated to have a role in the
and its levels are inversely associated with mortality64. pathophysiology of kwashiorkor80, although a large
randomized trial of an antioxidant mixture (containing
Oxidative stress. Oxidative stress has also been associ­ riboflavin, vitamin E, selenium and N-acetylcysteine) in
ated with severe malnutrition and, in particular, with Malawi failed to show decreased rates of kwashiorkor83,
kwashiorkor. Indeed, children with severe malnutrition despite a pilot study suggesting a potential beneficial
have reduced levels of antioxidants, including vitamin E treatment effect78. An imbalance between reactive oxy­
and glutathione, compared with children without mal­ gen species production and detoxification by peroxi­
nutrition, and this reduction is more-­pronounced in somes results in mitochondrial damage, which ultimately
­children with kwashiorkor78–82. Although a reduced reduces ATP production and impairs cellular function

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17067 | 5


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Table 1 | Anthropometric measures and risk of death in children <5 years of age
All deaths Pneumonia deaths Diarrhoeal deaths Measles deaths Deaths due to other
HR (95% CI) HR (95% CI) HR (95% CI) HR (95% CI) infectious diseases
HR (95% CI)
Height-for-age or length-for-age Z‑score
<–3 5.5 (4.6, 6.5) 6.4 (4.2, 9.8) 6.3 (4.6, 8.7) 6.0 (3.0, 12.0) 3.0 (1.6, 5.8)
–3 to <–2 2.3 (1.9, 2.7) 2.2 (1.4, 3.4) 2.4 (1.7, 3.3) 2.8 (1.4, 5.6) 1.9 (1.0, 3.6)
–2 to <–1 1.5 (1.2, 1.7) 1.6 (1.0, 2.4) 1.7 (1.2, 2.3) 1.3 (0.6, 2.6) 0.9 (0.5, 1.9)
≥–1 (control) 1.0 1.0 1.0 1.0 1.0
Weight-for-length Z‑score
<–3 11.6 (9.8, 13.8) 9.7 (6.1, 15.4) 12.3 (9.2, 16.6) 9.6 (5.1, 18.0) 11.2 (5.9, 21.3)
–3 to <–2 3.4 (2.9, 4.0) 4.7 (3.1, 7.1) 3.4 (2.5, 4.6) 2.6 (1.3, 5.1) 2.7 (1.4, 5.5)
–2 to <–1 1.6 (1.4, 1.9) 1.9 (1.3, 2.8) 1.6 (1.2, 2.1) 1.0 (0.6, 1.9) 1.7 (1.0, 2.8)
≥–1 (control) 1.0 1.0 1.0 1.0 1.0
Adapted from Black et al.2. CI, confidence interval; HR, hazard ratio.

in the liver. Mitochondrial dysfunction and ATP deple­ function, which is mainly responsible for hepatic lipid
tion together with specific nutrient deficiencies might oxidation, is impaired66,84,95,96. Indeed, impaired mito­
influence the response to an intercurrent infection and chondrial function would be expected to affect hepatic
contribute to the development of multi-organ failure84–87; synthetic pathways, and reduced glucose synthesis in
this process needs further characterization. children with kwashiorkor was shown to be correlated
with mitochondrial activity66. Other alterations in the
Cardiac function and haemodynamics. Cohort studies livers of children with kwashiorkor include reduced
of small numbers of children with severe malnutrition albumin synthesis97, although whether other hepatic syn­
have reported cardiac muscle atrophy and decreased thetic processes (such as the production of ­coagulation
cardiac output88, especially in children with kwashior­ factors) are altered has not been studied.
kor89. However, other studies have reported a normal
cardiac output when corrected for body surface area90,91, Enteropathy. Although much interest surrounds the
with impairments observed only in the most severely potential association of stunting with enteropathy98,
ill ­children, which was probably related to sepsis. The intestinal dysfunction also accompanies severe mal­
largest study to date excluded severely ill children and nutrition. Indeed, diarrhoea is common in children
showed no major differences in cardiac function between with malnutrition and is associated with poor clin­
hospitalized children with severe malnutrition and ical outcomes99–101. Several factors might contribute to
­children without malnutrition at an average of 4 days secretory and osmotic diarrhoea in individuals with
after admission91. The mechanisms leading to cardiac malnutrition, including intestinal infections and inflam­
atrophy have not been well studied, although one study mation102. In addition, poor nutrient digestion resulting
used a mouse model of calorie restriction to show that from impaired hepatobiliary and pancreatic exocrine
this was related to a proportional decrease in different function could contribute to nutrient malabsorption
subcellular ­components of cardiomyocytes92. and diarrhoea103–106. Malnutrition leads to small intesti­
nal villous blunting, thereby reducing intestinal absorp­
Hepatic function. Severe malnutrition, in particular tive capacity, including impaired monosaccharide and
kwashiorkor, is associated with changes in hepatic meta­ disaccharide absorption, which could contribute to
bolic function. A striking feature of kwashiorkor is the osmotic diarrhoea107.
presence of hepatic steatosis (that is, fatty liver)35. One Children with severe malnutrition have distinct
study suggested that the hepatic steatosis in kwashiorkor alterations in their intestinal microbiota that can affect
is not related to the impaired secretion of lipids (in the intestinal inflammation and function, as well as the
form of very low-density lipoproteins) by the liver93. growth of the child108. In addition, the faecal microbiota
In addition, an increase in fatty acid release from adi­ of children with kwashiorkor in Malawi was immature,
pose tissue that could be taken up by the liver has been with a reduced diversity, compared with the microbiota
observed in children with kwashiorkor in some62,94, but of their twins without malnutrition109. Interestingly, mice
not all63, studies, which might be related to whether the that received transplantation of the faecal microbiota
analyses were performed when the participants were in from children with kwashiorkor had a more profound
the fasted state. Impaired hepatic lipid oxidation might weight loss than mice transplanted with microbiota
also explain the hepatic steatosis observed in children from the twin without malnutrition, and this weight
with kwashiorkor63. This lipid oxidation is difficult to loss was associ­ated with signs of disturbed metabolic
assess in children, but some data from post-mortem pathways. In a separate birth cohort in Bangladesh, the
samples and animal models suggest that mitochondrial presence of an immature faecal microbiota was linked

6 | ARTICLE NUMBER 17067 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

with wasting110. Another study comparing the micro­ wasting are often apathetic, with slowed movements and
biomes between children with kwashiorkor and children impaired speech. However, the underlying mechanisms
with wasting found a more-modest difference in micro­ of these behavioural changes are poorly understood.
biome composition111. A potential limitation of study­ Although the association of severe malnutrition and
ing the microbiota in children with severe malnutrition growth in early life and development has been well docu­
is the use of antibiotic treatment before stool collection, mented, few studies have focused on long-term develop­
which profoundly affects microbiota composition. mental effects. Impaired development after an episode
Interest is growing regarding the interaction between of severe malnutrition has been reported in ­children116,
the bacterial microbiome, the virome112 and eukaryotic and psychosocial interventions have been shown to
organisms, but the functional consequences of micro­ improve their development117. However, differing case
biota alter­ations in children with severe malnutrition definitions and treatment strategies make it challenging
have yet to be well characterized. to disentangle the direct effects of severe malnutrition
from the effects of other risk factors and adversities that
Renal function. Studies assessing renal function (deter­ these children typically experience. Additional studies
mined by glomerular filtration rate) in children with are needed, particularly to understand the long-term
severe malnutrition have been limited. Given the fre­ effects on different domains of development. In contrast
quency of diarrhoea and dehydration in these ­children, to severe malnutrition, the cognitive deficits associated
the pre-renal contribution to reduced glomerular fil­ with stunting are well described. Indeed, for every 10%
tration might have a substantial role. Low glomeru­ increase in the prevalence of stunting, the proportion of
lar ­filtration rates in children with malnutrition and children reaching the end of primary school has been
dehydra­tion have been reported76, and subsequent stud­ estimated to drop by 7.9%118,119.
ies have reported impaired glomerular filtration rates
and signs of tubular dysfunction with reduced urine Other pathophysiological alterations. Cellular Na+/K+-
osmolality, but the clinical relevance of low glomerular ATPase pumps that maintain fluid, electrolyte and sub­
­filtration remains unclear113. strate levels might be impaired in children with severe
malnutrition. An increased membrane perme­ability was
Brain function. Severe malnutrition is associated found in leukocytes from children with ­kwashiorkor,
with acutely altered cerebral function and behavioural and reduced Na+/K+-ATPase activity has been observed
changes; children with kwashiorkor have cerebral in children with severe wasting, potentially as part of an
­atrophy114,115 and irritability, and children with severe adaptation to reduce energy expenditure120,121.

a Mitochondria malfunction b

↑ROS ↓Glutathione
Liver
↑TGs ? TGs
↓ATP ATP
↓Albumin Albumin
FFAs Acyl-CoA ↓Glucose FFAs Acyl-CoA Glucose

Pancreas

Amino ↓Insulin ↓Insulin


Amino
acids acids

Muscle
Adipose
tissue
Circulation Circulation

FFAs FFAs
Figure 3 | Metabolic changes in severe malnutrition. Reduced secretion of and are used for ATP production and the synthesis of essential proteins and
Nature Reviews | Disease Primers
insulin contributes to a catabolic state in both kwashiorkor (part a) and severe glucose. In kwashiorkor, this adaptive response is disturbed, which causes a
wasting (part b). In severe wasting, the reduced secretion of insulin leads to a reduced release of FFAs from adipose tissue and amino acids from muscle
lipolytic and proteolytic response and the release of free fatty acids (FFAs) and tissue. Mitochondrial damage in the liver is associated with increased reactive
amino acids into the bloodstream. FFAs are taken up by muscle tissue for oxygen species (ROS) production and reduced glutathione. Arrow thickness
oxidation. Both FFAs and amino acids are also partially taken up by the liver represents the amount of metabolites in that pathway. TGs, triglycerides.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17067 | 7


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Diagnosis, screening and prevention the number of standard deviations below or above the
Diagnosis reference median value125. The main growth references
Although case definitions of severe malnutrition for that are used for the diagnosis of severe malnutrition
epidemio­logical, biological and clinical purposes focus are those derived from the 2006 WHO Multi-Country
on anthropometry122 (BOX 2), malnutrition is a functional Growth Reference Study, which describes how ­children
problem and has been defined as a state resulting from ‘should’ grow under optimal health, environmental and
lack of uptake or intake of nutrition leading to altered nutritional conditions126. Severe acute malnutrition is
body composition, decreased body cell mass leading to defined by the WHO as a WHZ of >3 standard deviations
diminished physical and mental function and impaired below the reference value for median weight‑for‑height
clinical outcome from disease123. Anthropometry is used or the presence of bilateral pitting oedema127.
to assess severe malnutrition because the functional Interestingly, a review of all studies on the relation­
­deficits are difficult to measure directly. Anthropometry ship between anthropometric indices and mortality
is therefore a proxy and not a direct measure of malnu­ concluded that WHZs are the least effective predictor
trition. Importantly, no ‘gold standard’ measurement of mortality128 and that unadjusted mid-upper arm cir­
for the diagnosis of severe malnutrition exists. Different cumference (MUAC) was more-discriminatory for
anthropometric measures assess different aspects and assessing mortality in several countries in Africa and
durations of the underlying functional deficit, and some Asia129. The association between MUAC and mortality
relate more closely to body composition and clinical might be due to the preferential selection of c­ hildren
­outcomes than others. who are younger, have both stunting and wasting and
are particularly at risk130. A MUAC of <115 mm was
Anthropometry. The accepted diagnostic c­ riteria for introduced as an independent diagnostic ­criterion for
severe malnutrition have changed over time. Initially, the severe acute malnutrition in children aged 6–59 months
measurement of weight-for-age (that is, comparing the by the WHO in 2013 (REF.  131) (BOX  2) . Although
weight of the child with the weight of age-matched chil­ the use of MUAC for diagnosis is limited as this meas­
dren in a reference population) was used, and severe mal­ urement does not identify the same children with mal­
nutrition was defined as <60% of the reference weight124. nutrition as WHZs122, following the development of
This definition was later refined with the addition of community-­based management programmes that are
oedema as an additional criterion to diagnose kwashi­ effective in preventing malnutrition-­associated deaths,
orkor30. To focus on the highest risk group of the most MUAC has been used more frequently. An alternative
acutely malnourished children rather than on those with is to increase the MUAC cut-off value up to 120 mm to
a low weight-for-age who were mainly stunted, weight- pragmatically define severe malnutrition. Although the
for-height was later used. Weight-for-height compares additional children diagnosed with severe malnutrition
the weight of the child to the weight of well-nourished on the basis of an increased MUAC value are not the
children of the same height (length is used if children are same as those identified by WHZs, this approach can
<2 years old). The WHO recommended expressing meas­ better identify children with a higher risk of death than
urements as weight-for-height Z-scores (WHZs), that is, an approach using both MUAC and a WHZ132.

