Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Clostridioides difficile Infection in

Children: Recent Updates on


Epidemiology, Diagnosis, Therapy
Debbie-Ann Shirley, MD, MPH,a William Tornel,a Cirle A. Warren, MD,b,c Shannon Moonah, MD, ScMb

Clostridioides (formerly Clostridium) difficile is the most important infectious abstract


cause of antibiotic-associated diarrhea worldwide and a leading cause of
healthcare-associated infection in the United States. The incidence of C. difficile
infection (CDI) in children has increased, with 20 000 cases now reported
annually, also posing indirect educational and economic consequences. In
contrast to infection in adults, CDI in children is more commonly community-
associated, accounting for three-quarters of all cases. A wide spectrum of
disease severity ranging from asymptomatic carriage to severe diarrhea can
occur, varying by age. Fulminant disease, although rare in children, is associ-
ated with high morbidity and even fatality. Diagnosis of CDI can be challenging
as currently available tests detect either the presence of organism or disease- a
Pediatric Infectious Diseases, Department of Pediatrics,
causing toxin but cannot distinguish colonization from infection. Since coloni- b
Infectious Diseases and International Health, Department
of Medicine, and cComplicated C. difficile Clinic, UVA Health,
zation can be high in specific pediatric groups, such as infants and young
University of Virginia, Charlottesville, Virginia
children, biomarkers to aid in accurate diagnosis are urgently needed. Similar
Drs Shirley and Moonah conceptualized the review,
to disease in adults, recurrence of CDI in children is common, affecting 20% to drafted the manuscript, and reviewed and revised the
manuscript; Mr Tornel contributed to the design and
30% of incident cases. Metronidazole has long been considered the mainstay helped to draft the manuscript; Dr Warren critically
therapy for CDI in children. However, new evidence supports the safety and ef- reviewed the manuscript for important intellectual
content; and all authors approved the final manuscript
ficacy of oral vancomycin and fidaxomicin as additional treatment options, as submitted and agree to be accountable for all
whereas fecal microbiota transplantation is gaining popularity for recurrent in- aspects of the work.

fection. Recent advancements in our understanding of emerging epidemiologic DOI: https://doi.org/10.1542/peds.2023-062307

trends and management of CDI unique to children are highlighted in this re- Accepted for publication May 31, 2023

view. Despite encouraging therapeutic advancements, there remains a pressing Address correspondence to Debbie-Ann Shirley, MD, MPH,
Department of Pediatrics, PO Box 800386, Charlottesville,
need to optimize CDI therapy in children, particularly as it pertains to severe VA 22908. E-mail: ds3ru@virginia.edu
and recurrent disease. PEDIATRICS (ISSN Numbers: Print, 0031-4005; Online,
1098-4275).
This is an open access article distributed under the terms of
Clostridioides (formerly Clostridium) difficile is a spore-forming, anaerobic, the Creative Commons Attribution-NonCommercial-
NoDerivatives 4.0 International License (http://
Gram-positive bacillus first described in 1935 from the stool of healthy new- creativecommons.org/licenses/by-nc-nd/4.0/), which permits
born infants.1 Initially named Bacillus difficile, because it was difficult to isolate, noncommercial, distribution, and reproduction in any me-
dium, provided the original author and source are credited.
with time this pathogen has also proven difficult to treat and control. Most
FUNDING: This work was supported by the Hartwell
C. difficile bacteria secrete toxin A and toxin B. The presence of toxin B appears Foundation and the NIH, R01 DK131313 and R34
to be the primary cause of diarrhea, and sufficient by itself to induce disease, AI165304. The Hartwell Foundation and the NIH had no
role in the design and conduct of the study.
whereas other virulence factors, such as binary toxin, surface layer protein, and
CONFLICT OF INTEREST DISCLOSURES: Dr Warren is a
biofilm formation, may contribute to colonization, transmission, or disease medical advisor for SER-109. The rest of the authors
severity. have indicated they have no conflicts of interest
relevant to this article to disclose.
C. difficile has risen to become a leading cause of healthcare-associated infec-
tion among adult populations in the United States, resulting in an estimated
half-million attributable cases each year, leading to 223 900 people requiring To cite: Shirley D-A, Tornel W, Warren CA, et al.
hospital care and 12 800 deaths.2,3 CDI prolongs hospital stay and increases Clostridioides difficile Infection in Children: Recent
Updates on Epidemiology, Diagnosis, Therapy.
healthcare expenses, costing the US healthcare system at least 1 billion dollars Pediatrics. 2023;152(3):e2023062307
annually.3 The US Centers for Disease Control and Prevention (CDC) have

