The Human Microbiome
3695CHAPTER 471
1950
100
200
300
400 Crohn’s
disease
Multiple
sclerosis
Asthma
Measles
Mumps
Tuberculosis
Hepatitis A
Rheumatic
fever
Type 1
diabetes
0
50
100
Incidence of immune disorders (%)
Incidence of infectious diseases (%)
A 1950 1960 1970 1980 1990 2000 B 1960 1970 1980 1990 2000
FIGURE 471-5 There was an inverse relationship between the incidence of select infectious diseases and the
incidence of autoimmune disorders during the latter half of the twentieth century. A. Relative incidence of prototypical
infectious diseases from 1950 to 2000. B. Relative incidence of select autoimmune disorders from 1950 to 2000. (From
JF Bach: The effect of infections on susceptibility to autoimmune and allergic diseases. N Engl J Med 347:911, 2002.
Copyright © 2002, Massachusetts Medical Society. Reprinted with permission from Massachusetts Medical Society.)
exposures are required to prevent subsequent disease and that the
“westernization” of society has led to a decrease in such exposures. This
concept is now being applied beyond atopic diseases to other inflammatory and autoimmune diseases and is thought to reflect processes
that occur in later life as well.
■ RELATIONSHIP BETWEEN THE MICROBIOTA AND
SPECIFIC DISEASE STATES
The ideas inherent in the hygiene hypothesis—in sum, that microbial
exposure can affect long-term health outcomes—laid the theoretical foundation for translational microbiome studies. While most
of the studies described earlier sought to describe how the microbiota responds to specific and often transient influences (e.g., a
course of antibiotics, dietary interventions, travel), a multitude of
studies have characterized the microbiota in patients with various
diseases in the hope that a better understanding of the nature of
disease-specific microbial communities will provide insight into disease pathogenesis and potentially uncover novel treatment modalities.
Remarkably, virtually all of these studies have demonstrated differences
between the microbiotas of healthy controls and patients, irrespective
of the specific disease process examined. Although it is difficult to
generalize across all studies, a couple of general themes have emerged.
First, disease states are typically associated with microbiotas that are
less diverse than those of healthy individuals. This loss of diversity
can be measured either as a decrease in the number of species (alpha
diversity; often measured as the number of operational taxonomic units
or amplicon sequence variants, which are the bioinformatic equivalent
of species) or as a reduction in the microbial relatedness of the species
present (beta diversity). Often, both alpha and beta diversity decrease
in the setting of disease. Second, states of inflammation—regardless of
site or underlying disease process—are often associated with a relative
increase in the abundance of the bacterial family Enterobacteriaceae
and a decrease in the abundance of Lachnospiraceae.
Dissecting Correlation and Causality Given that most of these
investigations have been designed as case-control studies, it is difficult
to determine whether microbiologic findings are the cause or the effect
of the disease. Even studies that examine treatment-naïve patients at
the time of initial diagnosis are still confounded by this “chicken or
egg” issue. Moreover, prospective, longitudinal clinical studies—still
rare in the microbiome field—may simply yield correlations between
the microbiome and subclinical disease rather than necessarily proving causality. Experiments in animals—specifically, studies using
gnotobiotic mice (germ-free mice that have been colonized with
specified microbial communities)—have been critical in this regard
as they allow investigation of specific differences in microbial components while controlling for the host’s genetics, diet, and housing
conditions. Moreover, human microbes
can be transplanted into gnotobiotic mice
to permit in-depth mechanistic studies of
how these microbial communities affect
disease pathogenesis. This marriage of
human samples and animal experiments
has facilitated the identification of causal
roles played by some microbes in disease
pathogenesis; these findings provide a
critical proof of concept for the interplay
of the microbiota with human health.
However, the vast majority of microbiome studies are still at the level of correlation. The next several sections describe
the clinical and animal data for many
different disease processes. Given the
voluminous and rapidly changing nature
of this field, it is impossible to cover
all of the disease associations known
to date; rather, the following discussion
represents a combination of the leading exemplars of microbiome data and
nascent areas of significant clinical interest. In all cases, the hope is that
further study of the role of the microbiota will provide novel diagnostics, new therapeutic modalities, and/or additional insight into disease
pathogenesis.
Gastrointestinal Diseases Given that the intestines harbor the
largest number and greatest diversity of organisms in the body, much
work has focused on how the microbiota impacts gastrointestinal
diseases. Even though the luminal surface area of the gastrointestinal
tract is 30–40 square meters (~90% of which is contained within the
small intestine) and features marked anatomic and functional differences that result in many discrete macro- and micro-ecosystems, stool
is often used as a surrogate for the intestinal microbiota given the
relative ease of collecting samples. A few studies that have compared
the microbial profile in stool with the mucosa-adherent organisms
present in biopsy samples have demonstrated that stool is, in fact, a
reasonable proxy for biopsy samples; however, the relative microbial
“noise” present in stool can sometimes overwhelm the “signal,” making
biopsy samples more informative for some scientific questions. The key
issue is to ensure that the biopsy samples evaluated represent relatively
similar intestinal regions, as there are significant differences between
the organisms present in the crypt and the tip of the villus and between
microbes found in the ascending versus the descending colon.
OBESITY Obesity is a worsening epidemic throughout the world, and
multiple studies have linked the composition of the intestinal microbiota
to the development of obesity in animal models and in humans. Indeed,
many of the initial translational microbiome studies performed in mice
at the beginning of the twenty-first century focused on obesity. These
early studies suggested that the ratio of the relative abundance of Bacteroidetes to Firmicutes was lower in obese mice than in control animals.
Moreover, a causal relationship between the microbiota and obesity
was established by the finding that gnotobiotic mice colonized with the
microbiota from obese individuals had more rapid and more extensive
weight gain than gnotobiotic mice colonized with the microbiota from
lean individuals. Biologically, it is posited on the basis of metagenomic
surveys that the obesity-associated microbiome has an increased capacity to harvest energy from the diet. Notably, the relationship between the
Bacteroidetes/Firmicutes ratio and obesity did not hold in initial human
studies; however, the finding that this ratio increased in obese patients
who lost weight while on a fat- or carbohydrate-restricted diet suggested
some generalizability between mice and humans. Beyond this ratio of
major bacterial phyla, obesity was linked to a microbiome with a lower
alpha diversity. Over the past ~15 years, numerous human studies
examining the relationship between the microbiome and obesity have
been completed, all with mixed results. A recent meta-analysis of 10
studies including nearly 3000 individuals revealed an apparent lack of
relationship between the Bacteroidetes/Firmicutes ratio and obesity,
3696 PART 16 Genes, the Environment, and Disease
though there is ~2% lower diversity associated with obesity that is
statistically significant but of unclear biologic significance. This finding
highlights a problem common to microbiome studies: i.e., there is no
sense as to what magnitude of change is biologically meaningful. Ultimately, although murine studies have indicated a causal link between
the microbiota and obesity, the human data are less convincing, and
their significance may be limited because the studies primarily examined only high-level taxonomic information rather than also assessing
transcriptional or metabolic differences.
The rise in obesity has elicited a plethora of ideas about the type of
diet that might be most successful in leading to sustained weight loss.
It has become clear that the same dietary ingredient can have highly
diverse effects on blood glucose measurements in different people and
that this effect is mediated largely by the microbiome. These observations suggest that the “optimal” diet needs to be individualized in
the context of the person’s microbiome, which itself may continue to
change over the course of the diet. An intriguing parallel question is
whether the microbiota may also influence dietary preferences; such
an influence would suggest important feedback loops between the
microbiome and diet.
MALNUTRITION Representing the other end of the metabolic spectrum from obesity, malnutrition is also linked to an altered microbiome. Analysis of Malawian twin pairs (≤3 years of age) who were
discordant for kwashiorkor—a severe form of malnutrition—revealed
that kwashiorkor is associated with a microbiologically “immature”
fecal microbiota that resembles that of a chronologically younger child.
Transplantation of the fecal microbiota from these discordant twins
into gnotobiotic mice that were fed a diet similar in composition to
a typical Malawian diet established that the kwashiorkor-associated
microbiome is causally related to poor weight gain. Subsequent studies
demonstrated these same general trends in malnourished Bangladeshi children. Investigators were able to identify five bacterial species (Faecalibacterium prausnitzii, Ruminococcus gnavus, Clostridium
nexile, Clostridium symbiosum, and Dorea formicigenerans) that—
when administered together as a “cocktail” to mice colonized with a
kwashiorkor-associated microbiome—were able to prevent growth
impairments. Moreover, children with moderate acute malnutrition
fed therapeutic food purposefully designed for its ability to alter the
microbiota in defined manners have altered serum biomarkers consistent with improved growth. These results demonstrate that rationally
designed modulation of the microbiota may lead to improved health
outcomes.
INFLAMMATORY BOWEL DISEASE Ulcerative colitis and Crohn’s disease, the two predominant forms of inflammatory bowel disease (IBD),
are chronic gastrointestinal inflammatory conditions that differ in their
locations and patterns of inflammation (Chap. 326). The following
observations have led to the suggestion that IBD is the result of an
immune response to a dysbiotic microbiota in a genetically susceptible
individual: genes account for only ~20% of susceptibility to IBD (and
many of the relevant genes are related to host–microbe interactions),
antibiotic treatment reduces the clinical severity of disease, and relapses
of Crohn’s disease are prevented by diversion of the fecal stream. While
the microbiota clearly is not the only driver of disease, it is considered
to be an important element. Accordingly, numerous animal and clinical
studies have been designed to tease out the nature of the relationship
between the microbiota and IBD.
Most of these studies have focused on comparing the microbiome’s
composition in IBD patients with that in healthy controls, concentrating on microbial diversity and specific bacterial taxa that are associated
with health or disease. Unfortunately, few, if any, results have been
universally obtained, probably because of differences in study design,
inclusion criteria, and methodology (e.g., the use of stool, rectal swabs,
or biopsy samples; the choice of sequencing primers; the analysis
pipeline). Even with these differences among studies, patients with
IBD have been shown typically to have reduced alpha and beta diversity in their fecal microbiotas. Moreover, Clostridium clusters IV and
XIVa, which are polyphyletic and encompass several different bacterial
families, are generally reduced in patients with IBD. F. prausnitzii is a
notable example from Clostridium cluster IV that is often underrepresented in the stool of patients who have Crohn’s disease, with more
mixed results in biopsy samples. The bacterial family Lachnospiraceae,
which is largely contained in Clostridium cluster XIVa, and other
butyrate-producing organisms are also reduced in the stool of patients
with IBD. Some of these species produce butyrate by using acetate
generated by other members of the microbiome, and some of these acetate-producing species are similarly reduced (e.g., Ruminococcus albus).
These complex interactions and dependencies among bacterial species
pose unique challenges to definitive ascertainment of the cause–effect
relationships between microbes and disease. Even before researchers
were able to assess the entire microbiome at once, they often noted that
patients with Crohn’s disease had a higher representation of adherent
invasive E. coli in the ileal mucosa, an observation consistent with the
increased abundance of Enterobacteriaceae seen in more recent microbiome studies. Beyond bacteria, burgeoning evidence supports a role
for Caudovirales bacteriophages in IBD pathogenesis, though these
findings may merely reflect the underlying dysbiosis related to the loss
of bacterial diversity in IBD. Moreover, some data suggest that IBD
is also associated with fungal dysbiosis; several studies have demonstrated an increased ratio of Basidiomycota to Ascomycota. It is still
unclear whether any of these microbial associations reflect the cause of
IBD or merely serve as biomarkers of disease.
Studies of antibiotic-treated mice and gnotobiotic mice colonized
with IBD-associated microbiotas have been useful in confirming that
the microbiota affects colitis severity. Several bacterial species have
been identified as either promoting colitis in mice (e.g., Klebsiella
pneumoniae, Prevotella copri) or protecting against it (e.g., Bacteroides
fragilis, Clostridium species); however, these organisms do not always
correlate with the taxa identified as differentially abundant across
multiple clinical studies. In contrast, IgA-coated commensal organisms
isolated from patients with IBD promote more severe colitis in mice
than either IgA-uncoated bacteria from patients with IBD or IgAcoated bacteria from healthy controls. These data suggest that functional categorization of the microbiota based on immune recognition
(e.g., IgA coating) may be a useful approach for identifying pathogenic
organisms.