Box 2 | Anthropometric criteria for nonmicronutrient malnutrition in children 6–59 months of age*


Global acute malnutrition‡ Severe acute malnutrition
Any of the following: • Severe wasting and/or kwashiorkor
• Weight-for-height Z-score <−2 Moderate underweight¶
• Mid-upper arm circumference (MUAC) <125 mm • Weight-for-age Z-score ≥−3 to <−2
• The presence of nutritional oedema
Severe underweight¶
Moderate acute malnutrition‡,§ • Weight-for-age Z-score <−3
Either of the following:
Moderate stunting¶
• Weight-for-height Z-score ≥−3 to <−2
• Height-for-age Z-score ≥−3 to <−2
• MUAC ≥115 mm to <125 mm
in the absence of nutritional oedema Severe stunting¶
• Height-for-age Z-score <−3
Severe wasting‡,||
For younger children, typically those <2 years age, length is commonly
Either of:
measured instead of height, giving weight-for-length or length-for-age
• Weight-for-height Z-score <−3
values. Z-scores refer to the number of standard deviations away from
• MUAC <115 mm a reference population median. For example, a Z-score of −1 equates
in the absence of nutritional oedema to 1 standard deviation below the 2006 WHO Growth Standard (http://
www.who.int/childgrowth/en). *See REF. 131. ‡Acute conditions.
Kwashiorkor‡ §
Diagnosed according to weight-for-length or weight-for-height and/or
• Any weight-for-height Z-score
MUAC criteria; also referred to as ‘moderate wasting.’ ||Diagnosed
• Any MUAC according to weight-for-length or weight-for-height and/or MUAC
• The presence of nutritional oedema criteria. ¶Chronic conditions.

8 | ARTICLE NUMBER 17067 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Identification of oedema. Oedema is a sign of kwash­


iorkor and is assessed by pressing firmly down on the
third to fourth tarsal bones on the dorsal aspect of
the child’s foot for 3–5 seconds and then looking for
pitting oedema for 2–3 seconds. Oedema is graded
according to the location on the body; grade 1+
­indicates oedema limited to the feet and lower legs,
2+ indicates oedema that is present in the arms and
grade 3+ indicates oedema of the face. The presence of
symmetric, bilateral oedema of the feet in a vulnerable
child in a high-risk population, regardless of the child’s
anthropometric measurements, is generally sufficient to
diagnose kwashiorkor.

Infants <6 months of age. The diagnosis of severe mal­


nutrition in infants <6 months of age presents special
challenges. These include the practical difficulties of
undertaking anthropometric measurements on small
infants133 and that many individuals assume mal­nutrition
is rare at this age and so do not take measurements at
all134. However, the burden of malnutrition in this age
group is high135. The WHO currently recommends using
a weight-for-length Z-score (WLZ) of <−3 to identify Figure 4 | Assessment of oedema. Oedematous swelling
infants with severe malnutrition, but only because of Nature
of the lower extremities with skinReviews Disease
changes|in a childPrimers
with
the paucity of evidence for other criteria. Few prospec­ kwashiorkor.
tive studies have examined the discriminatory perfor­
mance of different anthropometric measures against
the risk of death, but both weight-for-age and MUAC by normative bodies such as the WHO and UNICEF.
have been suggested for infants >60 days old136. Similar This delay has contributed to the poor uptake of this
to what has been found in older children, MUAC identi­ practice by local practitioners and professional bodies.
fies a slightly different group of infants but has ­superior
performance for predicting mortality than WLZs for Prevention
populations in Gambia and Kenya137,138. A MUAC of Given the high morbidity and mortality associated with
<110 mm for infants >60 days of age has been proposed severe malnutrition, preventing it is one of the most
based on a clinical trial in Kenya, which confirms that important goals in global health. Nevertheless, preven­
this measurement can identify infants with a very high tion remains elusive in most impoverished and humani­
risk of death6,138. tarian settings, and it requires programmes that address
maternal and child malnutrition holistically. Many
Screening countries have reduced the overall prevalence of severe
In practice, given the challenges of measuring WHZs malnutrition and stunting through the reduction of
frequently or at a large scale in a community, screening inequities141–143. In most instances, this achievement has
programmes tend to use a MUAC of <115 mm to assess required a combination of economic growth, the use of
severe malnutrition in children 6 months to 5 years public sector programmes focused on reducing inequities
of age, and the assessment of oedema is used to diag­ and investments in nutrition-sensitive (those that directly
nose kwashiorkor (FIG. 4). Indeed, the best approach for affect nutrition, such as breastfeeding and complemen­
screening for severe malnutrition is to measure MUAC tary feeding support programmes) and nutrition-specific
frequently, ideally every month or every time a child is (those that indirectly affect nutrition, such as improved
unwell. All anthropometric indices are better at identify­ agriculture and social safety nets) interventions. Indeed,
ing high-risk children in the short term, that is, in the no single intervention has effectively reduced the rates
month following the measure128, and this applies to of severe malnutrition or stunting for individual chil­
MUAC as well139. A promising approach to screening for dren144,145, and packages of public health approaches
severe malnutrition is to provide mothers with MUAC are needed. These packages should include improved
tapes that have a cut-off value of 115 mm and show them access to clean water, sanitation and hygiene, improved
how to detect malnutrition using this measurement6,140. agricultural productivity to minimize food insecurity,
This technique is simple enough to be taught rapidly140 timely universal vaccinations, early and efficient access
and enables malnutrition to be detected early (before to primary health facilities and care for acute illnesses (for
the onset of complications), hence reducing the need example, pneumonia, diarrhoea, measles and malaria),
for initial inpatient treatment6. However, the use of an emphasis on exclusive and continued breastfeeding
MUAC tapes to screen for severe malnutrition still has and attention to effective complementary feeding. Each
substantial room for global expansion, which has been approach is likely to affect malnutrition generally and,
delayed owing to the lack of guidelines, until recently, in turn, affect the rates of severe malnutrition146.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17067 | 9


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Box 3 | Management of severe acute malnutrition of treatment for acute malnutrition when necessary.
They should also receive all recommended immuniza­
The management of children with severe malnutrition tions and have ready access to essential interven­
has several components: tions and therapeutics when required (including oral
• Therapeutic feeding with macronutrients and rehydra­tion salts for diarrhoea, antibiotics for pneumo­
micronutrients to achieve homeostasis and overcome nia and antimalarials, as well as antiretroviral therapy
the nutrient deficits and opportunistic infection prophylaxis for children
• The immediate identification and management who have or have been exposed to HIV infection)154.
of life-threatening infections and complications Early and comprehensive intervention for children
such as sepsis and metabolic derangements
with moderate malnutrition is important for limiting
• The treatment of subclinical infections and underlying the incidence of severe malnutrition as these children
conditions, such as HIV or tuberculosis
are assumed to have an increased risk of progression to
• The cognitive stimulation and emotional support severe malnutrition155.
of the child and psychosocial support for their parents
or caregivers
Management
• Linkage to medical, nutritional and social services
The goals of management of severe malnutrition are
during therapy and recovery
to prevent short-term mortality, achieve sustained
nutri­tional recovery to reduce susceptibility to life-­
threatening infections and to support neurocognitive
Despite several countries reducing the rates of mal­ development. Despite their striking clinical differences,
nutrition in general, inconsistent effects of vertical pro­ the treatment recommendations for kwashiorkor and
grammes on childhood malnutrition have been reported. severe wasting have several common elements (BOX 3).
Indeed, the largest and most comprehensive multi­centre The current management of severe malnutrition has
trial testing the use of small quantities of lipid-­containing evolved over several decades and is mostly based on
nutrient supplements did not show significant benefits expert opinion and a few observational studies and not
regarding stunting or wasting in Malawi147 and showed on data from modern randomized clinical trials156,157.
only modest benefits in Ghana148,149. The same inter­ Management is based on a series of WHO guidelines
vention did have a better effect on stunting and wasting and training programmes, which are practical and
in Burkina Faso150, but notably, this trial also included have been widely adopted. Indeed, the management
treatment for diarrhoea and malaria. These data empha­ guidelines have become so engrained in clinical prac­
size the importance of interventions beyond nutritional tice that it might be considered unethical to carry out
therapy alone in at-risk populations. randomized controlled trials except to show equivalence
At the level of the individual, a comprehensive pre­ or superior­ity to the current standard of care. Notably,
vention package should begin with teenage girls and many of the principles of management have emerged
young women before pregnancy and should include from emergencies, and translating these to low-income
comprehensive health education and the use of nutri­ and middle-­income countries, in which severe mal­
tional interventions aimed at optimizing their overall nutrition is a daily burden on health systems, remains
physical health, nutritional status and psychosocial a challenge158. In general, management includes the use
readiness for pregnancy and childbirth151. During preg­ of therapeutic foods along with antibiotics to treat any
nancy, women should be provided with evidence-based underlying infections. Management can be undertaken
prenatal and perinatal support, including micro­nutrient in communities for children with uncomplicated severe
and macronutrient supplementation, treatment for malnutrition (that is, children who are clinically stable
infectious diseases (including prophylaxis and treatment and do not display clinical features of complications,
for acute infections) and early and frequent education especially coincidental infections).
regarding neonatal nutrition and care152; this strategy is
believed to improve the health status of both the mother Therapeutic foods
and the infant. Therapeutic foods, such as ready-to-use therapeutic
Optimizing macronutrient intake during infancy to foods (RUTFs) and F-75 and F-100 milks, were designed
prevent the development of severe malnutrition begins for nutritional and metabolic stabilization and rehabili­
with the prompt initiation of breastfeeding, followed by tation, and their use aims to address anticipated calor­
exclusive breastfeeding for the first six months of life ific needs and treatment stage-appropriate protein,
and the subsequent progressive introduction of nutri­ electrolyte and micronutrient requirements, in addition
tious and diverse complementary foods, with continued to initially limiting exposure to nutrients that could be
breastfeeding for ≥2 years153. In addition, context-­ harmful to metabolically unstable children or those
specific micronutrient programmes can be used and with infections, such as sodium and iron. However, the
might include vitamin A, iron, multiple micro­nutrient precise nutrient requirements of children with severe
powders or small quantities of lipid-containing nutrient malnutrition and the bioavailability of these therapeutic
supplements. Additionally, zinc supplements can be used foods are not well established and might vary by setting
in children with acute diarrhoea. All ­children <5 years and the presence of comorbidities in some patients.
of age living in at-risk communities should undergo The formulations of F-75 and F-100 were derived and
frequent growth monitoring, with the commencement optimized based on metabolic studies of hospitalized