PEDIATRICS Volume 152, number 3, September 2023:e2023062307 STATE-OF-THE-ART REVIEW


Downloaded from http://publications.aap.org/pediatrics/article-pdf/doi/10.1542/peds.2023-062307/1523499/peds.2023-062307.pdf
by guest
identified C. difficile as an urgent threat, listing it as one EPIDEMIOLOGY
of the top 5 drug-resistant pathogens in need of aggres-
sive action.3 Incidence and Prevalence
Although CDI is more common and tends to be more The incidence of CDI as a cause of diarrhea in children has
severe in older adults, the burden of C. difficile to chil- increased over time, though the exact magnitude of this
dren is also significant (Fig 1). C. difficile is the most im- problem is challenging to quantify given limitations in diag-
portant infectious cause of antibiotic-associated diarrhea nosis.4,12,13 CDI is classified as healthcare-associated (onset
in children, contributing approximately 20 000 infections more than 3 days after hospitalization) or community-asso-
in the United States each year.4,5 Rates of CDI in children ciated (onset within 3 days of hospitalization provided no
have increased over the past 3 decades and the rate of documented overnight stay in a healthcare facility in the
pediatric hospitalizations resulting from or complicated preceding 12 weeks). CDI, considered mostly a healthcare-
by C. difficile increased 57% from 1997 to 2006.5,6 There associated infection in adults, contrasts as most commonly
is some more recent indication of leveling rates, particu- community-associated in children, nonetheless community-
larly in the inpatient setting.2,7,8 Changes in testing, diag- associated CDI rates have increased in both adults and
nosis, treatment and strain type can all contribute to children.2,4 Population-based surveillance conducted by the
fluctuations in incidence. Nonetheless, CDI-associated hospi- CDC Emerging Infections Program estimates the incidence
talizations cost 1.6 times more than non-CDI associated of community-associated CDI in children was as high as
hospitalizations in children, and CDI adds an additional 25.8 per 100 000 in 2019, accounting for 75% of cases,
4 days to length of stay for pediatric hospitalizations.9 compared with 9.0 per 100 000 reported as healthcare-as-
Broader, less well-described indirect consequences of CDI on sociated (Fig 1).12 Diversion of public health resources dur-
child health also bear consideration, including educational ing the coronavirus disease 2019 pandemic has since led to
consequences from lost days of schooling and economic delays in subsequent surveillance estimates.14
impact from missed parental days of work.10 The global impact of CDI in children is even harder to
The Infectious Diseases Society of America (IDSA) and quantify given even wider variations in diagnostic ap-
the Society for Healthcare Epidemiology of America proach.15 Regardless, high rates of CDI in children are re-
(SHEA) updated CDI clinical practice guidelines in 2017, ported worldwide. A recent cross-sectional study conducted
including, for the first time, specific recommendations re- among children presenting with acute gastroenteritis at a
garding CDI diagnosis and therapeutic considerations in tertiary children’s hospital in Zhejiang, China, for example,
children.11 Our understanding of CDI in children has con- reported 14.3% of cases solely attributed to CDI.16 Whereas,
tinued to grow since the release of these guidelines. This
C. difficile was detected from 19.7% of children being evalu-
review summarizes recent updates pertaining to the ep-
ated for diarrhea in Perth, Australia.16,17
idemiology, diagnosis, and management of CDI in chil-
dren, with a principle focus on developments over the Age
past 5 years.
The prevalence of detection of C. difficile in the stools of
children varies by age. Infants have high frequencies of
asymptomatic carriage, exceeding 40% within the first
year.18 Colonization, which begins shortly after birth,
peaks at 6 to 12 months of age.19–21 The frequency re-
mains as high as 22% in toddlers 1 to 2 years of age be-
fore declining to approach rates seen in healthy adults of
1% to 3%.11 This finding is consistent even among re-
source-limited settings, where detection of C. difficile is
also prevalent during the first year of life, then declining
during the second year of life, though cumulative inci-
dence varies by country.22
For reasons unknown, infants appear almost univer-
sally protected from clinical disease despite these high
colonization rates, even in the presence of toxin produc-
ing strains. Only rare cases of CDI in infants are repor-
ted,23 but other age-related causes such as necrotizing
enterocolitis may still be at play. A popularly purported
reason that infants do not develop diarrhea from CDI is
FIGURE 1 the lack of expression of receptors for binding C. difficile
The impact of Clostridioides difficile infection in children. toxins, based on limited animal data from the intestines