Cardiovascular Disease Inflammation helps drive the pathogenesis of atherosclerosis, and it has long been postulated that microbes
are involved in the atherosclerotic process. Early work demonstrated
that patients with cardiovascular disease have higher titers of antibody
to Chlamydia pneumoniae than control patients, that C. pneumoniae is
present within atherosclerotic lesions, and that C. pneumoniae can both
initiate and exacerbate atherosclerotic lesions in animal models. This
type of analysis has been extended to other bacteria, such as Porphyromonas gingivalis, with the idea that multiple different bacteria may
play some role in the pathogenesis of atherosclerosis.
More recent studies have demonstrated clinical correlations between
serum levels of trimethylamine N-oxide (TMAO) and atherosclerotic
heart disease. Given that red meat, eggs, and dairy products are important sources of carnitine and choline (both precursors of TMAO), it is
not surprising that levels of TMAO are higher in omnivores than in
vegans. Animal studies have confirmed that transfer of the gut microbiota from atherosclerosis-susceptible strains of mice to atherosclerosisresistant animals leads to increased serum levels of TMAO and a
dietary choline-dependent increase in atherosclerotic plaques; this
observation confirms the role of the gut microbiota in the generation
of TMAO and atherosclerosis. Moreover, treatment of atherosclerosissusceptible strains of mice with a structural analogue of choline that
inhibits the first enzymatic step in TMAO formation leads to decreased
circulating TMAO levels and, more importantly, restrains macrophage
foam-cell formation and atherosclerotic lesion development. In a
study of >4000 patients, plasma TMAO levels were also predictive
of incident thrombosis risk (myocardial infarction, stroke). Gnotobiotic animals were used to demonstrate that this risk was dependent
on the microbiota; although eight bacterial taxa were identified as
being associated with both plasma TMAO levels and thrombotic risk,
organisms with choline-utilization genes that represent the first step
The Human Microbiome
3697CHAPTER 471
of TMAO production were not more abundant in animals at greater
risk for thrombosis. This discrepancy highlights the complexity of the
microbiota and suggests that other aspects of the overall dynamics of
the microbial community may be in play.
Oncology Recent studies exploring the link between the microbiota
and cancer have demonstrated that specific members of the microbiota
can affect treatment efficacy in both a positive and a negative manner.
For example, therapy with antibody to programmed cell death ligand
1 (anti-PD-L1) has proven highly effective for a number of different
cancers (Chap. 73); however, a significant proportion of patients do not
respond even when their tumors have high PD-L1 expression levels, a
prerequisite for this type of checkpoint blockade inhibition. Three groups
have independently performed clinical studies—sometimes coupled
with gnotobiotic mouse experiments to verify causal relationships—to
demonstrate that specific bacteria can potentiate checkpoint blockade
inhibition in melanoma, non-small-cell lung cancer, and renal cell
carcinoma. Intriguingly, these groups identified different bacteria
(Bifidobacterium, Faecalibacterium, and Akkermansia species) as being
associated with the anticancer effects, even when the same oncologic
process was being studied. The biologic factors driving these differences are not yet clear but may relate to differences in adjunctive therapies, geography, and/or other as-of-yet unidentified factors. Although
these seemingly disparate findings raise concern about the generalizability of microbiome studies, it may be that identifying relevant bacterial species—as opposed to their bioactive molecules—does not offer
sufficient granularity for comparison across studies.
In a separate set of studies, the efficacy of therapy with antibody
to cytotoxic T lymphocyte–associated antigen 4 (anti-CTLA-4) was
associated with T-cell responses specific for either Bacteroides thetaiotaomicron or B. fragilis. In particular, administration of B. fragilis to
germ-free or antibiotic-treated mice restored the normally absent
anticancer response to anti-CTLA-4 therapy. While these examples
demonstrate potentiation of anticancer therapies by the microbiota,
other therapies can be antagonized. Some cancers, such as pancreatic
ductal adenocarcinoma, contain intratumoral bacteria, particularly
Gammaproteobacteria, that can metabolize the chemotherapeutic
agent gemcitabine and thereby contribute to the drug resistance of
these tumors. Overall, these examples highlight the microbiota’s critical impact—both direct and indirect—on the efficacy of drugs. Many
other notable examples have been described (e.g., involving cyclophosphamide, digoxin, levodopa, and sulfasalazine), and many more likely
remain to be discovered.
The application of microbiome science to hematopoietic stem cell
transplantation (HSCT) is an area of expanding interest, particularly
given the significant morbidity and mortality related to graft-versus-host
disease (GVHD). In light of studies in the 1970s showing that germ-free
mice developed less frequent and less severe gut GVHD than wild-type
mice, clinicians began to use antibiotics to decontaminate the gut of
patients undergoing HSCT. This decontamination approach yielded
mixed results, probably because of differences in the antibiotic regimens
used. The natural history of patients undergoing allogeneic HSCT
includes a substantial loss of diversity in the fecal microbiota, intestinal
domination (≥30% abundance in the fecal microbiota) by Enterococcus species and other pathogens, and increased mortality. Moreover,
a retrospective analysis of ~850 patients undergoing allogeneic HSCT
revealed that receipt of imipenem-cilastatin or piperacillin-tazobactam
for neutropenic fever was associated with increased GVHD-related
mortality at 5 years; this observation suggested that specific bacteria
may help protect against GVHD-related mortality. More detailed analyses revealed an association between the abundance of Blautia species
and protection against GVHD and mortality, though this correlation is
still being examined with regard to its causal relationship. Despite significant interest in examining these microbial relationships with HSCT,
little has yet been studied in the context of solid organ transplantation,
which likely represents the next frontier of transplantation-related
microbiome investigation.
Autoimmune Diseases The dramatic rise in the incidence of
many autoimmune diseases over the past few decades has been far
more rapid than can be explained simply by genetic factors (Fig. 471-5).
It is increasingly thought that environmental triggers, including the
microbiome, are partially responsible for the development of these
autoimmune diseases.
TYPE 1 DIABETES Type 1 diabetes (T1D) is an autoimmune disorder
characterized by T cell–mediated destruction of insulin-producing
pancreatic islets (Chap. 403). There is a clear genetic predisposition for
the disease: ~70% of patients with T1D have human leukocyte antigen
(HLA) risk alleles. However, only 3–7% of children with these risk
alleles actually develop disease, an observation that suggests a role for
other environmental factors. Studying a prospective, densely sampled,
longitudinal cohort of at-risk, HLA-matched children from Finland
and Estonia, investigators detailed changes in the microbiota prior to
development of disease. Although only 4 of the 33 children studied
developed T1D within the time frame of the study, a marked decrease
of ~25% in alpha diversity occurred after seroconversion but before
disease diagnosis. The low number of cases in this study unfortunately
precluded identification of any specific disease-associated taxa. A
follow-up study compared the microbiomes of a larger cohort of these
high-risk northern European children with those of low-risk Russian
children who lived in geographic proximity. Bacteroides species were
more abundant in the high-risk group than in the low-risk group, particularly at early ages. This difference was postulated to be associated
with an altered structure of the bacterial lipopolysaccharide to which
children were exposed at a young age. It was further suggested that
Bacteroides-derived lipopolysaccharide was not able to provide the
immunogenic stimulus necessary to prevent T1D. These two studies
offer attractive—though logistically complicated—options for future
clinical investigations aimed at exploring the role of the microbiome.
The first approach—longitudinally following individuals who are at
high risk for a given disease—may provide insight into host–microbe
relationships by mapping temporal changes in the microbiome with
disease onset. An important caveat with this type of study, though, is
that the associations identified may reflect preclinical disease rather
than specifically indicating causality for any observed changes. The
second approach illustrates how careful selection of study participants
may offer an opportunity to uncover more meaningful associations
that can subsequently be experimentally verified.
RHEUMATOID ARTHRITIS Similar to many other autoimmune diseases, rheumatoid arthritis (RA) is a multifactorial disease that comes
to clinical attention after an environmental factor triggers symptoms
in an individual with preexisting autoantibodies. Multiple lines of
evidence support the notion that RA pathogenesis is reliant on the
microbiota, including the findings that germ-free mice do not develop
symptoms in several RA models and that antibiotic treatment of mice
mitigates against RA development. Several taxa (e.g., Bacteroides species, Lactobacillus bifidus, and segmented filamentous bacteria) have
been implicated in promoting RA in murine models, and analysis of
the fecal microbiota of patients with newly diagnosed RA has indicated
that P. copri is a biomarker of disease. That this association with P. copri
does not exist for chronic, treated RA or for psoriatic arthritis suggests
some specificity for new-onset RA. A major limitation of this approach
is that the identified association is shown to be a biomarker of disease
(and, in this case, potentially of response to treatment), but no added
insight is gained into a possible causal relationship between P. copri and
RA. In fact, many of the patients with new-onset RA had no Prevotella
detected, and several of the healthy controls had significant levels of
Prevotella. The lack of a strict concordance between the presence (or
absence) of a specific taxon and a given disease state argues against a
possible causal role.
MULTIPLE SCLEROSIS Epidemiologic studies of twin pairs and at-risk
individuals moving between high- and low-risk geographic areas
indicate that genetics plays a minor component in multiple sclerosis
(MS) susceptibility relative to environmental factors. For example, in
monozygotic twin pairs in which one sibling has MS, the other sibling
also develops MS in only ~30% of cases. Although MS is a disease of
the central nervous system (CNS), there is growing evidence of a link
between MS and the microbiota, specifically that of the gut. Germ-free
3698 PART 16 Genes, the Environment, and Disease
animals and antibiotic-treated animals display reduced disease incidence and severity in an MS model. Similarly, some clinical studies
suggest improved disease outcomes in patients with MS who have been
treated with minocycline, while patients treated with long-term penicillin appear to have an increased disease risk. Although several studies
have compared the fecal microbiotas of healthy controls to those of
patients with MS, these studies have all been relatively small and have
yielded few results (if any) that are common throughout. Although
work relating the microbiome to MS is ongoing, it has opened the
door to exploring this link with other neurologic diseases. Already,
there are animal data demonstrating links between the microbiota
and both Parkinson’s disease and autism, and there are clinical data
assessing fecal microbiomes in relation to a variety of neurologic conditions. It is not quite clear how the gut microbiota is communicating
with the CNS—i.e., whether communication takes place via bacterial
metabolites that travel in the bloodstream and cross the blood-brain
barrier, via migration of whole organisms into the CNS, or via feedback
through the vagus nerve. Emerging data suggest that a subset of enteroendocrine cells in the intestinal epithelium is synaptically connected
to the CNS, which may provide another means for the gut microbiota
to impact neurologic function. Although our understanding of this
brain-gut axis is still in its infancy, research in this area has elicited
tremendous excitement as a tractable approach to potential treatments
for these challenging diseases.
Atopic Diseases The incidence and prevalence of allergic diseases
continue to steadily increase, as do more severe clinical presentations.
Life-threatening food allergies are now such a public health issue that
nut-free classrooms are the norm in many cities. The development of
allergic diseases often follows a stereotyped progression that begins
with atopic dermatitis (AD) and continues, in order, with food allergy,
asthma, and allergic rhinitis. The microbiome has been linked to all of
these conditions and has the potential to modulate effects anywhere
along this spectrum.