10 | ARTICLE NUMBER 17067 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

children with severe malnutrition, with the aim of prov­ As RUTFs provide individuals with nutrients at levels
iding the micronutrients and macronutrients that are that are nearly impossible to reach without supplemen­
likely to be deficient in a child with severe malnutrition. tation164, the direct assessment of the effect of RUTFs
F-75 is used for initial refeeding and F-100 is effective on weight gain compared with the effect of a local diet
in facilitating rapid catch-up weight gain but might not alone has not been carried out. However, children
correct all of the metabolic disturbances in children with treated with RUTFs have a higher proportion of recovery
severe malnutrition. and weight gain than children who received a local diet
RUTFs were developed to contain the same nutri­ ­supplemented with additional vitamins and minerals7,165.
tional content as the F-100 therapeutic milk formula Despite the benefits associated with use of thera­
but in a dehydrated form that can be administered in peutics foods, a rapid increase in macronutrient and
the home environment with no preparation or cooking micronutrient intake can lead to some negative health
required and minimal risk of contamination159. RUTFs consequences, including diarrhoea (due to carbohydrate
have no mandatory formulation but do have minimum malabsorption)166,167 and refeeding syndrome (BOX 4).
micronutrient and macronutrient concentrations and The risk of refeeding syndrome might be reduced when
quality control and microbiological safety standards, ­children consume therapeutic foods on demand based on
which are outlined by WHO and UNICEF160. The pre­ their appetite rather than being force-fed using a naso­
dominant formulation of RUTFs consists of p ­ eanut gastric tube168,169. However, no randomized ­trials testing
paste, milk powder, sugar, oil and micronutrients. the formulations, amounts or durations of f­ eeding of F-75
The high cost of RUTFs is largely due to their milk or F-100 have been published.
powder component, which might be only partially
overcome by using local production methods as this Antibiotics
is also associated with high costs, including the costs Current guidelines recommend empiric antibiotics for
of constructing and maintaining appropriate produc­ all children with severe malnutrition because of con­
tion facilities and importing ingredients, as well as high cerns that although some children might not initially
local taxes. Trials investigating the use of RUTFs with­ present with signs of infection, they might deteriorate
out milk powder have suggested an inferior effect on suddenly without fever or other warning signs170,171.
child growth161,162. The essential polyunsaturated fatty No prospective clinical trials showing the need for anti­
acid (PUFA) composition of RUTFs is not optimal8,163, biotics in children with complicated severe mal­nutrition
but a more-favourable composition, such as a reduced have been published. However, there are reports of
ω-6 PUFA content or increased long-chain ω-3 PUFA in vitro nonsusceptibility to β-lactam antibiotics and
content, presents challenges for shelf life; the effects of gentamicin, the recommended first-line agents, although
these ­compositions on growth and neurodevelopment the implications of these findings in terms of outcomes
are uncertain. have yet to be tested in clinical trials. Interpreting the
results of these studies is challenging owing to biases;
these studies are usually conducted in tertiary centres
Box 4 | Refeeding syndrome
and/or do not distinguish community-acquired from
hospital-acquired infections172.
Refeeding syndrome can contribute to poor clinical outcomes in children with severe For outpatient management of children with
malnutrition. Rapidly increased nutrient intake can cause a switch from a catabolic to uncompli­cated, severe malnutrition, the use of routine
an anabolic state, which can lead to a surge in insulin secretion, acute hypoglycaemia antibiotic therapy has been questioned because of the
and the transport of extracellular electrolytes into cells. This electrolyte flux can lead
cost and the risk of enhancing antimicrobial resistance.
to dangerously low blood concentrations of potassium, magnesium and phosphate,
which can result in lethargy, seizures, muscle weakness, impaired cardiac function
In one study in Malawi, children with severe mal­
and respiratory failure225. nutrition receiving amoxicillin or cefdinir had 36%
The precise incidence of refeeding syndrome is unknown, which is related to the lack and 44% reductions in mortality, respectively, and
of a precise definition and the fact that, in most settings, the accurate measurement of they had faster recoveries than children who received
electrolytes and other biomarkers cannot be carried out. In addition, insufficient intake placebo5. Of note, this area had a high prevalence of
of electrolytes and their loss through the gastrointestinal and renal systems can affect kwashiorkor compared with other centres. In another
electrolyte concentrations, making it challenging to relate abnormal plasma levels to study in Niger, mortality was unchanged in children
refeeding syndrome. A recent study in Uganda reported hypophosphataemia in 76% with uncomplicated severe wasting who had not been
of children with severe malnutrition 2 days after admission, which was associated previously treated for malnutrition and who were
with mortality168.
given oral amoxi­cillin compared with placebo in an
Interestingly, refeeding syndrome influenced the development of the main
milk-based refeeding formulation used for the stabilization of patients (F-75),
out­patient feeding programme attached to a referral
which is characterized by a low protein content and reduced energy content hospital173. However, amoxicillin treatment was associ­
(mostly from carbohydrates) compared with F-100, and contains phosphate, ated with a faster recovery and a 14% reduction in the
potassium, magnesium, vitamin A and zinc, among other micronutrients. However, proportion of children who were referred to hospital in
as F-75 provides individuals with an energy source that is mostly from carbohydrates, another study173. Collectively, evidence from these t­ rials
this leads to increased glucose levels and, potentially, a surge in insulin secretion and supports the use of empiric antibiotics in outpatient
a high demand for phosphorylated glycolytic intermediates, which could precipitate manage­ment172,174, although further research on other
severe hypophosphataemia. Whether reducing calorie intake or energy from types and severities of malnutrition, other care settings,
carbohydrates during the initial hospitalization of patients with severe malnutrition the effects on antimicrobial resistance, the potential use
would reduce the incidence and severity of refeeding syndrome is currently unknown.
of point-of-care biomarkers and costs are needed.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17067 | 11


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Determine the severity of malnutrition systemic and intestinal inflammation. Trials assessing the
use of oral prebiotics or probiotics among children with
severe malnutrition have not shown effects on inpatient
Severe Moderate mortality outcomes, but they do suggest subsequent bene­
fits for growth9,180, and in the future, the use of prebiotics
Complicated Uncomplicated
and probiotics are likely to be better directed by improved
knowledge of microbiota ontogeny and function112,181.
• Emergency triage and treatment Gut-protective therapies targeting inflammation, micro­
• Careful fluid management bial translocation, malabsorption, intraluminal nutrient
• Intravenous antibiotics • Oral antibiotics processing and normalization of the intestinal micro­
• Treatment of specific infections • Deworming
biota might yield novel and effective interventions. Other
• Milk-based therapeutic foods • RUTFs
• Psychosocial stimulation • Psychosocial stimulation potential strategies for reducing malnutrition-­associated
mortality and improving growth include the use of
anti-inflammatory agents182 and gut-trophic nutrients,
Supplementary feeding including alanyl-glutamine176,183 and other dipeptides.
Inpatient care
Outpatient care Discharge Uncomplicated severe malnutrition
Until the 2000s, all children with severe malnutrition
Figure 5 | Overview of management of severe and moderate malnutrition. The first were usually managed as inpatients with milk-based
step in the treatment of malnutrition is to determine the severity. Moderate malnutrition therapeutic foods and empiric broad-spectrum paren­
is typically treated with the use of supplementary foods, which are administered in
teral antibiotics, which assumed that these patients
addition to the child’s normal diet. Uncomplicated severe malnutrition is usually treated
with ready-to-use therapeutic foods (RUTFs), which are given instead of the child’s normal always had nutritional and metabolic complications and
diet, in addition to a course of oral antibiotics and along with other treatments, such as serious, sub-patent infections127. However, management
deworming and treatment for other non-life-threatening infections. The management of has been revolutionized by the detection of children with
complicated severe malnutrition requires admission to an inpatient facility with adequate uncomplicated malnutrition, who can be safely treated
paediatric emergency care facilities and a nutritional rehabilitation care programme. in the community via a proactive, public health-based
Children with complicated malnutrition can be discharged from inpatient facilities when approach to care. This is made possible by community
serious complications have resolved, and subsequently entered into outpatient screening that uses MUAC assessments and by feeding
management programmes. Management includes the use of therapeutic foods and children a RUTF at a dose of 175 kcal/kg per day129,184.
antibiotics and the treatment of other infections. After the completion of treatment for The effectiveness of RUTFs in decreasing the mor­
severe malnutrition, children are transitioned from RUTFs to their home diet, which is a
tality of children with severe malnutrition cannot be
potential point of faltering. However, aside from standardized supplementary feeding,
strategies to decrease this risk have not been assessed in trials. overstated. Early RUTF trials reported a lower mortal­
ity for children treated with RUTFs in their community
than for those previously treated in hospitals. Following
Other interventions these trials, community treatment programmes rapidly
Profuse watery diarrhoea, hypovolaemia and shock expanded, which dramatically improved treatment cover­
are common in children with severe malnutrition and are age to reach previously untreated children. Following
associated with high mortality, despite current manage­ from this, many community-based programmes have
ment guidelines99,175. The optimal composition and rate shown lower mortality rates compared with historical
of administration of intravenous or oral fluid therapy, or data for hospital-based treatment outcomes. Accordingly,
other supportive therapies, in settings that do not have the development of community-based programmes has
access to advanced paediatric intensive care is unclear and largely occurred in the absence of formal randomized
controversial176 as only two small trials have been con­ controlled trials (comparing children treated with RUTFs
ducted to study individuals with severe mal­nutrition177,178. with untreated children), as carrying out these trials
Another large trial indicated that aggressive fluid would be unethical. For the same reasons, the effective­
manage­ment in children without severe malnutrition ness of hospital-based treatment in reducing mortality
and diarrhoea, but with some signs of shock, is harm­ has never been formally established.
ful179, which is likely to be the case in c­ hildren with mal­ Decentralized services closer to patients’ homes can
nutrition. Studies that distinguish shock caused by sepsis avoid the costs and risks of inpatient care, including
from hypovolaemia caused by diarrhoea in ­children nosocomial infections185,186, and has enabled uncompli­
with severe malnutrition have not been c­ arried out, cated severe malnutrition (generally >90% of all children
but they are needed to inform management strategies with severe malnutrition) to be treated in the commu­
that could be implemented in hospital settings. nity184 (FIG. 5). Outcomes of children with uncompli­
Other features of severe malnutrition include chronic cated severe malnutrition treated in their community
systemic and intestinal inflammation, infection and are markedly better than of children with complicated
environ­mental enteric dysfunction (that is, inflammation, malnutrition of similar anthropometric status187,188.
reduced absorption and reduced barrier function in the
small intestine)49. Mortality risk is more closely related Complicated severe malnutrition
to markers of systemic rather than intestinal inflam­ Children with complicated severe malnutrition (that is,
mation102, although bacterial intestinal translocation children with a more-severe disease who are sick)
and aberrant immune function might contribute to both are managed as inpatients so that life-threatening

12 | ARTICLE NUMBER 17067 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

complications (such as infections) can be addressed feeding with, initially, less energy-dense food aims to pre­
along with nutritional and metabolic deficits. The aim of vent and treat hypoglycaemia, reduce sodium l­ evels, and
nutritional management is to address disturbed carbo­ increase potassium and phosphate levels. The adminis­
hydrate and energy metabolism, which affects blood glu­ tration of hyperosmolar and intravenous fluids is avoided
cose ­levels and body temperature regulation67,189, as well ­whenever possible190.
as the distribution of fluid and electrolytes. Regular Therapeutic feeding is cautiously started with milk-
based food (specifically, 130 ml/kg per day of F-75,
which provides 97.5 kcal/kg per day) that initially con­
Emergency triage and treatment tain low levels of protein and sodium and high levels
of potassium, which is designed to match the child’s
Check airway and breathing reduced metabolic capacity. Initial feeding aims to
improve metabolism, gut motility and nutrient absorp­
Assess circulation for shock tion, and clinical improvements during this phase
and check for coma manifest as improved appetite and reduced oedema
(if present). When appetite is restored and the signs of
Ongoing care infectious complications improve, children transition
to RUTFs or milk-based foods with higher energy and
protein levels (such as F-100 at 130–200 ml/kg per day
Check for and treat dehydration Stabilization phase
Broad-spectrum antibiotics which provides 130–200 kcal/kg per day) to gain weight.
and the early initiation of No other food should be given, with the exception of
Routine investigations therapeutic food breast milk, which should be given before therapeutic
foods are administered to keep stimulating the maternal