2 SHIRLEY et al
Downloaded from http://publications.aap.org/pediatrics/article-pdf/doi/10.1542/peds.2023-062307/1523499/peds.2023-062307.pdf
by guest
of newborn rabbits, however further corroboration in hu- Given that CDI is mostly community-associated in chil-
mans is required to substantiate this observation.24 Al- dren, recent attention has focused on identifying specific
though protection from passive transplacental transfer of risk factors for this setting. In a large case-control study
maternal antibody is also unlikely the explanation based conducted by the Kaiser-Permanente system, risk factors
on maternal cord blood sampling, colonization may be for community-associated CDI in children included non-
important for acquired protection later in life, as natural Hispanic ethnicity, antibiotic exposure to amoxicillin-
immunization against toxin A and B does seem to occur clavulanate, cephalosporins or clindamycin within the
following colonization, irrespective of feeding method, previous 12 weeks, a previous positive C. difficile test
though the durability of this protection is unknown.25 within 6 months, and increased health care visits within
Beyond infancy and toddlerhood, other specific pediat- the last year.4 However, traditional risk factors may not
ric populations are predisposed to higher rates of coloni- always be present and 13.6% of children lacked any
zation, including those with inflammatory bowel disease, identifiable risk factor in another multisite case-control
cystic fibrosis, and cancer as well as transplant recipi- study of younger children with community-associated
ents.26–30 Up to one-quarter of hospitalized children may CDI.34
be asymptomatically colonized31 and one-third or more Transmission
of pediatric oncology patients may have asymptomatic C.
Transmission of C. difficile occurs fecal-orally through inges-
difficile colonization on admission. In these older age
tion of spores, including by contact with contaminated
groups, colonization serves as a risk factor for subse-
surfaces in the environment. Whole genome sequencing in-
quent infection as CDI is more common in children with
dicates that transmission of C. difficile among symptomatic
chronic medical conditions. For example, nearly all clini-
children within healthcare settings may be less common
cal isolates from pediatric oncology patients diagnosed
than in adult populations. Only about 12% of healthcare-
with CDI during hospitalization were identical to their
associated transmission events could be linked to another
baseline admission isolates by pulsed-field gel electro- symptomatic patient at a medical center that implemented
phoresis analysis; the caveat here being that diarrhea in contact isolation for all patients with diarrhea.38 Many chil-
children with cancer can be multifactorial, and it may not dren with healthcare associated CDI are already colonized
always be possible to clinically distinguish the primary with C. difficile at the time of admission.18 Infants, young
etiology.18 children, and household contacts with active CDI may serve
Risk Factors as possible sources of community-associated C. difficile ex-
posure.18,39,40 Potential transmission from companion ani-
Antibiotic exposure is the most important predisposing mals has also been implicated.41
factor to the development of CDI.32–34 Diarrhea in gen-
eral is a common side effect of antibiotics, occurring in Molecular Epidemiology
more than one third of patients who receive them, with Ribotyping, a molecular technique often used for surveil-
mechanisms such as osmotic effect and promotility con- lance purposes, characterizes C. difficile strains based on
tributing. Antibiotics may specifically predispose to CDI specific differences in ribosomal RNA. C. difficile ribotype
through disruptions in intestinal microbial composition 106 has emerged as the most common strain in the
and metabolism, a state frequently referred to as dysbio- United States, superseding the hypervirulent ribotype
sis. Through complex interactions, the distorted micro- 027 strain (also known as BI/NAP1), which was previ-
bial gut community can selectively promote survival of C. ously the most common.12 In 2018, ribotype 106 ac-
difficile.35 For example, enterococci are highly abundant counted for about 16% of community-associated and
in the stools of children with CDI. Enterococci may pro- 12% of healthcare-associated infections in the United
duce fermentable amino acids, such as leucine and orni- States.12 Ribotype 106 is also the most common strain in
thine, whereas depleting arginine, leading to metabolic the pediatric population.42 Globally, ribotype 014 (ST2,
alterations in the gut that may further promote CDI.36 ST13, ST49/clade 1) appears to be common.43
Any antibiotic class can potentially predispose to CDI and
onset can occur within days to weeks of exposure. In CLINICAL
particular, third generation cephalosporins, clindamycin, C. difficile causes a wide spectrum of disease severity,
fluoroquinolones, and amoxicillin-clavulanate are strong ranging from asymptomatic carriage all the way to life
associations.33,37 Several other factors that predispose to threatening colitis. Diarrhea is thought to primarily result
CDI are described, including administration of proton from toxin-mediated intestinal epithelial cytotoxicity and
pump inhibitors, tube feeds, healthcare facility exposure, inflammation. Most symptomatic children will have mild-
prolonged hospitalization, and contact with a person in- to-moderate diarrhea. Severe CDI, defined by leukocyto-
fected with C. difficile.32,33 sis ($15, 000 cells/mm3) or renal impairment (serum