ATOPIC DERMATITIS The skin is the largest organ in the body, and
its different anatomic sites (e.g., antecubital fossa, volar forearm, alar
crease) represent distinct ecologic niches and harbor unique microbial
communities. Moreover, given that the skin serves as a critical interface between the body and the external environment (e.g., microbes),
it must be able to respond to unwanted microbes with an adequate
immune response. AD is an inflammatory skin disorder involving
immune dysfunction and a dysbiotic skin microbiota that is typically
marked by greater abundances of Staphylococcus aureus and a lesser
degree of bacterial diversity. Effective treatment of AD does not require
complete elimination of S. aureus but is associated with restoration of
the normal level of diversity. It is likely that this increase in bacterial
diversity reestablishes normal immune homeostasis in the skin; specific
members of the skin microbiota have been shown to induce protective
skin-restricted immune responses. Coagulase-negative staphylococci
(CoNS; primarily S. epidermidis and S. hominis) obtained from lesional
and nonlesional skin of patients with AD were functionally screened
and compared to CoNS from healthy controls; AD-lesional CoNS
were much less often able to produce antimicrobial peptides (lantibiotics) directed against S. aureus. To demonstrate that these lantibioticproducing CoNS were biologically relevant, they were incorporated
into a lotion and applied to the arms of patients with AD. Surprisingly,
a single application of the probiotic-laced lotion led to a decrease in
the abundance of S. aureus recovered; no such decrease was observed
when lantibiotic-negative strains were used. The authors of this study
did not specifically comment on the clinical improvement of the AD
lesions. Nevertheless, this is one of a limited number of studies that is
beginning to extend microbiome-related findings into clinical trials.
ASTHMA Asthma is characterized by the clinical triad of airflow
obstruction, bronchial hyperresponsiveness, and inflammation in the
lower respiratory tract. Although the long-standing dogma was that
the lungs are sterile, there is now convincing evidence for a constant
ebb and flow of bacteria within the lower airways. In healthy states, the
mucociliary escalator continually eliminates these bacteria soon after
they land in the airways; in disease states (e.g., cystic fibrosis, chronic
obstructive pulmonary disease), these bacteria establish long-term
colonization of the airways and influence disease pathogenesis. In
asthma specifically, both fecal and airway microbes have been linked
to clinical outcomes.
Early studies of the microbiome’s influence on asthma used
culture-based methods to assess the hypopharyngeal microbiota of
asymptomatic 1-month-old infants. Intriguingly, in one study, early-life
colonization with S. pneumoniae, Haemophilus influenzae, Moraxella
catarrhalis, or a combination of these organisms—but not S. aureus—
was significantly associated with persistent wheeze and asthma at
5 years of age. Eosinophilia and total IgE levels at 4 years of age were
also increased in children who were neonatally colonized with these
organisms. Although this study examined a fairly focused set of bacteria, it laid the experimental groundwork indicating that early-life
microbial exposures influence subsequent development of asthma.
A later longitudinal investigation of the fecal microbiota in a generalpopulation birth cohort of >300 children demonstrated that lower
abundances of the genera Lachnospira, Veillonella, Faecalibacterium,
and Rothia at 3 months of age were associated with an increased risk
for development of asthma. The fact that these bacterial changes were
no longer apparent when the children were 1 year of age is consistent
with the notion that microbial exposures early in life are important to
disease pathogenesis later in life. Transplantation of stool samples from
3-month-old children at risk for asthma into gnotobiotic mice resulted
in significant airway inflammation in a murine model of asthma; preand postnatal exposure of mice to a four-species cocktail (F. prausnitzii,
Veillonella parvula, Rothia mucilaginosa, and Lachnospira multipara)
inhibited airway inflammation, with a marked reduction in neutrophil
numbers in bronchoalveolar lavage fluid. These data suggest that earlylife modulation of the microbiome may be an effective strategy to help
prevent asthma, though the specific logistics (e.g., strains, dose, timing
of exposure, patient selection) remain to be clarified.
Infectious Diseases The increased susceptibility of antibiotictreated mice to infection with a wide range of enteric pathogens was
initially observed in the 1950s and led soon thereafter to the concept
of colonization resistance, which holds that the normal intestinal microbiota plays a critical role in preventing colonization—and therefore
disease production—by invading pathogens. Seminal work in the
1970s demonstrated that this protection is largely reliant on anaerobic
gram-positive organisms, and the subsequent half-century has been
spent trying to identify the specific microbes involved. Although much
of the work relating the microbiota to infection has focused on enteric
pathogens, the intestinal microbiota has also been clearly linked to
bacterial pneumonia in mouse models, and changes in the microbial
composition of the gut have been causally related to changes in the
severity of disease. Although this gut-lung axis clearly exists in animals,
its relevance in humans is still unclear. Several groups are beginning to
study the human lung microbiome in the context of pneumonia and
tuberculosis. Moreover, the relationships between the microbiota and
both systemic infections (e.g., HIV infection, sepsis) and the response
to vaccination are starting to be explored.
ENTERIC INFECTIONS Clostridium difficile infection (CDI) represents
a growing worldwide epidemic and is the leading cause of antibioticassociated diarrhea (Chap. 134). Roughly 15–30% of patients who are
successfully treated for CDI end up with recurrent disease. The strong
association between antibiotic exposure and CDI initially raised the
idea that the microbiota is inextricably linked to acquisition of disease,
presumably because of the loss of colonization resistance. Consistent
with the epidemiologic data, characterization of the fecal microbiota of patients with CDI revealed that it is a markedly less diverse,
dysbiotic community. Fecal microbiota transplantation (FMT)—the
“transplantation” of stool from a healthy individual into patients with
disease—was successfully used in the 1950s to treat four patients with
severe CDI and has recently been demonstrated in numerous studies
to be an effective therapy for recurrent CDI, with clinical cure in
85–90% of patients (as detailed below). Thus, FMT for recurrent CDI
has become the “poster child” for the idea that microbiome-based
The Human Microbiome
3699CHAPTER 471
therapies may transform the management of many diseases previously
considered to be refractory to medical therapy. Although FMT is
agnostic as to the underlying mechanism of protection, work is ongoing to identify specific microbes and host pathways that can protect
against CDI. Studying mice with differential susceptibilities to CDI
due to antibiotic-induced changes in their microbiota, investigators
identified a cocktail of four bacteria (Clostridium scindens, Barnesiella
intestihominis, Pseudoflavonifractor capillosus, and Blautia hansenii)
that conferred protection against CDI in a mouse model. Intriguingly,
treatment of mice with just C. scindens offered significant, though not
complete, protection in a bile acid–dependent manner. Clinical data
from patients who underwent HSCT also associated C. scindens with
protection from CDI, an observation that suggests the possibility of
translating these findings from mice to humans. This study provides
another example of the identification of relevant bacterial factors
through examination of microbial differences in populations that differ
in disease risk.
Microbiome-related changes associated with Vibrio cholerae infection include a striking loss of diversity (largely due to V. cholerae
becoming the dominant member of the microbiota) and an altered
composition that rapidly follows the onset of disease. These changes,
which occur in a reproducible and stereotypical manner, are reversible
with treatment of the disease. This recovery phase involves a microbial succession that is similar to the assembly and maturation of the
microbiota of healthy infants. In addition to V. cholerae, streptococcal
and fusobacterial species bloom during the early phases of diarrhea,
and the relative abundances of Bacteroides, Prevotella, Ruminococcus/
Blautia, and Faecalibacterium species increase during the resolution
phase and mark the return to a healthy adult microbiota. Analysis of
these microbial changes occurring in patients with cholera and in healthy
children led to the selection of 14 bacteria that were transplanted into
gnotobiotic mice, which were then challenged with V. cholerae. Bioinformatic analysis of specific taxa changing during cholera determined
that Ruminococcus obeum restrained V. cholerae growth. Subsequently,
this relationship was experimentally confirmed, and the R. obeum
quorum-sensing molecule AI-2 (autoinducer 2) was found to be responsible for restricting V. cholerae colonization via an unclear mechanism.
These studies highlight the potential for use of microbiome-based
therapies to prevent and/or treat infectious diseases. Moreover, they
suggest that temporal analysis of longitudinal microbiome data may be
an effective strategy for identifying microbes with causal relationships
to disease.
HIV INFECTION The augmentation of HIV pathogenesis by some
viral, bacterial, and parasitic co-infections suggests that a patient’s
underlying microbial environment can influence the severity of HIV
disease. Moreover, it has been hypothesized that the intestinal immune
system plays a significant role in regulating HIV-induced immune
activation; this seems particularly likely since the intestines are an early
site for viral replication and exhibit immune defects before peripheral
CD4+ T-cell counts decrease. Several studies have examined the intestinal microbiotas of HIV-infected individuals. Initial studies performed
in nonhuman primates infected with simian immunodeficiency virus
found no alteration in the bacterial components of the fecal microbiota; however, there were profound changes in the enteric virome.
In contrast, many recent studies exploring this issue in patients have
identified substantial differences in the HIV-associated fecal microbiota that correlate with systemic markers of inflammation. Curiously,
these microbial changes do not necessarily normalize with antiretroviral therapy; this finding suggests that the microbiota may have some
“memory” of the previously high HIV loads and/or that HIV infection
helps reset the “normal” microbiota. This memory-like capacity of the
microbiota has been demonstrated in animal models in the context of
other infections and in response to dieting.
Given that the majority of new HIV transmission events follow heterosexual intercourse, there has been significant interest in examining
the relationship between the vaginal microbiota and HIV acquisition.
A longitudinal study of South African adolescent girls who underwent high-frequency testing for incident HIV infection facilitated
the identification of bacteria that were associated with reduced risk
of HIV acquisition (Lactobacillus species other than L. iners) or with
enhanced risk (Prevotella melaninogenica, Prevotella bivia, Veillonella
montpellierensis, Mycoplasma, and Sneathia sanguinegens). In mice
inoculated intravaginally with Lactobacillus crispatus or P. bivia, the
latter organism induced a greater number of activated CD4+ T cells
in the female genital tract, a result suggesting that the increased risk
of HIV acquisition associated with P. bivia may be secondary to the
increased presence of target cells. In a separate study, the composition
of the vaginal microbiota was shown to modulate the antiviral efficacy
of a tenofovir gel microbicide. Although tenofovir reduced HIV acquisition by 61% in women who had a Lactobacillus-dominant vaginal
microbiota, it reduced HIV acquisition by only 18% in women whose
vaginal microbiota comprised primarily Gardnerella vaginalis and
other anaerobes. This difference in efficacy was due to the ability of
G. vaginalis to metabolize tenofovir faster than the target cells can take
up the drug and convert it into its active form, tenofovir diphosphate.
These findings illustrate how microbial ecology can be an important
consideration in choosing effective treatment regimens.
RESPONSE TO VACCINATION Second only to the provision of clean
water, vaccination has been the most effective public health intervention in the prevention of serious infectious diseases. Its effects are
mediated by antigen-specific antibodies and, in some cases, effector
T-cell responses. Although vaccines are clearly effective on a population scale, the magnitude of the immune response to vaccines can vary
among individuals by tenfold to a hundredfold. Although many factors
(e.g., genetics, maternal antibody levels, prior antigen exposures) can
affect vaccine immunogenicity, the microbiota is now recognized as
another important factor. Analysis of the fecal microbiotas of ~50
Bangladeshi children identified specific taxa that exhibited positive
associations (e.g., Actinomyces, Rothia, and Bifidobacterium species)
and negative associations (e.g., Acinetobacter, Prevotella, and staphylococcal species) with responses to vaccines against polio, tuberculosis (bacille Calmette-Guérin), tetanus, and hepatitis B. A study of
infants from Ghana revealed an inverse relationship between the fecal
abundance of Bacteroidetes and a response to the rotavirus vaccine.
Moreover, the nasal microbiota has been implicated as a factor that
contributes to the IgA response to live, attenuated influenza vaccines.
These correlations based on clinical data have been partially confirmed in animal studies. The best example is the demonstration that
the responses to non-adjuvanted viral subunit vaccines (inactivated
influenza and polio vaccines) are reliant on the microbiota, whereas
the responses to live or adjuvanted vaccines (live attenuated yellow
fever, Tdap/alum, an HIV envelope protein/alum vaccine) are not. A
causal role for the microbiome influencing vaccine-induced immunity
in humans was demonstrated by comparing microneutralization titers
following the inactivated influenza vaccine in individuals treated with
or without antibiotics, although an antibiotic-dependent effect was
only present in subjects who had low levels of preexisting immunity
to influenza. These data suggest that the microbiota may serve as an
adjuvant for certain vaccine types and in naïve populations. Confirmation of these findings in clinical settings may suggest ways to improve
vaccine efficacy in the future.