Regular structured assessment of medical and


Treatment of acute medical condition(s) milk supply. Additional micronutrients (such as zinc for
nutritional complications and feeding
diarrhoea) beyond what are included in the therapeutic
Actively diagnose chronic conditions food are generally unnecessary without specific signs of
HIV, tuberculosis, neurodisability etc. a deficiency190.
If the patient has Under current WHO recommendations, children
If the patient has an improving resolving oedema and are discharged from inpatient care when their serious
medical condition is regaining appetite complications have resolved rather than after they have
met specific anthropometric targets, and they continue
Deterioration Transition phase therapeutic feeding and supplementary feeding (for
at any time Step up to F-100 or RUTFs moderate malnutrition) as outpatients151 (FIG. 6). The
Breastfeeding and transition of patients from the hospital setting to home
nutrition counselling requires ensuring that the parent or caregiver under­
Rehabilitation phase stands the treatment phases and what is expected from
F-100 or RUTF them in addition to enrolling them in available nutri­
If medically fit for discharge tional, medical and social services.
from hospital Psychosocial
stimulation Children with complicated severe malnutrition
are usually admitted to hospital because of severe
Multidisciplinary decision to discharge infections and typically have a fatality rate of 12%
to >20% 168,191. HIV infection is present in 15–29%
Community-based therapeutic feeding
of children hospitalized with severe malnutrition in
Medical follow-up for chronic conditions and social services as required Sub-Saharan Africa and is associated with a threefold
increased risk of inpatient mortality168,192,193. Studies
Figure 6 | Detailed management of complicated severe malnutrition. The challenges aiming to reduce mortality in children with complicated
associated with the management of children with complicated severe malnutrition require
Nature Reviews | Disease Primers severe malnutrition have included increased nursing
the integration of structured, high-quality paediatric care and nutritional support, which
must happen in parallel and in a multidisciplinary manner. Paediatric care focuses on the support, systematic feeding or reductions in intra­venous
identification and treatment of the most immediately life-threatening problems, as laid out fluid administration and have been met with limited
in the WHO Emergency Triage and Treatment guidelines. Nutritional support focuses first success194–196, ­presenting a clear need for research to
on helping to restore physiological processes to a normal state and then on achieving improve care22,176.
rehabilitation. Initially, use of the ‘ABCD’ mnemonic can help focus attention on the
assessment and maintenance of the airway (A) and breathing (B), circulation (C) and Infants <6 months of age
disability (coma, convulsion and dehydration (D)). Children with complicated severe Infants <6 months of age with severe malnutrition are
malnutrition remain exceptionally vulnerable during treatment and can deteriorate rapidly fed with breast milk and infant formula, or diluted
without clear prior warning signs. Accordingly, monitoring and frequent re-evaluation are F-100 milk, often by using a technique called ‘supple­
essential. When prevalent, comorbidities such as tuberculosis and HIV should be actively
mentary suckling’. A key aim of treatment is to estab­
tested for. If community-based management of uncomplicated severe malnutrition is
available, the decision to discharge children from hospital is based on their appetite and lish or re­establish effective exclusive breastfeeding
the resolution of complications rather than on anthropometric parameters, and should wherever possible. These infants also receive empiric
be decided by nutritional and clinical staff. The treatment process in hospital and after antibiotics134. Further research is needed to determine
discharge should be explained to parents and caregivers, and they should know what help the efficacy of this strategy and other components of the
is available to them and what is expected of them. RUTFs, ready-to-use therapeutic foods. management of these infants197.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17067 | 13


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Box 5 | Selected research priorities in severe malnutrition who have been treated for severe malnutrition typically
have less lean mass (that is, fat-free mass) than children
• Determining the burden of severe malnutrition based on its incidence without malnutrition in the same community162,204,205.
• Determining the causes of death associated with malnutrition, including infection Indeed, children in Malawi had lower lean tissue and
and impaired energy metabolism were more severely stunted at an average of 7 years after
• Determining the pathophysiology of kwashiorkor and its implications for hospitalization for severe malnutrition, than controls, but
management had a similar head circumference and respiratory and
• Determining the mechanisms of functional immune impairment and the association cardiometabolic function204. In addition, children with
with nutritional rehabilitation prior severe malnutrition have weaker hand grip and
• Determining the long-term effect on body composition, metabolism and lower exercise tolerance than children who have not had
neurodevelopment of severe malnutrition and its treatment, with or without preterm malnutrition204,206. This ‘thrifty’ growth pattern indicates
and/or low birth rates that severe malnutrition in early childhood might predis­
• Devising preventive strategies that work on a large scale and are low in cost pose an individual to noncommunicable diseases later in
• Developing strategies for the integration of protocols to prevent severe malnutrition life. However, this predisposition is difficult to assess as
in fragile and humanitarian settings stunting and other insults might have been present before
• Improving the management of acute malnutrition in infants <6 months of age197 severe malnutrition developed in the children enrolled in
• Improving risk stratification to help identify which patients need inpatient care these studies. Although the effect of in utero exposure to
and those that need empiric antimicrobials, and determining which antimicrobials malnutrition and its risk factors leading to a wide range of
should be used noncommunicable diseases later in life is estab­lished207,208,
• Simplifying treatment protocols and developing protocols that can be used at the critical window or risk for these effects and the
the community level with minimal supervision consequent effect of infant and child malnutrition on
• Developing new formulations of ready-to-use therapeutic foods that optimize noncommunicable diseases later in life is still unclear209.
several nutritional parameters to maximize patient survival, weight gain, Severe malnutrition and stunting are associated with
correction of metabolic perturbations, and long-term cognitive, physical delayed child neurodevelopment, behavioural problems,
and immunological development lower school achievement and reduced adult earning
potential2,210. The use of psychosocial stimulation, such
as play and giving positive feedback training to mothers
Quality of life or prolonged home visiting programmes after infants or
Children with severe malnutrition typically live in set­ young children with severe wasting or kwashiorkor have
tings of pervasive poverty, low female literacy, food been discharged from the hospital, has been shown to
­insecurity, inadequate water and sanitation and over­ significantly improve infant development scores in the
stretched health systems198. Acute management inter­ short term, as well as educational achievement and IQ
ventions for the treatment of severe malnutrition do not into adolescence116,211,212. Several micronutrients are
fundamentally alter these underlying risks. Most chil­ potentially essential for cognitive development, including
dren with acute malnutrition are also severely stunted at PUFAs, thiamine, choline, vitamin D, folate, iron, copper,
presentation7,173, which indicates that they have chronic selenium and iodine213, and deficiencies in these might
ill health and malnutrition. Several studies in Malawi, vary among children with severe malnutrition according
Kenya, Gambia and Bangladesh have shown that anthro­ to the child’s age, location and the cause of malnutrition.
pometric recovery is not sufficient as children have a high However, supplementation with these micronutrients has
rate of death following discharge from inpatient care for not undergone clinical trials with neurodevelopmental
severe malnutrition, predominantly from pneumonia end points in the context of severe malnutrition.
and diarrhoea6,199. In addition, mortality is increased Further studies are needed to assess the optimal dur­
in children following treatment in community-based ation of treatment with therapeutic foods, factors limit­
nutrition programmes184,200. The reasons for this likely ing lean (muscle, organ and connective) tissue synthesis
contains a home environment that includes risk factors (including the role of high-quality dietary protein), how
for malnutrition, an undiagnosed chronic illness and zinc and other micronutrients can modulate lean tissue
that attending health care visits for episodes of illness or deposition, the benefits of psychosocial stimulation and
ongoing therapeutic feeding might be limited in certain physical activity and the long-term effects of malnutrition
areas200,201. In addition, some children with severe mal­ on health risks in adolescence and adulthood204.
nutrition might have a pre-existing vulnerability (such
as a mild immune deficit that would cause minimal Outlook
health problems in a resource-rich setting with a low Although this Primer has highlighted many of the neg­
prevalence of infectious diseases) that results in a cycle ative consequences of severe malnutrition, it is impor­
of illness and malnutrition and a high degree of vulner­ tant to note that our understanding of the epidemiology
ability to infection202. Certainly, not all siblings who grow and management of severe malnutrition has improved.
up in the same family are equally affected by the same Indeed, community-based management of severe mal­
home environment203. nutrition has revolutionized treatment programmes and
During treatment and recovery from severe mal­ has enabled large numbers of children to receive effective
nutrition, weight and MUAC usually increase rapidly, but nutritional and clinical care in various settings. Children
height does not, and growth is typically sufficient only to with severe malnutrition and a high risk of death can be
reach the baseline height-for-age Z-score173,203. Children easily identified, and many of them can be successfully

14 | ARTICLE NUMBER 17067 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

treated at the community level by parents with assis­ nets to address food security are other areas to explore.
tance from community health workers, in addition to Other research gaps regarding severe malnutrition are
outreach services. described in BOX 5 (REFS 218,219).
The use of appropriate prevention and manage­ An improved understanding of several areas of the
ment interventions for severe malnutrition could pre­ pathophysiology of severe malnutrition and the effi­
vent 61% of cases of wasting and almost 350,000 child cacy of potential interventions might be able to save the
deaths ­annually214, and several major international initi­ lives of these high-risk children. For example, under­
atives, such as No Wasted Lives215, are working towards standing the pathophysiology of kwashiorkor could lead
­ambitious targets to achieve this. Although progress to phenotype-­specific therapeutic interventions; this
towards lowering childhood mortality is ongoing, more requires research that uses modern tools for assessing
work is still required. Indeed, as more children sur­ metabolism and other physiological functions. In addi­
vive episodes of severe malnutrition, and as poverty tion, although much of the previous research on the
decreases in many countries, new challenges will emerge. pathophysiology of severe malnutrition has been inform­
For example, helping children to survive an episode of ative and hypothesis-generating, the research does not
severe malnutrition is no longer enough. Following the commonly meet today’s standards for quality in terms of
Global Strategy for Women’s, Children’s and Adolescents’ participant selection, the avoidance of bias and sample
Health (2016–2030), we must also help these children size. Going forward, the testing of well-founded hypoth­
to thrive214. To do this, curative and preventive services eses in appropriately designed randomized trials with
must be transformed. More effort is needed in previously the use of biomarkers, information on mechanisms and
neglected populations, such as infants <6 months of age216 sound clinical measures is required.
and children with underlying disabilities217 and other The overarching priority is to make severe malnutri­
identi­fied vulnerabilities. In addition, focus is needed tion a thing of the past in countries in Africa and Asia, as
on the prevention of relapse and readmission, although is the case in Europe and North America today. Achieving
how this can be achieved is uncertain. One large trial the SDGs for health and nutrition will be possible only
investigating the use of prophylactic co-trimoxazole for through addressing severe malnutrition, and to do this,
high-risk children following discharge from hospital in we must recognize that reducing the burden of morbidity
Kenya showed negative results for reducing the risk of and mortality associated with severe childhood malnutri­
relapse and death6. However, other broad-spectrum anti­ tion is a global moral imperative. To eradicate severe mal­
biotics or other therapies that could support a prolonged nutrition will require political will, investment in peace
period of immune system recovery might be beneficial. building and reducing man-made and natural disasters,
In addition, improved social support and social safety which are issues that should unite the world.