PEDIATRICS Volume 152, number 3, September 2023 3


Downloaded from http://publications.aap.org/pediatrics/article-pdf/doi/10.1542/peds.2023-062307/1523499/peds.2023-062307.pdf
by guest
creatinine >1.5 mg/dL) in adults, is poorly defined in several commercial tests available for diagnosis that ei-
children, necessitating clinical judgement to diagnose. ther detect the presence of C. difficile organism or toxin
Severe complications of CDI in children include develop- production, but each has limitations in application and
ment of pseudomembranous colitis, pneumatosis intesti- there is no gold standard (Table 1). Tests that detect
nalis, toxic megacolon, perforation, peritonitis, and shock the organism, such as nucleic acid amplification tests
with multisystem failure.44 Severe CDI is less common in (NAATs), are highly sensitive but do not distinguish colo-
children than adults, but does occur, and up to 8% of nization from infection. Tests that detect toxin, such as
will develop severe disease.45 By other estimates, up to enzyme immunoassays, are more specific but lack suffi-
0.1% to 1.2% may require colectomy, and all-cause mor- cient sensitivity for diagnosis. Detection of toxin does not
tality in children with CDI is about 2% to 4%.7 Imaging correlate with severity of symptoms in children nor can
and endoscopic findings of CDI are nonspecific. Disease stool toxin concentration reliably distinguish carriage
in children is often cecocolonic or colonic. In severe from infection.51 A prospective study of asymptomatic
cases, abdominal imaging may show colonic wall thicken- children with cancer, cystic fibrosis, or inflammatory
ing, colonic dilation or free air in the case of perforation bowel disease in which 21% were colonized with C. diffi-
(Fig 2). cile demonstrated that toxin detection did not differ
when compared with a cohort of children with symptom-
RECURRENT CDI atic CDI.52 Hence, correct interpretation of C. difficile test
results ultimately requires careful consideration of pa-
Recurrent CDI (rCDI) is as common in children as adults,
tient factors. Consequences of misinterpretation include
occurring in about 20% to 30% of cases despite treat-
unnecessary antibiotic exposure, potential drug adverse
ment.11,46,47 Recurrence is defined as new onset of CDI
effects, and delay in diagnosis of the true cause of
symptoms with a positive C. difficile specific laboratory
diarrhea.
test following an incident episode in the previous 2 to 8
Adherence to several strategies of diagnostic steward-
weeks. Ongoing dysbiosis perpetuates survival and prolif-
ship can further improve the predictive value of testing.
eration of C. difficile, leading to relapse or reinfection.48
First, diarrheal stool should preferentially only be col-
Risk factors for recurrence include the presence of co-
lected from patients with unexplained and new-onset di-
morbidities such as cancer, IBD, medical technology de-
arrhea, defined as 3 or more unformed stools within a
pendence, recent surgery, and antibiotic exposure.49 The
24 hour period.11 An exception to this would be in pa-
risk of additional recurrent episodes increase with each
tients with ileus or toxic megacolon suspected to be sec-
recurrence. Interestingly, nearly one-fourth of children
ondary to severe CDI. Second, testing should be avoided
referred for fecal microbiota transplantation (FMT) be-
when a more likely cause of diarrhea, such as laxative
cause of rCDI at one institution were found to have an al-
use, is apparent. Following similar reasoning, other more
ternative diagnosis, such as constipation or overflow
likely causes should be evaluated before testing children
diarrhea or inflammatory bowel disease, emphasizing the
under 2 years of age, and testing should generally be
need for careful consideration of alternative causes of re-
avoided in infants less than 1 year of age, given the high
current loose stools when faced with this dilemma.50
rates of asymptomatic colonization in these young age
groups. In a multicenter investigation of children under 2
LABORATORY DIAGNOSIS years of age, C. difficile detection by polymerase chain re-
The diagnosis of CDI in children is challenging, and de- action was twice as common in asymptomatic children
tection does not always equate to diagnosis. There are (28%) as in those presenting with acute gastroenteritis