MECHANISMS OF MICROBIOME-MEDIATED
EFFECTS
As highlighted in the examples above, numerous associations have
been made between the microbiome and various disease states. These
correlations have often been established at broad taxonomic levels,
with little or no insight into causality. Given that most clinical studies
of these relationships have a fairly small sample size (often <100) and
are simultaneously comparing numerous variables (i.e., each of the
bacterial species in the microbiota is effectively a different feature being
compared), many of these studies may not be adequately powered and
therefore may yield false-positive results. Testing of these correlations
in animal models of disease has been critical in demonstrating a causal
relationship between microbes and specific phenotypes. Because
microbiome-wide association studies typically result in a long list of
3700 PART 16 Genes, the Environment, and Disease
bacterial taxa that are correlated with a disease, it has been challenging
to know which organism to test further in mechanistic studies. Moreover, even if a specific bacterial species is identified in these analyses,
there is potentially enough strain-to-strain variation that the “functional” isolate may need to be recovered from the individuals studied;
a publicly available representative of the species may not confer the
same phenotype.
Despite all these difficulties, a handful of specific microbes have
now been linked to disease effects; some examples have been mentioned above. The next layer of challenges relates to identification
of the specific mechanisms that underlie these causal relationships.
Successes along these lines have been more limited, but approaches
are being developed to tackle the issue of defining specific bacterial
factors and metabolites that are responsible for the phenotypic changes.
Complicating factors are that many organisms, particularly those in
the phylum Firmicutes, are not readily genetically tractable and that
many of the phenotypes are not easy to assess with high-throughput
screening.
■ BACTERIAL FACTORS
B. fragilis polysaccharide A (PSA) is perhaps the best-studied
commensal-derived molecule that has been demonstrated to influence
disease outcomes in mouse models. PSA—one of at least eight capsular polysaccharides expressed by B. fragilis—has a unique zwitterionic
structure that incorporates both a positive and a negative charge within
each repeating unit. Studies in which mice have been treated either with
isogenic strains of B. fragilis that differ in PSA expression or with purified
PSA have shown that PSA confers protection—prophylactically and therapeutically—against experimental colitis and MS. PSA is recognized by
Toll-like receptor 2 on antigen-presenting cells, particularly plasmacytoid dendritic cells, and—in the setting of inflammation—induces
interleukin 10 (IL-10)–producing regulatory T cells (Tregs) that help
restrain inflammation.
B. fragilis is also the source of the only other microbiota-based bacterial factor identified thus far: an immunomodulatory glycosphingolipid that affects the numbers of invariant natural killer T (iNKT) cells.
It is not clear whether these glycosphingolipids activate or inhibit iNKT
cells; results have been discordant, probably because different glycosphingolipid species have been tested. Analysis of a specific purified
glycosphingolipid (Bf717) demonstrated that it inhibits endogenous
iNKT cell agonists in vitro and in vivo. Treatment of neonatal mice
with Bf717 leads to a decreased number of colonic iNKT cells in adulthood and to improved outcomes in a model of colitis.
■ BACTERIAL METABOLITES
The use of mass spectrometry to detect and profile tens of thousands
of different metabolites present in different bodily fluids has offered
the promise of deeper insight into microbially mediated processes that
underlie disease susceptibility. However, the fact that the overwhelming majority of these metabolites are not annotated, coupled with the
sheer volume of data generated, has so far limited the general utility
of these untargeted approaches. Instead, interest in more targeted
approaches has increased, with a current emphasis on examining the
role of short-chain fatty acids (SCFAs) and bile acids.
Short-Chain Fatty Acids Several groups have demonstrated
that SCFAs, the intestinal levels of which are largely determined
by bacterial metabolism, are important for the induction of Tregs,
though there is not agreement on which specific SCFA (propionate,
acetate, or butyrate) is most relevant. Wild-type mice colonized with
bacteria known to induce colonic Tregs have elevated cecal levels of
SCFAs. Colonization with any of three Bacteroides species (B. caccae,
B. massiliensis, and B. thetaiotaomicron) increases levels of acetate and
propionate, whereas colonization with Parabacteroides distasonis or
a mix of 17 human-derived Clostridium species elevates levels of all
three SCFAs. In all of these cases, though, the SCFAs inhibit histone
deacetylase, with a consequent increase in Foxp3 expression. Notably,
microbe-induced SCFA production has not been shown to be critical
for Treg induction by any of these organisms. In contrast, there appears
to be no correlation between SCFA levels and Treg numbers in mice
monocolonized with various Treg-inducing bacterial species. Taken
together, these data suggest important heterogeneity in the mechanisms underlying Treg development and do not rule out the possibility
of other, redundant mechanisms for Treg induction. In addition to
effects on Tregs, SCFAs also promote the epithelial barrier, impact
cell proliferation (directionality depends on the specific cell type and
SCFA), regulate host metabolism, and provide an energy source to
colonocytes.
Bile Acids Bile acids are produced in the liver but then are metabolized by intestinal bacteria to form deconjugated and secondary bile
acids. These microbially produced bile acid profiles act through complex signaling pathways to balance the metabolism of lipids and carbohydrates and to affect immune responses. Therefore, bile acids are now
being investigated as microbial metabolites that are critical to maintaining human health. As mentioned above, C. scindens helps protect
mice against CDI through a bile acid–dependent process. Alterations
in bile acid profiles due to underlying microbial dysbiosis have also
been associated with hepatic and colonic inflammation, hepatic cellular carcinoma, colorectal cancer, and impaired gut motility. Almost all
of these relationships have been documented at the level of correlation
and, at best, reflect a partial change in phenotype in the setting of bile
acid sequestrants (e.g., cholestyramine). Work is ongoing to determine
causal relationships between bacterial metabolism of bile acids and
changes in host physiology, though the most definitive evidence is that
microbe-produced bile acid metabolites influence Treg homeostasis.
Other Bacterial Metabolites Although most work has thus far
focused on SCFAs and bile acids, a few notable examples of other
bacterial metabolites have been implicated in maintaining health.
The microbiota metabolizes tryptophan into various products (e.g.,
kynurenine, indole and its derivatives) that influence immune function,
metabolic diseases, and neuronal function, among other things. Taurine enhances NLRP6 inflammasome–induced colonic IL-18 secretion,
while histamine, spermine, and putrescine suppress IL-18 secretion.
Desaminotyrosine produced by Clostridium orbiscindens confers protection from influenza by inducing type I interferon activity. In both
of these cases, the microbiota was initially shown to influence the phenotype, with either untargeted metabolomics or a more targeted screen
indicating a potential role for the indicated metabolites. Given the
thousands of different bacterial metabolites throughout the body, many
more metabolites will undoubtedly be linked to health and disease.
MOVING MICROBIOME SCIENCE FROM
BENCH TO BEDSIDE
The numerous microbiome–disease associations identified thus far
have generated a great deal of hope that understanding the relevant
microbe–host interactions will open the door to unlimited therapeutic
applications. Microbiome-based therapies offer several potential benefits. Patients often view such treatment as more “natural” than conventional drug therapy and are therefore more likely to comply with it.
Biologically, microbiome-based therapies are more likely to address one
of the root causes of disease (microbial dysbiosis) rather than simply
affecting the downstream sequelae. Finally, a given microbiome-based
therapy may serve as a “polypill” that is effective against several different
diseases stemming from similar microbial changes. Despite tremendous
interest in therapeutically exploiting the microbiome, there have thus
far been few clinical successes along these lines.
The most successful therapeutic application of microbiome science
has been the use of FMT, particularly for CDI. As mentioned earlier,
FMT involves “transplanting” stool from a healthy individual to a diseased patient, with the idea that the “healthy” microbiota will correct
whatever derangement may exist in the ill patient and therefore will
alleviate symptoms. Fundamentally, this notion is agnostic as to the
specific microbial dysbiosis and holds that any healthy microbiota will
be curative. The idea of FMT dates back to at least the fourth century,
when traditional Chinese doctors used a “yellow soup” (fresh human
fecal suspension) to successfully treat food poisoning and severe
The Human Microbiome
3701CHAPTER 471
diarrhea. The continued use of FMT through the centuries for the
treatment of diarrheal illnesses in both humans and animals, along
with the growing appreciation in recent years of the importance of the
microbiota, laid the groundwork for using FMT to treat CDI. Since the
first major prospective trial assessing FMT for recurrent CDI in 2013,
most of the numerous studies of FMT for CDI have demonstrated
remarkable efficacy, with an average clinical cure rate of ~85%. The
donor stool can be fresh or frozen (use of the latter allows biobanking
of samples from a limited number of prescreened donors) and can be
administered via nasogastric tube, nasoduodenal tube, colonoscopy,
enema, or oral capsules; the cure rate is slightly higher with lower-gastrointestinal administration than with upper-gastrointestinal treatment.
The optimal screening, preparation, and concentration of infused
donor stool have not yet been determined, and there have been cases
of antimicrobial-resistant pathogens transmitted by FMT that have led
to mortality. The most common adverse effects of FMT include altered
gastrointestinal motility (with constipation or diarrhea), abdominal
cramps, and bloating, all of which are generally transient and resolve
within 48 h. Although controlled studies of the use of FMT in immunosuppressed patients do not yet exist, meta-analyses of case reports
and case series have found no serious FMT-related adverse events in
>300 immunocompromised patients.
The successful use and the favorable short-term safety profile of
FMT for CDI have led to its expanded application for other indications.
At the end of 2020, >350 trials (listed at ClinicalTrials.gov) were investigating the efficacy of FMT for a range of indications, including CDI,
IBD (ulcerative colitis and Crohn’s disease), obesity, eradication of
multidrug-resistant organisms, anxiety and depression, cirrhosis, and
type 2 diabetes. The few published studies regarding indications other
than CDI have generally included small sample sizes and have offered
mixed results. In contrast to the successes in CDI, the results have been
more varied for patients with IBD, which is perhaps the second-beststudied indication. It is not clear whether these discrepancies are due
to heterogeneity in recipients (e.g., in terms of underlying disease
mechanisms or endogenous microbiotas), the donor material, and/
or the logistical details of FMT administration (e.g., route, frequency,
dose). However, these results demonstrate that—under the right
circumstances—modulation of the microbiota can be an effective
therapy for IBD.
Although FMT offers an important proof of concept that
microbiome-based therapies can be effective, treatment is difficult
to standardize across large populations because of variability among
stool donors and among the endogenous microbiotas of recipients.
In addition, FMT is fraught with safety concerns, and its mechanisms
of action are unclear. FMT likely represents the first generation of
microbiome-based therapies; subsequent generations will include the
use of more refined bacterial cocktails, single strains of bacteria, or
bacterial products and/or metabolites as the therapeutic intervention.
The field of probiotics has a complicated history: many different strains
have been tested against a multitude of diseases. Several meta-analyses
have combined results across bacterial strains and/or disease indications and have generally concluded that the data are not yet convincing
enough to support the use of the tested regimens. It should be noted
that the tested organisms have been chosen mainly on the basis of
their presumed safety profile rather than in light of a plausible biologic
link to disease. The hope is that more focused, mechanistic microbiome studies will identify specific commensal organisms—and their
underlying mechanisms of action—that are involved in disease pathogenesis and that will serve as the basis for the next wave of rationally
chosen probiotics, a few of which are currently in clinical trials. The
main hurdle in this endeavor has been identifying specific microbes
that are causally related to protection from disease.
PERSPECTIVE
The medical view of microbes has changed radically, moving from the
early-twentieth-century notion that we are engaged in a constant struggle with microbes—an “us-versus-them” mentality that focused on the
necessity of eradicating bacteria—to the more recent understanding
that we live in a carefully negotiated state of détente with our commensal organisms. Instead of holding a simple view of microbes as enemies
to be eliminated with antibiotics, scientists are increasingly recognizing
the critical role these organisms play in maintaining human health;
loss of these host–microbe interactions in the increasingly sterile
environment typical of Western civilization may have predisposed to
the increased incidence of autoimmune and inflammatory diseases.