1. United Nations. Sustainable development goal 2. 8. Hsieh, J. C. et al. High-oleic ready‑to‑use therapeutic of severe malnutrition in a child: a case–control study.
United Nations https://sustainabledevelopment.un.org/ food maintains docosahexaenoic acid status in severe Eur. J. Clin. Nutr. 48, 416–424 (1994).
sdg2 (2015). malnutrition. J. Pediatr. Gastroenterol. Nutr. 61, 18. Owor, M., Tumwine, J. K. & Kikafunda, J. K. Socio-
2. Black, R. E. et al. Maternal and child undernutrition 138–143 (2015). economic risk factors for severe protein energy
and overweight in low-income and middle-income 9. Kerac, M. et al. Probiotics and prebiotics for severe malnutrition among children in Mulago Hospital,
countries. Lancet 382, 427–451 (2013). acute malnutrition (PRONUT study): a double-blind Kampala. East Afr. Med. J. 77, 471–475 (2000).
This is an important global review of the efficacy randomised controlled trial in Malawi. Lancet 19. Saloojee, H., De Maayer, T., Garenne, M. L. & Kahn, K.
epidemiology, major risk factors and consequences 374, 136–144 (2009). What’s new? Investigating risk factors for severe
of malnutrition. 10. Katz, J. et al. Mortality risk in preterm and small-for- childhood malnutrition in a high HIV prevalence
3. Nandy, S. & Miranda, J. J. Overlooking gestational-age infants in low-income and middle- South African setting. Scand. J. Public Health Suppl.
undernutrition? Using a composite index of income countries: a pooled country analysis. Lancet 69, 96–106 (2007).
anthropometric failure to assess how underweight 382, 417–425 (2013). 20. Mishra, K., Kumar, P., Basu, S., Rai, K. & Aneja, S.
misses and misleads the assessment of undernutrition 11. van der Pols-Vijlbrief, R., Wijnhoven, H. A., Risk factors for severe acute malnutrition in children
in young children. Soc. Sci. Med. 66, 1963–1966 Schaap, L. A., Terwee, C. B. & Visser, M. Determinants below 5 y of age in India: a case–control study.
(2008). of protein-energy malnutrition in community-dwelling Indian J. Pediatr. 81, 762–765 (2014).
4. McDonald, C. M. et al. The effect of multiple older adults: a systematic review of observational 21. Ricci, J. A. & Becker, S. Risk factors for wasting and
anthropometric deficits on child mortality: meta- studies. Ageing Res. Rev. 18, 112–131 (2014). stunting among children in Metro Cebu, Philippines.
analysis of individual data in 10 prospective studies 12. Fritsch, P. Nutrition interventions for older people Am. J. Clin. Nutr. 63, 966–975 (1996).
from developing countries. Am. J. Clin. Nutr. 97, in emergencies: HelpAge International. HelpAge 22. Trehan, I., O’Hare, B. A., Phiri, A. & Heikens, G. T.
896–901 (2013). International http://nutritioncluster.net/wp-content/ Challenges in the management of HIV-infected
This review of data from ten prospective cohorts uploads/sites/4/2015/06/Nutrition-FINAL.pdf (2013). malnourished children in sub-Saharan Africa.
summarized the observed association of major 13. Gladstone, M. et al. Assessment of neurodisability AIDS Res. Treat. 2012, 790786 (2012).
anthropometric deficits and the additive effects and malnutrition in children in Africa. Semin. Pediatr. 23. Maxwell, D. et al. Emergency Food Security
of infections on risks of death in childhood. Neurol. 21, 50–57 (2014). Interventions (Humanitarian Practice Network, 2008).
5. Trehan, I. et al. Antibiotics as part of the management 14. Kerac, M. et al. The interaction of malnutrition and 24. Tolboom, J. J., Ralitapole-Maruping, A. P., Kabir, H.,
of severe acute malnutrition. N. Engl. J. Med. 368, neurologic disability in Africa. Semin. Pediatr. Neurol. Molatseli, P. & Anderson, J. Severe protein energy
425–435 (2013). 21, 42–49 (2014). malnutrition in Lesotho, death and survival in
This landmark study demonstrated the beneficial 15. United Nations Children’s Fund, World Health hospital, clinical findings. Trop. Geogr. Med. 38,
impact on overall mortality and recovery of Organization & World Bank Group. Joint child 351–358 (1986).
administering antibiotics to children treated malnutrition estimates 2017. WHO http://apps.who. 25. Institute for Health Metrics and Evaluation.
for severe malnutrition. int/gho/data/node.wrapper.nutrition-2016 (2017). Global Burden of Disease Results Tool.
6. Berkley, J. A. et al. Daily co‑trimoxazole 16. Frison, S., Checchi, F. & Kerac, M. Omitting edema GHDx http://ghdx.healthdata.org/gbd-results-
prophylaxis to prevent mortality in children with measurement: how much acute malnutrition are we tool?params=querytool-permalink/
complicated severe acute malnutrition: a missing? Am. J. Clin. Nutr. 102, 1176–1181 (2015). d34931ea04210ce4039fbf42ec0a62c8 (2015).
multicentre, double-blind, randomised placebo- This article presented a secondary data analysis 26. Pelletier, D. L., Frongillo, E. A. Jr, Schroeder, D. G.
controlled trial. Lancet Glob. Health 4, e464–e473 of 852 nutrition cross-sectional survey data sets & Habicht, J. P. The effects of malnutrition on child
(2016). of children 6–59 months of age and showed that mortality in developing countries. Bull. World Health
7. Bhandari, N. et al. Efficacy of three feeding regimens the prevalence of oedematous malnutrition varied Organ. 73, 443–448 (1995).
for home-based management of children with from 0% to 32.9% and that many children with 27. Caulfield, L. E., de Onis, M., Blossner, M. & Black, R. E.
uncomplicated severe acute malnutrition: oedema did not have wasting. Undernutrition as an underlying cause of child deaths
a randomised trial in India. BMJ Glob. Health 17. Islam, M. A., Rahman, M. M. & Mahalanabis, D. associated with diarrhea, pneumonia, malaria, and
1, e000144 (2016). Maternal and socioeconomic factors and the risk measles. Am. J. Clin. Nutr. 80, 193–198 (2004).