FIGURE 2
Severe Clostridioides difficile infection in children. (A) Abdominal radiograph showing thickened colon most prominent in the transverse co-
lon. (B) Abdominal sonogram showing hypoechoic diffuse large bowel wall thickening. (C) CT Abdomen and pelvis showing markedly thick-
ened colon wall in an adolescent with fulminant C. difficile colitis.

4 SHIRLEY et al
Downloaded from http://publications.aap.org/pediatrics/article-pdf/doi/10.1542/peds.2023-062307/1523499/peds.2023-062307.pdf
by guest
TABLE 1 Summary of Clostridioides difficile Testinga
Test Target Strengths Limitations
Tests to detect presence of C. difficile organism
GDH EIA Detects C. difficile-specific enzyme  High sensitivity;  Low specificitya;
 rapid, takes hours to complete;  does not distinguish toxigenic
 inexpensive from nontoxigenic strains
NAATs Detects genes for C. difficile toxin  High sensitivity;  Lower positive predictive value;
(toxin A/B)  good specificity;  does not detect presence of
 can be rapid toxin
Toxigenic culture Detects toxin producing C. difficile  High sensitivity;  Technically complex to perform;
strains  high specificity;  takes days to complete;
 allows for antimicrobial  used mostly as reference for
susceptibility testing validation or in research
settings
Tests to detect presence of C. difficile toxin
Toxin EIA Detects free toxin in stool (toxin A/B)  High specificity;  Low sensitivity (variable but as
 rapid, takes hours to complete; low as 35%)111
 inexpensive
CCNA Demonstrates free toxin B  High sensitivity;  Technically complex to perform;
 high specificity  takes day to complete;
 used mostly as reference for
validation or in research
settings
a
The 2017 IDSA and SHEA guideline recommends a 2-step algorithm (ie, glutamate dehydrogenase EIA plus toxin EIA, arbitrated by NAAT or NAAT plus toxin EIA) as the best diag-
nostic approach.11
CCNA, cell culture cytotoxicity neutralization assay; EIA, enzyme immunoassay; GDH, Glutamate dehydrogenase; NAATs, nucleic acid amplification tests.