The field of microbiome research has made great strides over the past
decade in cataloguing the normal microbiota and is now on the cusp of
being able to identify clinically actionable microbe–host relationships.
The recent explosion of “–omics” technologies (e.g., metagenomics,
metatranscriptomics, metabolomics) has enabled the generation of
vast amounts of data, but it is not yet clear how best to integrate data
sets in order to gain useful insights into host–microbe relationships.
The use of FMT has demonstrated that modulation of an individual’s
microbiota can effectively treat certain diseases; however, models
with which to predict specifically how a microbiota will change after
modulation—and what potentially untoward effects these changes
might have—are still lacking. Implicit in this limitation is our ignorance about what microbial configuration is optimal and how a given
microbiota should be rationally altered to obtain an ideal outcome.
Despite initial hyperbolic hype and a few false starts, microbiome
research now stands at the forefront of an ability to treat the fundamental basis of many diseases. As the field continues to mature, it will need
to move beyond correlations and address causation. The identification
of causal microbes and their mechanisms of action will create a “microbial toolbox” from which relevant bioactive strains can be chosen on
a per-patient basis to correct specific underlying microbial dysbioses.
In the near future, our knowledge base regarding the microbiome and
its relationship to health and disease will be robust enough that this
information can be applied in making important treatment decisions.
■ FURTHER READING
Goodrich JK et al: Conducting a microbiome study. Cell 158:250,
2014.
Human Microbiome Project Consortium: Structure, function and
diversity of the healthy human microbiome. Nature 486:207, 2012.
Lynch SV, Pederson O: The human intestinal microbiome in health
and disease. N Engl J Med 375:2369, 2016.
Rajilic’-Stojanovic’ M, deVos WM: The first 1000 cultured species
of the human gastrointestinal microbiota. FEMS Microbiol Rev
38:996, 2014.
Schmidt TSB et al: The human gut microbiome: From association to
modulation. Cell 172:1198, 2018.
Stefan KL et al: Commensal microbiota modulation of natural resistance to virus infection. Cell 183:1, 2020.
This page intentionally left blank
Global Medicine PART 17
Global Issues in Medicine
Joseph J. Rhatigan, Paul Farmer
472
WHY GLOBAL HEALTH?
Global health has emerged as an important field within medicine.
Some scholars have defined global health as the field of study and
practice concerned with improving the health of all people and
achieving health equity worldwide, with an emphasis on addressing
transnational problems. No single review can do much more than
identify the leading problems in applying evidence-based medicine in
settings of great poverty or across national boundaries. However, this is
a moment of opportunity: only relatively recently have persistent calls
for global health equity been matched by an unprecedented investment in addressing the health problems of poor people worldwide.
To ensure that this opportunity is not wasted, we must strengthen
health systems and improve health care delivery to address the true
burden and distribution of disease. This chapter introduces the major
international bodies that address health problems; identifies the more
significant barriers to improving the health of people who to date have
not, by and large, had access to modern medicine; and summarizes
population-based data on the most common health problems faced
by people living in poverty. Examining specific problems—notably
HIV/AIDS (Chap. 202) but also tuberculosis (Chap. 178), malaria
(Chap. 224), Ebola (Chap. 210), and key “noncommunicable”
chronic diseases (NCDs)—helps sharpen the discussion of barriers
to prevention, diagnosis, and care as well as the means of overcoming
them. This chapter closes by discussing global health equity, drawing
on notions of social justice that once were central to international
public health but had fallen out of favor during the last decades of the
twentieth century.
A BRIEF HISTORY OF GLOBAL
HEALTH INSTITUTIONS
Concern about illness across national boundaries dates back many
centuries, predating the Black Plague and other pandemics. One of
the first organizations founded explicitly to tackle cross-border health
issues was the Pan American Sanitary Bureau, which was formed
in 1902 by 11 countries in the Americas. The primary goal of what
later became the Pan American Health Organization was the control
of infectious diseases across the Americas. Of special concern was
yellow fever, which had been running a deadly course through much
of South and Central America and halted the construction of the Panama Canal. In 1948, the United Nations formed the first truly global
health institution: the World Health Organization (WHO). In 1958,
under the aegis of the WHO and in line with a long-standing focus
on communicable diseases that cross borders, leaders in global health
initiated the effort that led to what some see as the greatest success in
international health: the eradication of smallpox. Naysayers were surprised when the smallpox eradication campaign, which engaged public
health officials throughout the world, proved successful in 1979 despite
Cold War tensions.
At the International Conference on Primary Health Care in Alma-Ata
(in what is now Kazakhstan) in 1978, public health officials from
around the world agreed on a commitment to “Health for All by the
Year 2000,” a goal to be achieved by providing universal access to
primary health care worldwide. Critics argued that the attainment of
this goal by the proposed date was impossible. In the ensuing years,
a strategy for the provision of selective primary health care emerged.
This strategy included four inexpensive interventions collectively
known as GOBI: growth monitoring, oral rehydration, breast-feeding,
and immunizations for diphtheria, whooping cough, tetanus, polio,
tuberculosis, and measles. GOBI later was expanded to GOBI-FFF,
which also included female education, food, and family planning. Some
public health figures saw GOBI-FFF as an interim strategy to achieve
“health for all,” but others criticized it as a retreat from the bolder commitments of Alma-Ata.
The influence of the WHO waned during the 1980s. In the early
1990s, many observers argued that, with its vastly superior financial
resources and its close—if unequal—relationships with the governments of poor countries, the World Bank had eclipsed the WHO as the
most important multilateral institution working in the area of health.
One of the stated goals of the World Bank was to help poor countries
identify “cost-effective” interventions worthy of public funding and
international support. At the same time, international financial institutions encouraged many of those nations to reduce public expenditures
in health and education in order to stimulate economic growth as part
of (later discredited) policies imposing restrictions as a condition for
access to credit and assistance through the World Bank, the International Monetary Fund, and regional development banks. There was a
resurgence of many diseases—including malaria, trypanosomiasis, and
schistosomiasis—in Africa. Tuberculosis, an eminently curable disease,
remained the world’s leading infectious killer of adults. Half a million
women per year died in childbirth during the last decade of the twentieth century, and few of the world’s largest philanthropic or funding
institutions focused on global health equity.
HIV/AIDS, first described in the medical literature in 1981, precipitated a change. In the United States, the advent of this newly described
infectious killer marked the culmination of a series of events that
dashed previous hopes of “closing the book” on infectious diseases. In
Africa, which would emerge as the global epicenter of the pandemic,
HIV disease strained tuberculosis control programs, and malaria
continued to claim as many lives as ever: at the dawn of the twentyfirst century, these three diseases alone killed nearly 6 million people
each year. New research, new policies, and new funding mechanisms
were called for. The past two decades have seen the rise of important
multilateral global health financing institutions such as the Global
Fund to Fight AIDS, Tuberculosis, and Malaria; bilateral efforts such
as the U.S. President’s Emergency Plan for AIDS Relief (PEPFAR); and
private philanthropic organizations such as the Bill & Melinda Gates
Foundation. With its 193 member states and 150 country offices, the
WHO remains important in matters relating to the cross-border spread
of infectious diseases and other health threats. In the aftermath of the
epidemic of severe acute respiratory syndrome in 2003, the WHO’s
International Health Regulations—which provide a legal foundation
for that organization’s direct investigation into a wide range of global
health problems, including pandemic influenza, in any member state—
were strengthened and brought into force in May 2007.
Even as attention to and resources for health problems in poor
countries grow, the lack of coherence in and among global health
institutions may undermine efforts to forge a more comprehensive
and effective response. The WHO remains underfunded despite the
ever-growing need to engage a wider and more complex range of health
issues, such as the Ebola outbreak of 2013–2016 in West Africa. This
may be what some have called “the golden age of global health,” but
leaders of major global health organizations must work together to
design an effective architecture that will make the most of opportunities to link new resources for and commitments to global health equity
with the emerging understanding of disease burden and the unmet
need to create robust and resilient national health systems. To this end,
new and old players in global health must invest heavily in discovery
(relevant basic science), development of new tools (preventive, diagnostic, and therapeutic), and modes of delivery that will ensure the equitable provision of health products and services to all who need them.
The adoption of the Sustainable Development Goals (SDGs) in 2015
by the United Nations serves as an example of effective cooperation.
The SDGs articulate 17 overarching goals across several domains to
3704 PART 17 Global Medicine
be achieved by 2030. Goal 3 specifically relates to global health and
contains 13 distinct targets to be met, including reducing maternal
and child mortality; ending the epidemics of HIV, tuberculosis, and
malaria; and reducing the burden of NCDs.
Included in the SDGs is a commitment to achieve universal health
coverage (UHC), providing universal access to high-quality essential
health services at an affordable cost worldwide. Championed by the
WHO, the World Bank, and many civil society organizations, Goal
3 will measure coverage of 16 essential health services and assess the
financial burden of health spending by households in every country.
THE ECONOMICS OF GLOBAL HEALTH
Political and economic concerns have often guided global health interventions. As mentioned, early efforts to control yellow fever were tied
to the completion of the Panama Canal. However, the precise nature
of the link between economics and health remains a matter for debate.
Some economists and demographers argue that improving the health
status of populations must begin with economic development; others
maintain that addressing ill health is the starting point for development
in poor countries. In either case, there is increasing consensus that
investments in health care delivery and the control of communicable
diseases lead to increased productivity. The question is where to find
the necessary resources to start the predicted “virtuous cycle.”
During the past two decades, spending on health in poor countries
has increased dramatically. According to a study from the Institute for
Health Metrics and Evaluation (IHME) at the University of Washington,
total development assistance for health worldwide grew to $38.9 billion
in 2018—up from $5.6 billion in 1990, but down 3.3% from 2017. In
2018, the leading contributors included the United States, the United
Kingdom, and other private philanthropy, with the largest channels of
funding being nongovernmental organizations (NGOs) and U.S. bilateral aid agencies. It appears that the growth of development assistance
for health has plateaued: from 2013 to 2018, its annualized growth
rate was –0.3%, and it is unclear whether its growth will resume in the
future.
MORTALITY AND THE GLOBAL BURDEN OF
DISEASE
Refining metrics is an important task for global health: only relatively
recently have there been solid assessments of the global burden of
disease. The first study to look seriously at this issue, conducted in
1990, laid the foundation for the first report on Disease Control
Priorities in Developing Countries and for the World Bank’s 1993 World
Development Report Investing in Health. Those efforts represented
a major advance in the understanding of health status in developing
countries. Investing in Health has been especially influential: it familiarized a broad audience with cost-effectiveness analysis for specific
health interventions and with the notion of disability-adjusted life
years (DALYs). The DALY, which has become a standard measure of
the impact of a specific health condition on a population, combines
absolute years of life lost and years lost due to disability for incident
cases of a condition. (See Fig. 472-1 and Table 472-1 for an analysis of
the global disease burden by DALYs.)
In 2012, the IHME and partner institutions began publishing results
from the Global Burden of Diseases, Injuries, and Risk Factors Study
2010 (GBD 2010). GBD 2010 is the most comprehensive effort to date
to produce longitudinal, globally ambitious, and comparable estimates
of the burden of diseases, injuries, and risk factors. This report reflects
the expansion of the available data on health in the poorest countries
and of the capacity to quantify the impact of specific conditions on
a population. It measures current levels and recent trends for major
diseases, injuries, and risk factors worldwide. The GBD 2010 team
revised and improved the health-state severity weight system, collated
published data, and used household surveys to enhance the breadth
and accuracy of disease burden data. Updated reports were released in
2013, 2015, and 2017. As analytic methods and data quality improve,
important trends can be identified in a comparison of global disease
burden estimates from 1990 to 2017.
■ GLOBAL MORTALITY
Of the 55.9 million deaths worldwide in 2017, 19% (10.4 million) were
due to communicable diseases, maternal and neonatal conditions, and
nutritional deficiencies—a marked decrease compared with figures for
1990, when these conditions accounted for 32% of global mortality.