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17067 | 15


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

28. Sethuraman, K., Lansdown, R. & Sullivan, K. Women’s 51. Takele, Y. et al. Malnutrition in healthy individuals superimposed acute stress. Arch. Dis. Child 48,
empowerment and domestic violence: the role of results in increased mixed cytokine profiles, altered 714–716 (1973).
sociocultural determinants in maternal and child neutrophil subsets and function. PLoS ONE 11, 76. Kerpel-Fronius, E. Pathophysiology of Infantile
undernutrition in tribal and rural communities in e0157919 (2016). Malnutrition: Protein-Energy Malnutrition and Failure
South India. Food Nutr. Bull. 27, 128–143 (2006). 52. Hughes, S. M. et al. Dendritic cell anergy results to Thrive (Akademiai Kiado, 1983).
29. Kinyoki, D. K. et al. Conflict in Somalia: impact on from endotoxemia in severe malnutrition. J. Immunol. 77. Kalk, W. J. et al. Thyroid hormone and carrier protein
child undernutrition. BMJ Global Health 2, e000262 183, 2818–2826 (2009). interrelationships in children recovering from
(2017). 53. Hoang, T., Agger, E. M., Cassidy, J. P., kwashiorkor. Am. J. Clin. Nutr. 43, 406–413 (1986).
30. [No authors listed.] Classification of infantile Christensen, J. P. & Andersen, P. Protein energy 78. Becker, K. et al. Effects of antioxidants on glutathione
malnutrition. Lancet 2, 302–303 (1970). malnutrition during vaccination has limited influence levels and clinical recovery from the malnutrition
31. Cohen, S., Nathan, J. A. & Goldberg, A. L. Muscle on vaccine efficacy but abolishes immunity if syndrome kwashiorkor — a pilot study. Redox Rep.
wasting in disease: molecular mechanisms and administered during Mycobacterium tuberculosis 10, 215–226 (2005).
promising therapies. Nat. Rev. Drug Discov. 14, infection. Infect. Immun. 83, 2118–2126 (2015). 79. Fechner, A. et al. Antioxidant status and nitric
58–74 (2015). 54. Saucillo, D. C., Gerriets, V. A., Sheng, J., oxide in the malnutrition syndrome kwashiorkor.
32. Argiles, J. M., Busquets, S. & Lopez-Soriano, F. J. Rathmell, J. C. & Maciver, N. J. Leptin metabolically Pediatr. Res. 49, 237–243 (2001).
Cytokines in the pathogenesis of cancer cachexia. licenses T cells for activation to link nutrition and 80. Golden, M. H. & Ramdath, D. Free radicals in the
Curr. Opin. Clin. Nutr. Metab. Care 6, 401–406 (2003). immunity. J. Immunol. 192, 136–144 (2014). pathogenesis of kwashiorkor. Proc. Nutr. Soc. 46,
33. Grant, R. W. & Stephens, J. M. Fat in flames: influence 55. Palacio, A., Lopez, M., Perez-Bravo, F., Monkeberg, F. 53–68 (1987).
of cytokines and pattern recognition receptors on & Schlesinger, L. Leptin levels are associated with 81. Sive, A. A., Subotzky, E. F., Malan, H., Dempster, W. S.
adipocyte lipolysis. Am. J. Physiol. Endocrinol. Metab. immune response in malnourished infants. J. Clin. & Heese, H. D. Red blood cell antioxidant enzyme
309, E205–E213 (2015). Endocrinol. Metab. 87, 3040–3046 (2002). concentrations in kwashiorkor and marasmus.
34. Bowen, T. S., Schuler, G. & Adams, V. Skeletal muscle 56. Kerpel-Fronius, E. & Kaiser, E. Hypoglycaemia in Ann. Trop. Paediatr. 13, 33–38 (1993).
wasting in cachexia and sarcopenia: molecular infantile malnutrition. Acta Paediatr. Scand. Suppl. 82. Manary, M. J., Leeuwenburgh, C. & Heinecke, J. W.
pathophysiology and impact of exercise training. 172, 119+ (1967). Increased oxidative stress in kwashiorkor. J. Pediatr.
J. Cachexia Sarcopenia Muscle 6, 197–207 (2015). 57. Golden, M. The effects of malnutrition in the 137, 421–424 (2000).
35. Williams, C. D. A nutritional disease of childhood metabolism of children. Trans. R. Soc. Trop. Med. Hyg. 83. Ciliberto, H. et al. Antioxidant supplementation for
associated with a maize diet. Arch. Dis. Child 8, 82, 3–6 (1988). the prevention of kwashiorkor in Malawian children:
423–433 (1933). 58. Whitehead, R. G. & Alleyne, G. A. Pathophysiological randomised, double blind, placebo controlled trial.
36. Laditan, A. A. & Reeds, P. J. A study of the age of factors of importance in protein-calorie malnutrition. BMJ 330, 1109 (2005).
onset, diet and the importance of infection in the Br. Med. Bull. 28, 72–79 (1972). 84. van Zutphen, T. et al. Malnutrition-associated liver
pattern of severe protein-energy malnutrition in 59. Jahoor, F., Badaloo, A., Reid, M. & Forrester, T. steatosis and ATP depletion is caused by peroxisomal
Ibadan, Nigeria. Br. J. Nutr. 36, 411–419 (1976). Protein metabolism in severe childhood malnutrition. and mitochondrial dysfunction. J. Hepatol. 65,
37. Sullivan, J., Ndekha, M., Maker, D., Hotz, C. Ann. Trop. Paediatr. 28, 87–101 (2008). 1198–1208 (2016).
& Manary, M. J. The quality of the diet in Malawian 60. Jahoor, F., Badaloo, A., Reid, M. & Forrester, T. 85. Brooks, S. E., Doherty, J. F. & Golden, M. H.
children with kwashiorkor and marasmus. Protein kinetic differences between children with Peroxisomes and the hepatic pathology of childhood
Matern. Child Nutr. 2, 114–122 (2006). edematous and nonedematous severe childhood malnutrition. West Indian Med. J. 43, 15–17 (1994).
38. Coulthard, M. G. Oedema in kwashiorkor is caused undernutrition in the fed and postabsorptive states. 86. Ruggieri, A. J., Levy, R. J. & Deutschman, C. S.
by hypoalbuminaemia. Paediatr. Int. Child Health 35, Am. J. Clin. Nutr. 82, 792–800 (2005). Mitochondrial dysfunction and resuscitation in sepsis.
83–89 (2015). 61. Jahoor, F., Badaloo, A., Reid, M. & Forrester, T. Crit. Care Clin. 26, 567–575 (2010).
39. Coward, D. G. & Whitehead, R. G. Experimental Sulfur amino acid metabolism in children with severe 87. Leite, H. P. & de Lima, L. F. Metabolic resuscitation
protein-energy malnutrition in baby baboons. childhood undernutrition: methionine kinetics. in sepsis: a necessary step beyond the hemodynamic?
Attempts to reproduce the pathological features Am. J. Clin. Nutr. 84, 1400–1405 (2006). J. Thorac Dis. 8, E552–E557 (2016).
of kwashiorkor as seen in Uganda. Br. J. Nutr. 28, 62. Lewis, B., Wittmann, W., Krut, L. H., Hansen, J. D. 88. Webb, J. G., Kiess, M. C. & Chan-Yan, C. C.
223–237 (1972). & Brock, J. F. Free fatty acid flux through plasma in Malnutrition and the heart. CMAJ 135, 753–758
40. Golden, M. H., Golden, B. E. & Jackson, A. A. Albumin protein malnutrition of infants. Clin. Sci. 30, 371–375 (1986).
and nutritional oedema. Lancet 1, 114–116 (1980). (1966). 89. Wharton, B. A., Howells, G. R. & McCance, R. A.
41. Golden, M. H. Oedematous malnutrition. Br. Med. 63. Badaloo, A. V., Forrester, T., Reid, M. & Jahoor, F. Cardiac failure in kwashiorkor. Lancet 2, 384–387
Bull. 54, 433–444 (1998). Lipid kinetic differences between children with (1967).
42. Scrimshaw, N. S. Historical concepts of interactions, kwashiorkor and those with marasmus. Am. J. Clin. 90. El‑Sayed, H. L., Nassar, M. F., Habib, N. M.,
synergism and antagonism between nutrition and Nutr. 83, 1283–1288 (2006). Elmasry, O. A. & Gomaa, S. M. Structural and
infection. J. Nutr. 133, 316S–321S (2003). 64. Bartz, S. et al. Severe acute malnutrition in childhood: functional affection of the heart in protein energy
43. Bourke, C. D., Berkley, J. A. & Prendergast, A. J. hormonal and metabolic status at presentation, malnutrition patients on admission and after
Immune dysfunction as a cause and consequence of response to treatment, and predictors of mortality. nutritional recovery. Eur. J. Clin. Nutr. 60, 502–510
malnutrition. Trends Immunol. 37, 386–398 (2016). J. Clin. Endocrinol. Metab. 99, 2128–2137 (2014). (2006).
44. Rytter, M. J., Kolte, L., Briend, A., Friis, H. 65. Wharton, B. Hypoglycaemia in children with 91. Silverman, J. A. et al. The effects of malnutrition on
& Christensen, V. B. The immune system in children kwashiorkor. Lancet 1, 171–173 (1970). cardiac function in African children. Arch. Dis. Child
with malnutrition — a systematic review. PLoS ONE 9, 66. Bandsma, R. H. et al. Mechanisms behind decreased 101, 166–171 (2016).
e105017 (2014). endogenous glucose production in malnourished 92. Gruber, C. et al. Myocardial remodelling in left
45. Ahmed, T. et al. An evolving perspective about the children. Pediatr. Res. 68, 423–428 (2010). ventricular atrophy induced by caloric restriction.
origins of childhood undernutrition and nutritional 67. Spoelstra, M. N. et al. Kwashiorkor and marasmus J. Anat. 220, 179–185 (2012).
interventions that includes the gut microbiome. are both associated with impaired glucose clearance 93. Badaloo, A., Reid, M., Soares, D., Forrester, T.
Ann. NY Acad. Sci. 1332, 22–38 (2014). related to pancreatic β-cell dysfunction. Metabolism & Jahoor, F. Relation between liver fat content and
This review summarized some of the major insights 61, 1224–1230 (2012). the rate of VLDL apolipoprotein B-100 synthesis in
and hypotheses into how the interplay between 68. Becker, D. J., Pimstone, B. L., Hansen, J. D., children with protein-energy malnutrition. Am. J. Clin.
microbial, environmental and host factors MacHutchon, B. & Drysdale, A. Patterns of insulin Nutr. 81, 1126–1132 (2005).
contribute to severe malnutrition and how this response to glucose in protein-calorie malnutrition. 94. Lewis, B., Hansen, J. D., Wittman, W., Krut, L. H.
knowledge might be used to develop new therapies Am. J. Clin. Nutr. 25, 499–505 (1972). & Stewart, F. Plasma free fatty acids in kwashiorkor
against malnutrition. 69. Baig, H. A. & Edozien, J. C. Carbohydrate metabolism and the pathogenesis of the fatty liver. Am. J. Clin.
46. Assa, A. et al. Vitamin D deficiency promotes in kwashiorkor. Lancet 2, 662–665 (1965). Nutr. 15, 161–168 (1964).
epithelial barrier dysfunction and intestinal 70. Reis, M. A. et al. Glucose-induced insulin secretion 95. Waterlow, J. C. Oxidative phosphorylation in the
inflammation. J. Infect. Dis. 210, 1296–1305 (2014). is impaired and insulin-induced phosphorylation of livers of normal and malnourished human infants.
47. Wong, S. C. et al. Growth and the growth hormone– the insulin receptor and insulin receptor substrate‑1 Proc. R. Soc. Lond. B Biol. Sci. 155, 96–114 (1961).
insulin like growth factor 1 axis in children with chronic are increased in protein-deficient rats. J. Nutr. 127, 96. Brooks, S. E., Goldon, M. H. & Taylor, E. Hepatic
inflammation: current evidence, gaps in knowledge, 403–410 (1997). ultrastructure in children with protein-energy
and future directions. Endocr. Rev. 37, 62–110 (2016). 71. Ferreira, F. et al. Decreased insulin secretion in islets malnutrition. West Indian Med. J. 41, 139–145
48. Guerrant, R. L. et al. Biomarkers of environmental from rats fed a low protein diet is associated with (1992).
enteropathy, inflammation, stunting, and impaired a reduced PKAα expression. J. Nutr. 134, 63–67 97. Morlese, J. F. et al. Albumin kinetics in edematous and
growth in children in Northeast Brazil. PLoS ONE 11, (2004). nonedematous protein-energy malnourished children.
e0158772 (2016). 72. Soriano, S. et al. Reduced insulin secretion in protein Am. J. Clin. Nutr. 64, 952–959 (1996).
This study of systemic biomarkers of enteropathy malnourished mice is associated with multiple changes 98. Keusch, G. T. et al. Implications of acquired
and growth predictors in 375 children 6–26 in the β-cell stimulus-secretion coupling. environmental enteric dysfunction for growth
months of age in northeast Brazil demonstrated Endocrinology 151, 3543–3554 (2010). and stunting in infants and children living in low-
how environmental enteric dysfunction contributes 73. Robinson, H. M. & Seakins, A. Fasting pancreatic and middle-income countries. Food Nutr. Bull. 34,
to growth stunting and predisposes children glucagon in Jamaican children during malnutrition 357–364 (2013).
to severe malnutrition. and subsequent recovery. Pediatr. Res. 16, 99. Irena, A. H., Mwambazi, M. & Mulenga, V. Diarrhea
49. Owino, V. et al. Environmental enteric dysfunction 1011–1015 (1982). is a major killer of children with severe acute
and growth failure/stunting in global child health. 74. Jaya Rao, K. S., Srikantia, S. G. & Gopalan, C. malnutrition admitted to inpatient set‑up in Lusaka,
Pediatrics 138, e20160641 (2016). Plasma cortisol levels in protein-calorie malnutrition. Zambia. Nutr. J. 10, 110 (2011).
50. Naylor, C. & Petri, W. A. Jr. Leptin regulation of Arch. Dis. Child 43, 365–367 (1968). 100. Maitland, K. et al. Children with severe malnutrition:
immune responses. Trends Mol. Med. 22, 88–98 75. Paisley, R. B., Angers, M. & Frenk, S. Plasma cortisol can those at highest risk of death be identified with
(2016). levels in malnourished children with and without the WHO protocol? PLoS Med. 3, e500 (2006).