(14%), exemplifying the worth of incorporating age strat- institutions to improve appropriate test ordering.58 Gaining
ification in testing approach.53 Third, prolonged carriage in popularity, the use of multiplex gastrointestinal polymer-
can occur despite treatment, so repeat testing for proof ase chain reaction panels that include C. difficile targets may
of test of cure is not helpful.54 unintentionally increase inappropriate testing, so some labo-
The recommended diagnostic approach has evolved ratories preferentially suppress C. difficile results from the
with time from toxin-based assays to NAAT-based testing panel unless specifically requested, as another way to limit
and currently to multistep testing. As the best-perform- over testing in populations with low prevalence of CDI.59
ing approach for diagnosis, the 2017 IDSA and SHEA The search for novel stool biomarkers to improve diagnosis
guidelines recommend using a stool toxin test as part of is an ongoing and pressing need. Measurement of fecal
a 2-step algorithm (Fig 3), unless institutions have devel- metabolites,60 proinflammatory cytokines,61 and volatile
oped other agreed upon clinical and laboratory criteria organic compounds62 all hold promise as potential future
for test submission, in which case detection of toxigenic makers.
C. difficile by NAAT alone is considered acceptable.11
More recently in 2021, the American College of Gastroen- MANAGEMENT
terologists developed guidelines for the preferred man- The management of CDI is determined by the number
agement of CDI in adults, which are similar but differ and severity of episodes. Children who do not respond to
slightly to the 2017 IDSA and SHEA guidelines by recom- targeted treatment within 5 days should be re-evaluated
mending use of testing algorithms that include a highly for other causes. There is currently insufficient evidence
sensitive and a highly specific testing modality to distin- to support probiotics as an alternative to antimicrobial
guish colonization from active infection.55 Despite socie- therapy for CDI treatment.63
tal guidance, testing practices for diagnosis of C. difficile
in children varies considerably, suggesting further oppor- First Episode
tunities to improve diagnosis exist. Judicious testing of C. For symptomatic children, initial management should include
difficile ranks among one of the top 12 high-priority re- discontinuation of the inciting antibiotic, when possible, to
search topics in healthcare-associated infections and anti- limit further intestinal microbial disruption. Current antimi-
microbial stewardship by the CDC.56,57 Incorporation of crobial therapies targeted against the initial CDI episode of
clinical decision support around test ordering in elec- mild or moderate severity include oral metronidazole and
tronic health records and preauthorization of testing are oral vancomycin (Table 2). Metronidazole, which has been a
additional strategies successfully implemented by some mainstay treatment of pediatric CDI, was removed as a

PEDIATRICS Volume 152, number 3, September 2023 5


Downloaded from http://publications.aap.org/pediatrics/article-pdf/doi/10.1542/peds.2023-062307/1523499/peds.2023-062307.pdf
by guest
FIGURE 3
A laboratory testing approach for diagnosis of Clostridioides difficile infection in children.a
a
Based on the 2017 IDSA and SHEA guideline, which recommends a 2-step algorithm, including s stool toxin test (ie, glutamate dehydroge-
nase EIA plus Toxin EIA, arbitrated by NAAT or NAAT plus toxin EIA) as the best diagnostic approach.11 Discordant results require further clin-
ical evaluation. c/w, consistent with; CDI, Clostridioides difficile infection; EIA, enzyme immunoassay; GDH, glutamate dehydrogenase; ID,
Infectious Disease; NAAT, nucleic acid amplification test; Toxin A/B, Toxin A and B enzyme immunoassay; yo, years old.

recommended first line choice for adults in the 2017 IDSA 10 days. Children had to have negative rotavirus testing re-
and SHEA guidelines.11 Vancomycin and fidaxomicin are sults to be eligible, and children with inflammatory bowel
now recommended as preferred first line therapies in adults disease were excluded.70 No difference in clinical response at
based on improved clinical cure rates and lower recurrence the end of therapy was shown, however at 30 days after
risk.11,64–67 High quality data to support treatment guideline therapy, the rate of sustained response was higher with fi-
recommendation in children were lacking at the time, how- daxomcin (68%) compared with vancomycin (50%), with an
ever postguideline shifts in prescribing practice with de- adjusted treatment difference of 18.8% (95% confidence in-
creased use of metronidazole and increased use of oral terval 1.5%–35.3%). Children under 2 years of age were
vancomycin in children with CDI have been observed.7,68 included, so it is unclear if other etiologies contributed to
A retrospective, comparative effectiveness study of 192 diarrhea in younger participants.71 Regardless, based on
children hospitalized with nonsevere CDI has since shown these positive results, in 2020 the US Food and Drug Admin-
that those treated with vancomycin had earlier resolution of istration (FDA) expanded approval of fidaxomicin for treat-
symptoms (86.3%) compared with those treated with metro- ment of CDI to include children 6 months of age and older.
nidazole (71.1%) by day 5, whereas recurrence was simi- These 2 studies open up new therapeutic options in children.
lar.69 The recent landmark SUNSHINE study represents the Based on limited data extrapolated from care in adults,
first clinical trial conducted in children with CDI. In this phase children with more severe disease are treated with oral van-
3 trial, 142 children with CDI were age-stratified and ran- comycin, which can be administered via gastric feeding tube
domized to receive either oral fidaxomicin or vancomycin for in intubated children. Intravenous metronidazole is often