Among the fraction of all deaths related to communicable diseases,
maternal and neonatal conditions, and nutritional deficiencies, 75%
occurred in sub-Saharan Africa and southern Asia. While the proportion of deaths due to these conditions has decreased significantly
in the past decade, there has been a dramatic rise in the number of
deaths from NCDs, which increased between 2007 and 2017 by 23%.
The leading cause of death worldwide in 2017 was ischemic heart
disease, accounting for 8.9 million deaths (16% of total deaths). In
high-income countries, ischemic heart disease accounted for 17% of
total deaths, and in low- and middle-income countries, it accounted
for 7% and 17%, respectively. It is noteworthy that ischemic heart disease was responsible for just 5% of total deaths in sub-Saharan Africa
(Table 472-2). In second place—causing 11% of global mortality—was
stroke, which accounted for 8% of deaths in high-income countries,
6% and 12% in low- and middle-income countries, respectively, and
4% in sub-Saharan Africa. Although chronic obstructive pulmonary
disease (COPD) was the third leading cause of death globally and was
the fifth leading cause in high-income countries (accounting for 5% of
all deaths), this condition did not figure among the top 15 causes in
sub-Saharan Africa. Among the 10 leading causes of death in sub-Saharan
Africa, six were infectious diseases, with HIV/AIDS, lower respiratory
infections, diarrheal diseases, and malaria ranking as dominant contributors to disease burden. In high-income countries, however, only
one infectious disease—lower respiratory infection—ranked among
the top 10 causes of death.
The GBD 2017 found that the worldwide mortality figure among
children under 5 years of age dropped from 16.4 million in 1970 to
11.8 million in 1990 and to 5.4 million in 2017—a decrease that far
surpassed predictions. Of childhood deaths in 2017, 2.4 million (44%)
occurred in the neonatal period. Just under one-third of deaths among
children under 5 years old occurred in southern Asia and slightly
more than one-half in sub-Saharan Africa, but only ~1% occurred in
high-income countries.
The global burden of death due to HIV/AIDS and malaria was on an
upward slope until 2004, but significant progress has been made since
then. Global deaths from AIDS fell from 2.0 million in 2006 to 955,000
in 2017, while malaria deaths dropped from 1.2 million to 620,000 over
the same period. Despite these improvements, malaria and HIV/AIDS
continue to be major burdens in particular regions, with global implications. Although it has only a minor impact on mortality outside sub
-Saharan Africa and Southeast Asia, malaria is the fifth leading cause of
death of children under 5 years of age worldwide. HIV infection ranked
28th in global DALYs in 1990 but was the 11th leading cause of disease
burden in 2017, with sub-Saharan Africa bearing the vast majority of
this burden (Fig. 472-1).
The world’s population is living longer: global life expectancy has
increased significantly over the past 50 years from 58.8 years in 1970 to
73 years in 2017. This demographic change, accompanied by the fact
that the prevalence of NCDs increases with age, is dramatically shifting
the burden of disease toward NCDs, which have surpassed communicable, maternal, nutritional, and neonatal causes. By 2017, 73% of total
deaths at all ages and 62% of all DALYs were due to NCDs. Increasingly, the global burden of disease comprises conditions and injuries
that cause disability rather than death.
Worldwide, although both life expectancy and years of life lived
in good health have risen, years of life lived with disability have also
increased. Globally, the total burden of disability increased by 50%
between 1950 and 2017. Despite the higher prevalence of diseases
common in older populations (e.g., dementia and musculoskeletal
disease) in developed and high-income countries, best estimates from
2017 reveal that disability resulting from cardiovascular diseases,
chronic respiratory diseases, and the long-term impact of communicable diseases was greater in low- and middle-income countries. In
3705 Global Issues in Medicine CHAPTER 472
most developing countries, people lived shorter lives and experienced
disability and poor health for a greater proportion of their lives. Indeed,
47% of the global burden of disease occurred in southern Asia and
sub-Saharan Africa, which together account for only 38% of the world’s
population.
■ HEALTH AND WEALTH
Clear disparities in burden of disease (both communicable and noncommunicable) across country income levels are strong indicators
that poverty and health are inherently linked. Numerous studies have
documented the link between poverty and health within nations as
well as across them. Poverty remains one of the most important root
causes of poor health worldwide, and the global burden of poverty
continues to be high. Among the 7.3 billion people alive in 2015, 10%
(736 million) lived on less than $1.90 (in 2011 U.S. dollars) per day—a
standard measurement of extreme poverty—and 25% lived on less than
$3.20 per day. Just under one-fifth of all children under the age of 14
worldwide lived in extreme poverty in 2015. The extreme poverty rate
has declined steadily since 1990, and by 2015, there were more than
one billion fewer people living in poverty despite growth in the global
population of more than two billion during that time period. However, improvement has been uneven across the globe. In sub-Saharan
Africa, people living in extreme poverty increased from 278 million to
413 million during the same period.
■ RISK FACTORS FOR DISEASE BURDEN
The GBD study found that the three leading risk factors for global disease burden in 2017 were (in order of frequency) high systolic blood
pressure, smoking, and high fasting plasma glucose—a substantial
change from 1990, when child wasting was ranked first. Although
ranking ninth in 2017, child wasting remains the third leading risk
factor for death worldwide among children under 5 years of age. In
an era that has seen obesity become a major health concern in many
developed countries—and the fourth leading risk factor worldwide—
the persistence of undernutrition is cause for consternation. Child
wasting is still a dominant risk factor for disease burden in sub-Saharan
Africa. In its rural reaches, no health care initiative, however generously funded, will be effective or comprehensive without addressing
undernutrition.
In an analysis that examined how specific diseases and injuries are
affected by environmental risk, the WHO estimated that 24% of all
deaths and 28% of deaths among children <5 years of age in 2016 were
due to modifiable environmental factors: some 1.6 million children die
every year from causes related to unhealthy environments, including
the nearly 300,000 deaths stemming from a lack of access to clean
water and sanitation. Many of these modifiable factors lead to child
and adult deaths from infectious pathologies; others lead to deaths
from malignancies. Etiology and nosology are increasingly difficult to
parse with regard to environmental harm. Risk factors such as indoor
1 Neonatal disorders
1990 Rank 2017 Rank
Global
Both sexes, all ages, DALYs
2 Lower respiratory infect
3 Diarrheal diseases
4 Ischemic heart disease
5 Stroke
6 Congenital defects
7 Road injuries
8 Tuberculosis
9 COPD
10 Measles
11 Malaria
12 Low back pain
13 Protein-energy malnutrition
14 Drowning
15 Self-harm
16 Meningitis
17 Headache disorders
18 Dietary iron deficiency
19 Diabetes
20 Cirrhosis
21 Depressive disorders
23 Falls
24 Lung cancer
28 HIV/AIDS
1 Neonatal disorders
Communicable, maternal, neonatal,
and nutritional diseases
Non-communicable diseases Injuries
2 Ischemic heart disease
3 Stroke
4 Lower respiratory infect
5
472
WHY GLOBAL HEALTH?
Global health has emerged as an important field within medicine.
Some scholars have defined global health as the field of study and
practice concerned with improving the health of all people and
achieving health equity worldwide, with an emphasis on addressing
transnational problems. No single review can do much more than
identify the leading problems in applying evidence-based medicine in
settings of great poverty or across national boundaries. However, this is
a moment of opportunity: only relatively recently have persistent calls
for global health equity been matched by an unprecedented investment in addressing the health problems of poor people worldwide.
To ensure that this opportunity is not wasted, we must strengthen
health systems and improve health care delivery to address the true
burden and distribution of disease. This chapter introduces the major
international bodies that address health problems; identifies the more
significant barriers to improving the health of people who to date have
not, by and large, had access to modern medicine; and summarizes
population-based data on the most common health problems faced
by people living in poverty. Examining specific problems—notably
HIV/AIDS (Chap. 202) but also tuberculosis (Chap. 178), malaria
(Chap. 224), Ebola (Chap. 210), and key “noncommunicable”
chronic diseases (NCDs)—helps sharpen the discussion of barriers
to prevention, diagnosis, and care as well as the means of overcoming
them. This chapter closes by discussing global health equity, drawing
on notions of social justice that once were central to international
public health but had fallen out of favor during the last decades of the
twentieth century.
A BRIEF HISTORY OF GLOBAL
HEALTH INSTITUTIONS
Concern about illness across national boundaries dates back many
centuries, predating the Black Plague and other pandemics. One of
the first organizations founded explicitly to tackle cross-border health
issues was the Pan American Sanitary Bureau, which was formed
in 1902 by 11 countries in the Americas. The primary goal of what
later became the Pan American Health Organization was the control
of infectious diseases across the Americas. Of special concern was
yellow fever, which had been running a deadly course through much
of South and Central America and halted the construction of the Panama Canal. In 1948, the United Nations formed the first truly global
health institution: the World Health Organization (WHO). In 1958,
under the aegis of the WHO and in line with a long-standing focus
on communicable diseases that cross borders, leaders in global health
initiated the effort that led to what some see as the greatest success in
international health: the eradication of smallpox. Naysayers were surprised when the smallpox eradication campaign, which engaged public
health officials throughout the world, proved successful in 1979 despite
Cold War tensions.
At the International Conference on Primary Health Care in Alma-Ata
(in what is now Kazakhstan) in 1978, public health officials from
around the world agreed on a commitment to “Health for All by the
Year 2000,” a goal to be achieved by providing universal access to
primary health care worldwide. Critics argued that the attainment of
this goal by the proposed date was impossible. In the ensuing years,
a strategy for the provision of selective primary health care emerged.
This strategy included four inexpensive interventions collectively
known as GOBI: growth monitoring, oral rehydration, breast-feeding,
and immunizations for diphtheria, whooping cough, tetanus, polio,
tuberculosis, and measles. GOBI later was expanded to GOBI-FFF,
which also included female education, food, and family planning. Some
public health figures saw GOBI-FFF as an interim strategy to achieve
“health for all,” but others criticized it as a retreat from the bolder commitments of Alma-Ata.
The influence of the WHO waned during the 1980s. In the early
1990s, many observers argued that, with its vastly superior financial
resources and its close—if unequal—relationships with the governments of poor countries, the World Bank had eclipsed the WHO as the
most important multilateral institution working in the area of health.
One of the stated goals of the World Bank was to help poor countries
identify “cost-effective” interventions worthy of public funding and
international support. At the same time, international financial institutions encouraged many of those nations to reduce public expenditures
in health and education in order to stimulate economic growth as part
of (later discredited) policies imposing restrictions as a condition for
access to credit and assistance through the World Bank, the International Monetary Fund, and regional development banks. There was a
resurgence of many diseases—including malaria, trypanosomiasis, and
schistosomiasis—in Africa. Tuberculosis, an eminently curable disease,
remained the world’s leading infectious killer of adults. Half a million
women per year died in childbirth during the last decade of the twentieth century, and few of the world’s largest philanthropic or funding
institutions focused on global health equity.
HIV/AIDS, first described in the medical literature in 1981, precipitated a change. In the United States, the advent of this newly described
infectious killer marked the culmination of a series of events that
dashed previous hopes of “closing the book” on infectious diseases. In
Africa, which would emerge as the global epicenter of the pandemic,
HIV disease strained tuberculosis control programs, and malaria
continued to claim as many lives as ever: at the dawn of the twentyfirst century, these three diseases alone killed nearly 6 million people
each year. New research, new policies, and new funding mechanisms
were called for. The past two decades have seen the rise of important
multilateral global health financing institutions such as the Global
Fund to Fight AIDS, Tuberculosis, and Malaria; bilateral efforts such
as the U.S. President’s Emergency Plan for AIDS Relief (PEPFAR); and
private philanthropic organizations such as the Bill & Melinda Gates
Foundation. With its 193 member states and 150 country offices, the
WHO remains important in matters relating to the cross-border spread
of infectious diseases and other health threats. In the aftermath of the
epidemic of severe acute respiratory syndrome in 2003, the WHO’s
International Health Regulations—which provide a legal foundation
for that organization’s direct investigation into a wide range of global
health problems, including pandemic influenza, in any member state—
were strengthened and brought into force in May 2007.