16 | ARTICLE NUMBER 17067 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

101. Talbert, A. et al. Diarrhoea complicating severe acute 124. Gomez, F. et al. Mortality in second and third degree 144. Vaivada, T., Gaffey, M. F., Das, J. K. & Bhutta, Z. A.
malnutrition in Kenyan children: a prospective malnutrition. J. Trop. Pediatr. (Lond.) 2, 77–83 (1956). Evidence-based interventions for improvement of
descriptive study of risk factors and outcome. 125. Waterlow, J. C. et al. The presentation and use of maternal and child nutrition in low-income settings:
PLoS ONE 7, e38321 (2012). height and weight data for comparing the nutritional what’s new? Curr. Opin. Clin. Nutr. Metab. Care 20,
102. Attia, S. et al. Mortality in children with complicated status of groups of children under the age of 10 years. 204–210 (2017).
severe acute malnutrition is related to intestinal and Bull. World Health Organ. 55, 489–498 (1977). 145. Bhutta, Z. A. et al. Evidence-based interventions for
systemic inflammation: an observational cohort study. 126. de Onis, M., Garza, C., Onyango, A. W. & Martorell, R. improvement of maternal and child nutrition: what
Am. J. Clin. Nutr. 104, 1441–1449 (2016). WHO child growth standards. Acta Paediatr. 450, can be done and at what cost? Lancet 382, 452–477
103. Bartels, R. H. et al. Both exocrine pancreatic S1–S101 (2006). (2013).
insufficiency and signs of pancreatic inflammation are 127. World Health Organization. Management of Severe This comprehensive review provided an essential
prevalent in children with complicated severe acute Malnutrition: A Manual for Physicians and Other data-driven overview of what interventions might
malnutrition: an observational study. J. Pediatr. 174, Senior Health Workers (WHO, 1999). be most cost-effective in decreasing the burden
165–170 (2016). 128. Pelletier, D. L. The relationship between child of maternal and child malnutrition globally
104. Zhang, L. et al. Impaired bile acid homeostasis in anthropometry and mortality in developing countries: and what the benefits might be on a global scale.
children with severe acute malnutrition. PLoS ONE 11, implications for policy, programs and future research. 146. Nabwera, H. M., Fulford, A. J., Moore, S. E.
e0155143 (2016). J. Nutr. 124, 2047S–2081S (1994). & Prentice, A. M. Growth faltering in rural
105. Schneider, R. E. & Viteri, F. E. Luminal events of lipid 129. Myatt, M., Khara, T. & Collins, S. A review of methods Gambian children after four decades of interventions:
absorption in protein-calorie malnourished children; to detect cases of severely malnourished children in a retrospective cohort study. Lancet Glob. Health 5,
relationship with nutritional recovery and diarrhea. II. the community for their admission into community- e208–e216 (2017).
Alterations in bile acid content of duodenal aspirates. based therapeutic care programs. Food Nutr. Bull. 27, 147. Ashorn, P. et al. Supplementation of maternal diets
Am. J. Clin. Nutr. 27, 788–796 (1974). S7–S23 (2006). during pregnancy and for 6 months postpartum and
106. Murphy, J. L. et al. Maldigestion and malabsorption 130. Briend, A., Khara, T. & Dolan, C. Wasting and infant diets thereafter with small-quantity lipid-based
of dietary lipid during severe childhood malnutrition. stunting — similarities and differences: policy nutrient supplements does not promote child growth
Arch. Dis. Child 87, 522–525 (2002). and programmatic implications. Food Nutr. Bull. 36, by 18 months of age in rural Malawi: a randomized
107. Kvissberg, M. A. et al. Carbohydrate malabsorption S15–S23 (2015). controlled trial. J. Nutr. 145, 1345–1353 (2015).
in acutely malnourished children and infants: This review highlighted the physiological, 148. Adu-Afarwuah, S. et al. Small-quantity, lipid-based
a systematic review. Nutr. Rev. 74, 48–58 (2016). epidemiological and clinical similarities and nutrient supplements provided to women during
108. Subramanian, S. et al. Cultivating healthy growth and differences between wasting and stunting, pregnancy and 6 mo postpartum and to their infants
nutrition through the gut microbiota. Cell 161, 36–48 emphasizing that an integrated and thoughtful from 6 mo of age increase the mean attained length
(2015). approach toward children with both was the most of 18‑mo‑old children in semi-urban Ghana:
This important review indicated that microbiota effective in decreasing morbidity and mortality. a randomized controlled trial. Am. J. Clin. Nutr. 104,
assembly is perturbed in children with malnutrition, 131. World Health Organization & UNICEF. WHO Child 797–808 (2016).
resulting in persistent microbiota immaturity that Growth Standards and the Identification of Severe 149. Adu-Afarwuah, S. et al. Lipid-based nutrient
is not rescued by current nutritional interventions Acute Malnutrition in Infants and Children (WHO, supplement increases the birth size of infants of
and could be related to the pathogenesis and 2009). primiparous women in Ghana. Am. J. Clin. Nutr. 101,
sequelae of malnutrition. 132. Briend, A., Maire, B., Fontaine, O. & Garenne, M. 835–846 (2015).
109. Smith, M. I. et al. Gut microbiomes of Malawian Mid‑upper arm circumference and weight-for-height 150. Hess, S. Y. et al. Small-quantity lipid-based nutrient
twin pairs discordant for kwashiorkor. Science 339, to identify high-risk malnourished under-five children. supplements, regardless of their zinc content, increase
548–554 (2013). Matern. Child Nutr. 8, 130–133 (2012). growth and reduce the prevalence of stunting and
110. Subramanian, S. et al. Persistent gut microbiota 133. Mwangome, M. K., Fegan, G., Mbunya, R., wasting in young Burkinabe children: a cluster-
immaturity in malnourished Bangladeshi children. Prentice, A. M. & Berkley, J. A. Reliability and randomized trial. PLoS ONE 10, e0122242 (2015).
Nature 510, 417–421 (2014). accuracy of anthropometry performed by community 151. World Health Organization. Essential Nutrition
111. Kristensen, K. H. et al. Gut microbiota in children health workers among infants under 6 months in rural Actions: Improving Maternal, Newborn, Infant and
hospitalized with oedematous and non-oedematous Kenya. Trop. Med. Int. Health 17, 622–629 (2012). Young Child Health and Nutrition (WHO, 2013).
severe acute malnutrition in Uganda. PLoS Negl. 134. ENN/UCL/ACF. MAMI Project. Technical review: 152. World Health Organization. WHO Recommendations
Trop. Dis. 10, e0004369 (2016). current evidence, policies, practices & programme on Antenatal Care for a Positive Pregnancy Experience
112. Reyes, A. et al. Gut DNA viromes of Malawian twins outcomes. ENN http://www.ennonline.net/ (WHO, 2016).
discordant for severe acute malnutrition. Proc. Natl mamitechnicalreview (2010). 153. Kramer, M. S. & Kakuma, R. Optimal duration of
Acad. Sci. USA 112, 11941–11946 (2015). 135. Kerac, M. et al. Prevalence of wasting among under exclusive breastfeeding. Cochrane Database Syst. Rev.
113. Klahr, S. & Alleyne, G. A. Effects of chronic protein- 6‑month-old infants in developing countries and 8, CD003517 (2012).
calorie malnutrition on the kidney. Kidney Int. 3, implications of new case definitions using WHO growth 154. World Health Organization. Towards a Grand
129–141 (1973). standards: a secondary data analysis. Arch. Dis. Child Convergence for Child Survival and Health: A Strategic
114. Gunston, G. D., Burkimsher, D., Malan, H. & Sive, A. A. 96, 1008–1013 (2011). Revie of Options for the Future Building on Lessons
Reversible cerebral shrinkage in kwashiorkor: an MRI This analysis brought important attention to the Learnt from IMNCI (WHO, 2016).
study. Arch. Dis. Child 67, 1030–1032 (1992). growing need to address malnutrition in children 155. Annan, R. A., Webb, P. & Brown, R. Management
115. Atalabi, O. M., Lagunju, I. A., Tongo, O. O. <6 months of age. of moderate acute malnutrition (MAM): Current
& Akinyinka, O. O. Cranial magnetic resonance 136. Lelijveld, N., Kerac, M., McGrath, M., Mwangome, M. Knowledge and Practice (CMAM Forum, 2014).
imaging findings in kwashiorkor. Int. J. Neurosci. 120, & Berkley, J. A. A Review of Methods to Detect 156. Picot, J. et al. The effectiveness of interventions
23–27 (2010). Cases of Severely Malnourished Infants Less Than to treat severe acute malnutrition in young children:
116. Grantham-McGregor, S., Powell, C., Walker, S., 6 Months for their Admission into Therapeutic Care a systematic review. Health Technol. Assess. 16,
Chang, S. & Fletcher, P. The long-term follow‑up of (ENN, 2017). 1–316 (2012).
severely malnourished children who participated in 137. Mwangome, M. K., Fegan, G., Fulford, T., 157. Tickell, K. D. & Denno, D. M. Inpatient management
an intervention program. Child Dev. 65, 428–439 Prentice, A. M. & Berkley, J. A. Mid-upper arm of children with severe acute malnutrition: a review
(1994). circumference at age of routine infant vaccination of WHO guidelines. Bull. World Health Organ. 94,
This landmark study demonstrated how children to identify infants at elevated risk of death: 642–651 (2016).
with severe malnutrition benefit from early and a retrospective cohort study in the Gambia. 158. Gross, R. & Webb, P. Wasting time for wasted
sustained psychosocial interventions in addition Bull. World Health Organ. 90, 887–894 (2012). children: severe child undernutrition must be resolved
to nutritional rehabilitation. 138. Mwangome, M. et al. Diagnostic criteria for severe in non-emergency settings. Lancet 367, 1209–1211
117. Nahar, B. et al. Effects of psychosocial stimulation acute malnutrition among infants aged under 6 mo. (2006).
on growth and development of severely malnourished Am. J. Clin. Nutr. 105, 1415–1423 (2017). 159. Murray, E. & Manary, M. Home-based therapy for
children in a nutrition unit in Bangladesh. Eur. J. Clin. 139. Briend, A. & Zimicki, S. Validation of arm severe acute malnutrition with ready‑to‑use food.
Nutr. 63, 725–731 (2009). circumference as an indicator of risk of death in one to Paediatr. Int. Child Health 34, 266–270 (2014).
118. Grantham-McGregor, S. et al. Developmental potential four year old children. Nutr. Res. 6, 249–261 (1986). This review provided historical context on the
in the first 5 years for children in developing countries. 140. Blackwell, N. et al. Mothers Understand And Can do it evolution and widespread acceptance of RUTFs in
Lancet 369, 60–70 (2007). (MUAC): a comparison of mothers and community making it possible to care for children with severe
119. Walker, S. P. et al. Child development: risk factors health workers determining mid-upper arm malnutrition in the community.
for adverse outcomes in developing countries. Lancet circumference in 103 children aged from 6 months 160. World Health Organization, World Food Programme,
369, 145–157 (2007). to 5 years. Arch. Public Health 73, 26 (2015). United Nations System Standing Committee on
120. Patrick, J. & Golden, M. Leukocyte electrolytes 141. Tam, Y., Huicho, L., Huayanay-Espinoza, C. A. Nutrition & United Nations Children’s Fund.
and sodium transport in protein energy malnutrition. & Restrepo-Mendez, M. C. Remaining missed Community-Based Management of Severe Acute
Am. J. Clin. Nutr. 30, 1478–1481 (1977). opportunities of child survival in Peru: modelling Malnutrition (WHO, 2007).
121. Lang, F. Mechanisms and significance of cell volume mortality impact of universal and equitable coverage 161. Oakley, E. et al. A ready‑to‑use therapeutic food
regulation. J. Am. Coll. Nutr. 26, 613S–623S (2007). of proven interventions. BMC Public Health 16, 1048 containing 10% milk is less effective than one with
122. World Health Organization Expert Committee. (2016). 25% milk in the treatment of severely malnourished
Physical status: the use and interpretation of 142. Monteiro, C. A. et al. Narrowing socioeconomic children. J. Nutr. 140, 2248–2252 (2010).
anthropometry. Geneva. WHO http://www.who.int/ inequality in child stunting: the Brazilian experience, 162. Bahwere, P. et al. Cereals and pulse-based
childgrowth/publications/physical_status/en/ 1974–2007. Bull. World Health Organ. 88, 305–311 ready‑to‑use therapeutic food as an alternative to the
index.html (1995). (2010). standard milk- and peanut paste-based formulation
123. Cederholm, T. et al. Diagnostic criteria for malnutrition 143. Huicho, L. et al. Factors behind the success story for treating severe acute malnutrition: a noninferiority,
— an ESPEN Consensus Statement. Clin. Nutr. 34, of under-five stunting in Peru: a district ecological individually randomized controlled efficacy clinical
335–340 (2015). multilevel analysis. BMC Pediatr. 17, 29 (2017). trial. Am. J. Clin. Nutr. 103, 1145–1161 (2016).

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17067 | 17


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

163. Jones, K. D. et al. Ready‑to‑use therapeutic food 187. Bahwere, P., Mtimuni, A., Sadler, K., Banda, T. the metabolic syndrome? Nutr. Rev. 70, 642–653
with elevated n-3 polyunsaturated fatty acid content, & Collins, S. Long term mortality after community and (2012).
with or without fish oil, to treat severe acute facility based treatment of severe acute malnutrition: 210. Martorell, R. Improved nutrition in the first 1000 days
malnutrition: a randomized controlled trial. BMC Med. analysis of data from Bangladesh, Kenya, Malawi and and adult human capital and health. Am. J. Hum. Biol.
13, 93 (2015). Niger. J. Public Health Epidemiol. 4, 215–225 (2012). 29, e22952 (2017).
164. Ferguson, E. L., Briend, A. & Darmon, N. Can optimal 188. Shewade, H. D. et al. Effectiveness of indigenous 211. Hamadani, J. D., Huda, S. N., Khatun, F. & Grantham-
combinations of local foods achieve the nutrient density ready‑to‑use therapeutic food in community-based McGregor, S. M. Psychosocial stimulation improves
of the F100 catch‑up diet for severe malnutrition? management of uncomplicated severe acute the development of undernourished children in rural
J. Pediatr. Gastroenterol. Nutr. 46, 447–452 (2008). malnutrition: a randomized controlled trial from India. Bangladesh. J. Nutr. 136, 2645–2652 (2006).
165. Manary, M. J., Ndkeha, M. J., Ashorn, P., Maleta, K. J. Trop. Pediatr. 59, 393–398 (2013). 212. Walker, S. P., Chang, S. M., Powell, C. A., Simonoff, E.
& Briend, A. Home based therapy for severe 189. Smith, S. E., Ramos, R. A., Refinetti, R., Farthing, J. P. & Grantham-McGregor, S. M. Effects of psychosocial
malnutrition with ready‑to‑use food. Arch. Dis. Child & Paterson, P. G. Protein-energy malnutrition induces stimulation and dietary supplementation in early
89, 557–561 (2004). an aberrant acute-phase response and modifies the childhood on psychosocial functioning in late
166. Bandsma, R. H. et al. Impaired glucose absorption circadian rhythm of core temperature. Appl. Physiol. adolescence: follow‑up of randomised controlled trial.
in children with severe malnutrition. J. Pediatr. 158, Nutr. Metab. 38, 844–853 (2013). BMJ 333, 472 (2006).
282–287.e1 (2011). 190. World Health Organization. Pocket Book of Hospital 213. Grantham-McGregor, S. A review of studies of the
167. Nyeko, R., Kalyesubula, I., Mworozi, E. & Bachou, H. Care for Children: Guidelines for the Management effect of severe malnutrition on mental development.
Lactose intolerance among severely malnourished of Common Illnesses with Limited Resources J. Nutr. 125, 2233S–2238S (1995).
children with diarrhoea admitted to the nutrition unit, (WHO, 2013). 214. Every Woman Every Child. The global strategy for
Mulago hospital, Uganda. BMC Pediatr. 10, 31 (2010). 191. Chimhuya, S., Kambarami, R. A. & Mujuru, H. women’s, children’s and adolescents’ health
168. Rytter, M. J. et al. Risk factors for death in children The levels of malnutrition and risk factors for mortality (2016–2030). Every Woman Every Child http://
during inpatient treatment of severe acute at Harare Central Hospital-Zimbabwe: an observation globalstrategy.everywomaneverychild.org/pdf/
malnutrition: a prospective cohort study. Am. J. Clin. study. Cent. Afr. J. Med. 53, 30–34 (2007). EWEC_globalstrategyreport_200915_FINAL_WEB.pdf
Nutr. 105, 494–502 (2017). 192. Fergusson, P., Tomkins, A. & Kerac, M. Improving (2016).
169. Vemula, P., Abela, O. G., Narisetty, K., Rhine, D. survival of children with severe acute malnutrition 215. Action Against Hunger. No wasted lives. Action
& Abela, G. S. Potassium toxicity at low serum in HIV-prevalent settings. Int. Health 1, 10–16 (2009). Against Hunger https://www.actionagainsthunger.org.
potassium levels with refeeding syndrome. 193. Heikens, G. T. et al. Case management of HIV-infected uk/blog/no-wasted-lives (2017).
Am. J. Cardiol. 115, 147–149 (2015). severely malnourished children: challenges in the area 216. Kerac, M., Mwangome, M., McGrath, M., Haider, R.
170. Chisti, M. J., Salam, M. A., Sharifuzzaman of highest prevalence. Lancet 371, 1305–1307 & Berkley, J. A. Management of acute malnutrition
& Pietroni, M. A. Occult pneumonia: an unusual but (2008). in infants aged under 6 months (MAMI): current
perilous entity presenting with severe malnutrition 194. Brewster, D. R., Manary, M. J. & Graham, S. M. Case issues and future directions in policy and research.
and dehydrating diarrhoea. J. Health Popul. Nutr. 27, management of kwashiorkor: an intervention project Food Nutr. Bull. 36, S30–S34 (2015).
808–812 (2009). at seven nutrition rehabilitation centres in Malawi. 217. Groce, N. E., Kerac, M., Farkas, A., Schultink, W.
171. Jones, K. D. J. & Berkley, J. A. Severe Acute Eur. J. Clin. Nutr. 51, 139–147 (1997). & Bieler, R. B. Inclusive nutrition for children
Malnutrition and Infection (CMAM Forum, 2013). 195. Bachou, H., Tumwine, J. K., Mwadime, R. K., Ahmed, T. and adults with disabilities. Lancet Glob. Health 1,
172. Williams, P. C. M. & Berkley, J. A. Severe Acute & Tylleskar, T. Reduction of unnecessary transfusion e180–181 (2013).
Malnutrition Update: Current WHO Guidelines and intravenous fluids in severely malnourished 218. Angood, C., Khara, T., Dolan, C., Berkley, J. A.
and the WHO Essential Medicine List for Children children is not enough to reduce mortality. Ann. Trop. & WaSt Technical Interest, G. Research priorities
(WHO, 2016). Paediatr. 28, 23–33 (2008). on the relationship between wasting and stunting.
173. Isanaka, S. et al. Routine amoxicillin for uncomplicated 196. Manary, M. J. & Brewster, D. R. Intensive nursing PLoS ONE 11, e0153221 (2016).
severe acute malnutrition in children. N. Engl. J. Med. care of kwashiorkor in Malawi. Acta Paediatr. 89, 219. Wazny, K. et al. Setting global research priorities
374, 444–453 (2016). 203–207 (2000). for integrated community case management (iCCM):
174. Million, M., Lagier, J. C. & Raoult, D. Meta-analysis 197. Angood, C. et al. Research priorities to improve the results from a CHNRI (Child Health and Nutrition
on efficacy of amoxicillin in uncomplicated severe acute management of acute malnutrition in infants aged less Research Initiative) exercise. J. Glob. Health 4,
malnutrition. Microb. Pathog. 106, 76–77 (2017). than six months (MAMI). PLoS Med. 12, e1001812 020413 (2014).
175. Waterlow, J. Treatment of children with malnutrition (2015). 220. Garenne, M. et al. Incidence and duration of severe
and diarrhoea. Lancet 354, 1142 (1999). 198. Lazzaroni, S. & Wagner, N. Misfortunes never come wasting in two African populations. Public Health Nutr.
176. Brewster, D. R. Inpatient management of severe singly: structural change, multiple shocks and child 12, 1974–1982 (2009).
malnutrition: time for a change in protocol and malnutrition in rural Senegal. Econ. Hum. Biol. 23, 221. Hure, A., Oldmeadow, C. & Attia, J. Invited
practice. Ann. Trop. Paediatr. 31, 97–107 (2011). 246–262 (2016). commentary: improving estimates of severe acute
177. Akech, S. et al. Volume expansion with albumin 199. Chisti, M. J. et al. Post-discharge mortality in children malnutrition requires more data. Am. J. Epidemiol.
compared to gelofusine in children with severe with severe malnutrition and pneumonia in 184, 870–872 (2016).
malaria: results of a controlled trial. PLoS Clin. Trials Bangladesh. PLoS ONE 9, e107663 (2014). 222. Emergency Nutrition Network. Caseload calculations
1, e21 (2006). 200. Chang, C. Y. et al. Children successfully treated for (incidence rate). ENN http://www.en-net.org/
178. Akech, S. O., Karisa, J., Nakamya, P., Boga, M. moderate acute malnutrition remain at risk for question/514.aspx (2011).
& Maitland, K. Phase II trial of isotonic fluid malnutrition and death in the subsequent year after 223. Grellety, E. & Golden, M. H. Weight-for-height
resuscitation in Kenyan children with severe recovery. J. Nutr. 143, 215–220 (2013). and mid-upper-arm circumference should be used
malnutrition and hypovolaemia. BMC Pediatr. 10, 71 This trial demonstrated that children with moderate independently to diagnose acute malnutrition: policy
(2010). malnutrition who appear to achieve nutritional implications. BMC Nutr. 2, 10 (2016).
179. Maitland, K. et al. Mortality after fluid bolus in African recovery still have high rates of morbidity and 224. Briend, A. et al. Low mid-upper arm circumference
children with severe infection. N. Engl. J. Med. 364, mortality in the year following discharge. identifies children with a high risk of death who should
2483–2495 (2011). 201. Chuma, J. & Maina, T. Catastrophic health care be the priority target for treatment. BMC Nutr. 2, 63
180. Grenov, B. et al. Effect of probiotics on diarrhea in spending and impoverishment in Kenya. BMC Health (2016).
children with severe acute malnutrition: a randomized Serv. Res. 12, 413 (2012). 225. Friedli, N. et al. Revisiting the refeeding syndrome:
controlled study in Uganda. J. Pediatr. Gastroenterol. 202. Morgan, G. What, if any, is the effect of malnutrition Results of a systematic review. Nutrition 35, 151–160
Nutr. 64, 396–403 (2017). on immunological competence? Lancet 349, (2017).
181. Velly, H., Britton, R. A. & Preidis, G. A. Mechanisms of 1693–1695 (1997). This comprehensive review provided an excellent
cross-talk between the diet, the intestinal microbiome, 203. Kerac, M. et al. Follow‑up of post-discharge growth overview of refeeding syndrome.
and the undernourished host. Gut Microbes 8, and mortality after treatment for severe acute
98–112 (2017). malnutrition (FuSAM study): a prospective cohort Acknowledgements
182. Jones, K. D. J. et al. Mesalazine in the initial study. PLoS ONE 9, e96030 (2014). The authors are grateful to Grace Belayneh for her thoughtful
management of severely acutely malnourished 204. Lelijveld, N. et al. Chronic disease outcomes after assistance in the coordination of this work and facilitation of
children with environmental enteric dysfunction: severe acute malnutrition in Malawian children communication among the writing group.
a pilot randomized controlled trial. BMC Med. 12, (ChroSAM): a cohort study. Lancet Glob. Health 4,
133 (2014). e654–e662 (2016). Author contributions
183. Lima, A. A. et al. Effects of glutamine alone or in 205. Jackson, A. A. Protein requirements for catch‑up Introduction (Z.A.B.); Epidemiology (M.K. and Z.A.B.);
combination with zinc and vitamin A on growth, growth. Proc. Nutr. Soc. 49, 507–516 (1990). Mechanisms/pathophysiology (R.H.B. and J.A.B.); Diagnosis,
intestinal barrier function, stress and satiety-related 206. Benefice, E., Fouere, T., Malina, R. M. & Beunen, G. screening and prevention (A.B., I.T. and M.K.); Management
hormones in Brazilian shantytown children. Clinics Anthropometric and motor characteristics of (J.A.B. and I.T.); Quality of life (J.A.B.); Outlook (Z.A.B., M.K.
(Sao Paulo) 69, 225–233 (2014). Senegalese children with different nutritional histories. and A.B.); Overview of Primer (Z.A.B.).
184. Trehan, I. & Manary, M. J. Management of severe Child Care Health Dev. 22, 151–165 (1996).
acute malnutrition in low-income and middle-income 207. Fall, C. & Osmond, C. Commentary: the developmental Competing interests
countries. Arch. Dis. Child 100, 283–287 (2015). origins of health & disease: an appreciation of the life The authors declare no competing interests.
185. Aiken, A. M. et al. Risk and causes of paediatric & work of Professor David, J. P. Barker, 1938–2013.
hospital-acquired bacteraemia in Kilifi District Int. J. Epidemiol. 42, 1231–1232 (2013). Publisher’s note
Hospital, Kenya: a prospective cohort study. Lancet 208. Gluckman, P. D., Hanson, M. A. & Pinal, C. The Springer Nature remains neutral with regard to jurisdictional
378, 2021–2027 (2011). developmental origins of adult disease. Matern. Child claims in published maps and institutional affiliations.
186. Bachmann, M. O. Cost-effectiveness of community- Nutr. 1, 130–141 (2005).
based treatment of severe acute malnutrition in 209. DeBoer, M. D. et al. Early childhood growth failure How to cite this article
children. Expert Rev. Pharmacoecon. Outcomes Res. and the developmental origins of adult disease: do Bhutta, Z. A. et al. Severe childhood malnutrition. Nat. Rev.
10, 605–612 (2010). enteric infections and malnutrition increase risk for Dis. Primers 3, 17067 (2017).

18 | ARTICLE NUMBER 17067 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like