Even as attention to and resources for health problems in poor
countries grow, the lack of coherence in and among global health
institutions may undermine efforts to forge a more comprehensive
and effective response. The WHO remains underfunded despite the
ever-growing need to engage a wider and more complex range of health
issues, such as the Ebola outbreak of 2013–2016 in West Africa. This
may be what some have called “the golden age of global health,” but
leaders of major global health organizations must work together to
design an effective architecture that will make the most of opportunities to link new resources for and commitments to global health equity
with the emerging understanding of disease burden and the unmet
need to create robust and resilient national health systems. To this end,
new and old players in global health must invest heavily in discovery
(relevant basic science), development of new tools (preventive, diagnostic, and therapeutic), and modes of delivery that will ensure the equitable provision of health products and services to all who need them.
The adoption of the Sustainable Development Goals (SDGs) in 2015
by the United Nations serves as an example of effective cooperation.
The SDGs articulate 17 overarching goals across several domains to
3704 PART 17 Global Medicine
be achieved by 2030. Goal 3 specifically relates to global health and
contains 13 distinct targets to be met, including reducing maternal
and child mortality; ending the epidemics of HIV, tuberculosis, and
malaria; and reducing the burden of NCDs.
Included in the SDGs is a commitment to achieve universal health
coverage (UHC), providing universal access to high-quality essential
health services at an affordable cost worldwide. Championed by the
WHO, the World Bank, and many civil society organizations, Goal
3 will measure coverage of 16 essential health services and assess the
financial burden of health spending by households in every country.
THE ECONOMICS OF GLOBAL HEALTH
Political and economic concerns have often guided global health interventions. As mentioned, early efforts to control yellow fever were tied
to the completion of the Panama Canal. However, the precise nature
of the link between economics and health remains a matter for debate.
Some economists and demographers argue that improving the health
status of populations must begin with economic development; others
maintain that addressing ill health is the starting point for development
in poor countries. In either case, there is increasing consensus that
investments in health care delivery and the control of communicable
diseases lead to increased productivity. The question is where to find
the necessary resources to start the predicted “virtuous cycle.”
During the past two decades, spending on health in poor countries
has increased dramatically. According to a study from the Institute for
Health Metrics and Evaluation (IHME) at the University of Washington,
total development assistance for health worldwide grew to $38.9 billion
in 2018—up from $5.6 billion in 1990, but down 3.3% from 2017. In
2018, the leading contributors included the United States, the United
Kingdom, and other private philanthropy, with the largest channels of
funding being nongovernmental organizations (NGOs) and U.S. bilateral aid agencies. It appears that the growth of development assistance
for health has plateaued: from 2013 to 2018, its annualized growth
rate was –0.3%, and it is unclear whether its growth will resume in the
future.
MORTALITY AND THE GLOBAL BURDEN OF
DISEASE
Refining metrics is an important task for global health: only relatively
recently have there been solid assessments of the global burden of
disease. The first study to look seriously at this issue, conducted in
1990, laid the foundation for the first report on Disease Control
Priorities in Developing Countries and for the World Bank’s 1993 World
Development Report Investing in Health. Those efforts represented
a major advance in the understanding of health status in developing
countries. Investing in Health has been especially influential: it familiarized a broad audience with cost-effectiveness analysis for specific
health interventions and with the notion of disability-adjusted life
years (DALYs). The DALY, which has become a standard measure of
the impact of a specific health condition on a population, combines
absolute years of life lost and years lost due to disability for incident
cases of a condition. (See Fig. 472-1 and Table 472-1 for an analysis of
the global disease burden by DALYs.)
In 2012, the IHME and partner institutions began publishing results
from the Global Burden of Diseases, Injuries, and Risk Factors Study
2010 (GBD 2010). GBD 2010 is the most comprehensive effort to date
to produce longitudinal, globally ambitious, and comparable estimates
of the burden of diseases, injuries, and risk factors. This report reflects
the expansion of the available data on health in the poorest countries
and of the capacity to quantify the impact of specific conditions on
a population. It measures current levels and recent trends for major
diseases, injuries, and risk factors worldwide. The GBD 2010 team
revised and improved the health-state severity weight system, collated
published data, and used household surveys to enhance the breadth
and accuracy of disease burden data. Updated reports were released in
2013, 2015, and 2017. As analytic methods and data quality improve,
important trends can be identified in a comparison of global disease
burden estimates from 1990 to 2017.
■ GLOBAL MORTALITY
Of the 55.9 million deaths worldwide in 2017, 19% (10.4 million) were
due to communicable diseases, maternal and neonatal conditions, and
nutritional deficiencies—a marked decrease compared with figures for
1990, when these conditions accounted for 32% of global mortality.
Among the fraction of all deaths related to communicable diseases,
maternal and neonatal conditions, and nutritional deficiencies, 75%
occurred in sub-Saharan Africa and southern Asia. While the proportion of deaths due to these conditions has decreased significantly
in the past decade, there has been a dramatic rise in the number of
deaths from NCDs, which increased between 2007 and 2017 by 23%.
The leading cause of death worldwide in 2017 was ischemic heart
disease, accounting for 8.9 million deaths (16% of total deaths). In
high-income countries, ischemic heart disease accounted for 17% of
total deaths, and in low- and middle-income countries, it accounted
for 7% and 17%, respectively. It is noteworthy that ischemic heart disease was responsible for just 5% of total deaths in sub-Saharan Africa
(Table 472-2). In second place—causing 11% of global mortality—was
stroke, which accounted for 8% of deaths in high-income countries,
6% and 12% in low- and middle-income countries, respectively, and
4% in sub-Saharan Africa. Although chronic obstructive pulmonary
disease (COPD) was the third leading cause of death globally and was
the fifth leading cause in high-income countries (accounting for 5% of
all deaths), this condition did not figure among the top 15 causes in
sub-Saharan Africa. Among the 10 leading causes of death in sub-Saharan
Africa, six were infectious diseases, with HIV/AIDS, lower respiratory
infections, diarrheal diseases, and malaria ranking as dominant contributors to disease burden. In high-income countries, however, only
one infectious disease—lower respiratory infection—ranked among
the top 10 causes of death.
The GBD 2017 found that the worldwide mortality figure among
children under 5 years of age dropped from 16.4 million in 1970 to
11.8 million in 1990 and to 5.4 million in 2017—a decrease that far
surpassed predictions. Of childhood deaths in 2017, 2.4 million (44%)
occurred in the neonatal period. Just under one-third of deaths among
children under 5 years old occurred in southern Asia and slightly
more than one-half in sub-Saharan Africa, but only ~1% occurred in
high-income countries.
The global burden of death due to HIV/AIDS and malaria was on an
upward slope until 2004, but significant progress has been made since
then. Global deaths from AIDS fell from 2.0 million in 2006 to 955,000
in 2017, while malaria deaths dropped from 1.2 million to 620,000 over
the same period. Despite these improvements, malaria and HIV/AIDS
continue to be major burdens in particular regions, with global implications. Although it has only a minor impact on mortality outside sub
-Saharan Africa and Southeast Asia, malaria is the fifth leading cause of
death of children under 5 years of age worldwide. HIV infection ranked
28th in global DALYs in 1990 but was the 11th leading cause of disease
burden in 2017, with sub-Saharan Africa bearing the vast majority of
this burden (Fig. 472-1).
The world’s population is living longer: global life expectancy has
increased significantly over the past 50 years from 58.8 years in 1970 to
73 years in 2017. This demographic change, accompanied by the fact
that the prevalence of NCDs increases with age, is dramatically shifting
the burden of disease toward NCDs, which have surpassed communicable, maternal, nutritional, and neonatal causes. By 2017, 73% of total
deaths at all ages and 62% of all DALYs were due to NCDs. Increasingly, the global burden of disease comprises conditions and injuries
that cause disability rather than death.
Worldwide, although both life expectancy and years of life lived
in good health have risen, years of life lived with disability have also
increased. Globally, the total burden of disability increased by 50%
between 1950 and 2017. Despite the higher prevalence of diseases
common in older populations (e.g., dementia and musculoskeletal
disease) in developed and high-income countries, best estimates from
2017 reveal that disability resulting from cardiovascular diseases,
chronic respiratory diseases, and the long-term impact of communicable diseases was greater in low- and middle-income countries. In
3705 Global Issues in Medicine CHAPTER 472
most developing countries, people lived shorter lives and experienced
disability and poor health for a greater proportion of their lives. Indeed,
47% of the global burden of disease occurred in southern Asia and
sub-Saharan Africa, which together account for only 38% of the world’s
population.
■ HEALTH AND WEALTH
Clear disparities in burden of disease (both communicable and noncommunicable) across country income levels are strong indicators
that poverty and health are inherently linked. Numerous studies have
documented the link between poverty and health within nations as
well as across them. Poverty remains one of the most important root
causes of poor health worldwide, and the global burden of poverty
continues to be high. Among the 7.3 billion people alive in 2015, 10%
(736 million) lived on less than $1.90 (in 2011 U.S. dollars) per day—a
standard measurement of extreme poverty—and 25% lived on less than
$3.20 per day. Just under one-fifth of all children under the age of 14
worldwide lived in extreme poverty in 2015. The extreme poverty rate
has declined steadily since 1990, and by 2015, there were more than
one billion fewer people living in poverty despite growth in the global
population of more than two billion during that time period. However, improvement has been uneven across the globe. In sub-Saharan
Africa, people living in extreme poverty increased from 278 million to
413 million during the same period.
■ RISK FACTORS FOR DISEASE BURDEN
The GBD study found that the three leading risk factors for global disease burden in 2017 were (in order of frequency) high systolic blood
pressure, smoking, and high fasting plasma glucose—a substantial
change from 1990, when child wasting was ranked first. Although
ranking ninth in 2017, child wasting remains the third leading risk
factor for death worldwide among children under 5 years of age. In
an era that has seen obesity become a major health concern in many
developed countries—and the fourth leading risk factor worldwide—
the persistence of undernutrition is cause for consternation. Child
wasting is still a dominant risk factor for disease burden in sub-Saharan
Africa. In its rural reaches, no health care initiative, however generously funded, will be effective or comprehensive without addressing
undernutrition.
In an analysis that examined how specific diseases and injuries are
affected by environmental risk, the WHO estimated that 24% of all
deaths and 28% of deaths among children <5 years of age in 2016 were
due to modifiable environmental factors: some 1.6 million children die
every year from causes related to unhealthy environments, including
the nearly 300,000 deaths stemming from a lack of access to clean
water and sanitation. Many of these modifiable factors lead to child
and adult deaths from infectious pathologies; others lead to deaths
from malignancies. Etiology and nosology are increasingly difficult to
parse with regard to environmental harm. Risk factors such as indoor
1 Neonatal disorders
1990 Rank 2017 Rank
Global
Both sexes, all ages, DALYs
2 Lower respiratory infect
3 Diarrheal diseases
4 Ischemic heart disease
5 Stroke
6 Congenital defects
7 Road injuries
8 Tuberculosis
9 COPD
10 Measles
11 Malaria
12 Low back pain
13 Protein-energy malnutrition
14 Drowning
15 Self-harm
16 Meningitis
17 Headache disorders
18 Dietary iron deficiency
19 Diabetes
20 Cirrhosis
21 Depressive disorders
23 Falls
24 Lung cancer
28 HIV/AIDS
1 Neonatal disorders
Communicable, maternal, neonatal,
and nutritional diseases
Non-communicable diseases Injuries
2 Ischemic heart disease
3 Stroke
4 Lower respiratory infect
5 COPD
6 Diarrheal diseases
7 Diabetes
8 Road injuries
9 Low back pain
10 Congenital defects
11 HIV/AIDS
12 Headache disorders
13 Malaria
14 Tuberculosis
15 Depressive disorders
16 Cirrhosis
17 Lung cancer
18 Falls
21 Self-harm
24 Dietary iron deficiency
32 Meningitis
39 Drowning
41 Protein-energy malnutrition
67 Measles
FIGURE 472-1 Global disability-adjusted life-year (DALY) ranks for the top causes of disease burden in 1990 and 2017. COPD, chronic obstructive pulmonary disease.
(From the Institute for Health Metrics and Evaluation [IHME]. GBD Compare. Seattle, WA: IHME, University of Washington, 2017. Available at http://vizhub.healthdata.org/
gbd-compare. Accessed May 1, 2020.)
3706 PART 17 Global Medicine
TABLE 472-1 Leading Causes of Burden of Disease (DALYs), 2017
DISEASE OR INJURY
DALYs
(MILLIONS)
PERCENTAGE OF
TOTAL DALYs
World 2464.9 100
1. Neonatal disorders 185.8 7.4
2. Ischemic heart disease 170.3 6.8
3. Stroke 132.1 5.3
4. Lower respiratory infection 106.5 4.3
5. COPD 81.6 3.3
6. Diarrheal diseases 81.0 3.2
7. Diabetes 67.9 2.7
8. Road injuries 67.8 2.7
9. Low back pain 64.9 2.6
10. Congenital defects 60.9 2.4
Low- and Middle-Income Countriesa
1. Neonatal disorders 179.7 8.3
2. Ischemic heart disease 145.3 6.7
3. Stroke 117.1 5.4
4. Lower respiratory infection 101.1 4.7
5. Diarrheal disease 79.9 3.7
6. COPD 69.4 3.2
7. Road injuries 60.3 2.8
8. Congenital defects 57.2 2.6
9. Diabetes 56.0 2.6
10. HIV/AIDS 53.6 2.5
High-Income Countriesa
1. Ischemic heart disease 24.3 7.3
2. Low back pain 19.0 5.7
3. Stroke 14.2 4.3
4. Lung cancer 12.2 3.7
5. COPD 11.8 3.6
6. Diabetes 11.7 3.5
7. Alzheimer’s disease 10.9 3.3
8. Headache disorders 10.1 3.0
9. Falls 8.9 2.7
10. Drug use disorders 8.4 2.5
Sub-Saharan Africa
1. Neonatal disorders 66.8 12.8
2. Lower respiratory infection 44.7 8.6
3. HIV/AIDS 41.8 8.0
4. Malaria 40.1 7.7
5. Diarrheal diseases 39.3 7.5
6. Congenital defects 19.9 3.8
7. Tuberculosis 16.9 3.2
8. Meningitis 12.6 2.4
9. Protein-energy malnutrition 10.7 2.0
10. Road injuries 10.2 2.0
a
The World Bank classifies high-income countries as those whose gross national income
(GNI) per capita is ≥$12,376. Low- and middle-income countries are categorized as low
income (GNI per capita, <$1025), lower-middle income (GNI per capita, $1026–$3995), and
upper-middle income (GNI per capita, $3996–$12,375) (http://data.worldbank.org/about/
country-classifications).
Abbreviations: COPD, chronic obstructive pulmonary disease; DALYs, disability-adjusted
life-years.
Source: Institute for Health Metrics and Evaluation, University of Washington (2020). Data
available at http://www.healthdata.org/gbd/data-visualizations. Accessed April 30, 2020.
TABLE 472-2 Leading Causes of Death Worldwide, 2017
DISEASE OR INJURY
DEATHS
(MILLIONS)
PERCENTAGE OF
TOTAL DEATHS
World 55.8 100
1. Ischemic heart disease 8.9 15.9
2. Stroke 6.2 11.0
3. COPD 3.2 5.7
4. Lower respiratory infection 2.6 4.6
5. Alzheimer’s disease 2.5 4.5
6. Lung cancer 1.9 3.4
7. Neonatal disorders 1.8 3.2
8. Diarrheal diseases 1.6 2.8
9. Diabetes 1.4 2.4
10. Cirrhosis 1.3 2.4
Low- and Middle-Income Countriesa
1. Ischemic heart disease 7.2 15.8
2. Stroke 5.3 11.7
3. COPD 2.7 6.0
4. Lower respiratory infection 2.1 4.7
5. Neonatal disorders 1.8 3.9
6. Diarrheal diseases 1.5 3.4
7. Alzheimer’s disease 1.4 3.2
8. Lung cancer 1.2 2.7
9. Tuberculosis 1.2 2.6
10. Diabetes 1.1 2.5
High-Income Countriesa
1. Ischemic heart disease 1.7 16.6
2. Alzheimer’s disease 1.0 10.1
3. Stroke 0.8 7.9
4. Lung cancer 0.6 6.0
5. COPD 0.5 4.6
6. Lower respiratory infection 0.4 4.1
7. Colorectal cancer 0.4 3.4
8. Chronic kidney disease 0.3 2.4
9. Diabetes 0.2 2.1
10. Cirrhosis 0.2 2.0
Sub-Saharan Africa
1. Neonatal disorders 0.7 9.4
2. HIV/AIDS 0.7 9.3
3. Lower respiratory infection 0.7 9.2
4. Diarrheal diseases 0.6 7.3
5. Malaria 0.5 7.1
6. Ischemic heart disease 0.4 5.4
7. Tuberculosis 0.4 5.2
8. Stroke 0.3 4.4
9. Congenital defects 0.2 2.7
10. Road injuries 0.2 2.2
a
The World Bank classifies high-income countries as those whose gross national
income (GNI) per capita is ≥$12,376. Low- and middle-income countries are
categorized as low income (GNI per capita, <$1025), lower-middle income (GNI per
capita, $1026–$3995), and upper-middle income (GNI per capita, $3996–$12,375)
(http://data.worldbank.org/about/country-classifications).
Abbreviation: COPD, chronic obstructive pulmonary disease.
Source: Institute for Health Metrics and Evaluation, University of Washington (2020).
Data available at http://www.healthdata.org/gbd/data-visualizations. Accessed
April 30, 2020.
air pollution due to use of solid fuels, exposure to secondhand tobacco
smoke, and outdoor air pollution account for 55% of DALYs due to
lower respiratory infections globally. Various forms of unintentional
injury and malaria top the list of health problems to which environmental factors contribute.
The third edition of Disease Control Priorities (DCP3), published as
a set of serial volumes based on content area, provides evidence-based
recommendations and cost-effectiveness analyses for numerous interventions, with attention to strategies for strengthening health systems. Cost-effectiveness analyses that compare relatively equivalent
3707 Global Issues in Medicine CHAPTER 472
FIGURE 472-2 An HIV and tuberculosis (TB)–co-infected patient in Rwanda before (left) and
after (right) 6 months of treatment.
interventions in order to facilitate sound decisions under constraint are
necessary; however, these analyses, as the DCP3 authors acknowledge,
are unreliable when based on an incomplete knowledge of cost and
evolving evidence of effectiveness. As both resources and objectives for
global health initiatives grew, cost-effectiveness analyses (particularly
those based on older evidence) sometimes steered policy makers and
public health experts toward low-cost but ultimately ineffective interventions or away from higher-priced but effective ones. Thus, we use
the term global health equity to emphasize the need to ensure equitable
access to high-value health interventions. To illustrate these points,
it is instructive to look to HIV/AIDS, which in the course of the last
four decades has become one of the world’s leading infectious causes
of adult death.
■ HIV INFECTION/AIDS
Chapter 202 provides an overview of the global HIV epidemic today.
Approximately 36 million people worldwide were living with HIV
infection in 2017, and it has been the underlying cause of death for
almost 1 million people—especially concentrated in sub-Saharan
Africa—annually. As of 2017, 54 countries were on track to meet the
2020 target of 81% antiretroviral therapy (ART) coverage, but only
12 countries are expected to meet the 2030 target of 90% coverage.
Here the discussion will be limited to HIV/AIDS in the developing
world. Lessons learned from tackling HIV/AIDS in low-resource
settings are highly relevant to discussions of other chronic diseases,
including NCDs, for which effective therapies have been developed. In
the United States, after the mid-1990s, ART transformed HIV infection
from an inescapably fatal disease into a manageable chronic illness.
Across high-income countries, improved ART has dramatically prolonged life expectancy for people living with HIV infection, which now
approaches that of the general population. This success rate exceeds
that obtained with almost any treatment for adulthood cancer or for
complications of coronary artery disease. In developing countries,
treatment has been offered broadly only since 2003. Before 2003, many
arguments were raised to justify not moving forward rapidly with
ART programs for people living with HIV/AIDS in resource-limited
settings. The standard litany included the price of therapy compared
with the poverty of patients, the complexity of the intervention, the
lack of infrastructure for laboratory monitoring, and the lack of
trained health care providers. Narrow cost-effectiveness arguments
that created false dichotomies—prevention or treatment rather than
their synergistic integration—too often went unchallenged
by policy makers, public health experts, and health economists. As a cumulative result of these delays in the face of
health disparities both old and new, there were millions of
premature deaths.
Disparities in access to HIV treatment did give rise to
widespread moral indignation and a new type of health
activism. In several middle-income countries, including
Brazil, public programs have helped bridge the global
access gap. Other innovative projects pioneered by international NGOs in diverse settings such as Haiti and
Rwanda have established that a simple approach to ART
based on intensive community engagement and social
and economic support for patients and their community-based health workers can achieve remarkable results
(Fig. 472-2).
During the past decade, the availability of ART has
increased sharply in the low- and middle-income countries
that have borne the greatest burden of the HIV/AIDS pandemic. In 2000, few people living with HIV/AIDS in these
nations had access to ART, whereas by 2018, 62% of people
living with HIV infection were receiving ART, including
64% of people living with HIV in sub-Saharan Africa and
53% in Southeast Asia. In light of these dramatic gains,
coverage targets have grown more ambitious; for example,
in 2014, UNAIDS set the 90-90-90 targets, which aimed to
have 90% of people living with HIV know their status, 90%
of those with HIV treated with ART, and 90% of those on
treatment achieving viral load suppression by 2020. These are to be
followed by the 95-95-95 targets for 2030, with the objective of ending
the AIDS epidemic by 2030.
This scale-up was made possible by several developments: a staggering drop in the cost of generically manufactured ART, the development
of a standardized approach to treatment, substantial investments by
funders, and the political commitment of governments to afford ART
as a public good. Civil-society AIDS activists spurred many of these
efforts.
Starting in the early 2000s, a combination of factors, including work
by the Clinton HIV/AIDS Initiative and Médecins Sans Frontières,
led to the availability of generic ART medications. While first-line
ART cost >$10,000 per patient per year in 2000, first-line regimens in
low- and middle-income countries are now available for <$75 per year.
At the same time, fixed-dose combinations made multidrug regimens
easier to administer. Also around this time, the WHO began advocating a public health approach to the treatment of people with AIDS in
low-resource settings; this approach promised—thanks to dropping
viremia—to lower transmission rates and, if universally available, to
end almost all mother-to-child transmission. Derived from models of
care pioneered by the NGO Partners In Health and other groups, this
approach proposed the use of standard first-line treatment regimens
based on a simple five-drug formulary, with a more complex (and
more expensive) set of second-line options in reserve. Clinical protocols were standardized, and intensive training packages for health
professionals and community health workers were developed and
implemented in many countries. Early rollout efforts were supported
by new funding from the Global Fund and PEPFAR. In 2003, lack
of access to ART was declared a global public health emergency by
the WHO and UNAIDS, and those two agencies launched the 3 by
5 Initiative, setting an ambitious target: to have 3 million people in
developing countries on treatment by the end of 2005. Many countries
set corresponding national targets and have worked to integrate ART
into their national AIDS programs and health systems and to harness
the synergies between HIV/AIDS treatment and prevention activities.
External funding to fight HIV/AIDS in such settings increased dramatically during this period and beyond, rising from $300 million in 1996
to $9.1 billion in 2017. The integration of prevention and care led to
a sharp drop in transmission—a 96% decline according to one review
of the impact of ART rollout in heavily burdened countries in Africa
and the Caribbean.
No comments:
Post a Comment
اكتب تعليق حول الموضوع