6 SHIRLEY et al
Downloaded from http://publications.aap.org/pediatrics/article-pdf/doi/10.1542/peds.2023-062307/1523499/peds.2023-062307.pdf
by guest
TABLE 2 Summary of Therapies for Children With Clostridioides difficile Infection^
Presentation Drug# Route Dose Frequency Duration
Initial infection
Nonsevere Metronidazole* or PO 7.5 mg/kg per dose TID or QID 10 d
(max 500 mg)
Vancomycin PO 10 mg/kg per dose QID 10 d
(max 125 mg)
Severea or fulminant Vancomycin PO or PR 10 mg/kg per dose QID 10 d
(max 500 mgb)
± Metronidazole IV 10 mg/kg per dose TID 10 d
(max 500 mg)
Recurrent infection
First recurrence Metronidazole* or PO 7.5 mg/kg per dose TID or QID 10 d
(max 500 mg)
Vancomycin PO 10 mg/kg per dose QID 10 d
(max 125 mg)
Second or Vancomycin or PO 10 mg/kg per dose QID 10 d (if not used previously)
subsequent (max 125 mg)
recurrencec Vancomycin tapered PO 10 mg/kg per dose QID 10–14 d, then BID x 7 d, then once daily × 7
and pulsed or (max 125 mg) d, then every 2–3 d for 2–8 wk
Vancomycin; followed PO 10 mg/kg per dose QID; TID 10 d; 20 d
by rifaximin chaser (max 125 mg); no
or pediatric dosing for
rifaximin0 (max 400 mg)
FMT under IND — — — —
^
Data based on the 2017 IDSA and SHEA guidelines.11 *Some experts would preferentially administer vancomycin over metronidazole for initial treatment or recurrence based
on accumulating data to suggest vancomycin may be more effective. # Fidaxomicin is a newly US FDA approved option for treatment in children, weight based dosing 16 mg/kg
per dose (max 200 mg per dose) BID x 10 d, available as tablet and liquid formulations for children $6 mo of age and $4 kg.111 'Rifaximin is not US FDA approved for use in
children <12 y of age. 1/– consider addition of. BID, twice daily; FDA, Food and Drug Administration; d, days; IV, intravenous; mg, milligrams; PO, by mouth; PR, per rectum; QID,
4 times daily; TID, 3 times daily.
a
No validated definition of severity in children, based on clinical judgment.
b
Consider serum trough levels when using higher dosing levels to prevent systemic toxicity.
c
Consider fecal microbiota transplantation under investigational use for patients with multiple recurrences following standard antibiotic treatments.

added on, though it is unclear whether colonic concentrations oral vancomycin in children (Table 2). Pulse-tapered vanco-
of metronidazole are adequate in this situation. There are no mycin regimens or vancomycin course followed by rifaximin
optimal treatments for fulminant disease. When complica- chaser can be prescribed for second or subsequent recur-
tions, such as hypotension, shock, ileus, or toxic megacolon rences after that. Secondary prophylaxis with oral vancomy-
arise, multiple interventions are instituted simultaneously in cin while receiving systemic antibiotics may reduce the risk
hopes of improving outcomes. When ileus is present, admin- of recurrent CDI in the highest risk pediatric patients with an
istration of vancomycin via retention enema can be at- established history of CDI.76,77 Bezlotoxumab, an antitoxin B
tempted. Surgical intervention, in the form of either organ- monoclonal antibody providing modest risk reduction, is cur-
preserving diverting loop ileostomy or more invasive colec- rently recommended in adults at high risk of recurrence.64,78
tomy or FMT are other deliberations.11,72,73 Further studies Results of the phase 3 trial in children are expected soon.
are needed to improve outcomes from severe disease. FMT, which involves transfer of stool from a healthy
The optimal management of children who require donor to the gastrointestinal tract of a recipient with
treatment with other concomitant antibiotics beyond the rCDI has gained popularity as a potential way to restore
recommended duration of CDI targeted therapy is un- disrupted intestinal microbial composition. Traditionally,
known. In practice, some prolong the course of CDI treat- donor stool is transferred via endoscopy or enteric cap-
ment, though relapse rates were similar in adults who sules for treatment, but enema may be an option if the
received extended CDI therapy beyond 14 days in retro- other methods are unavailable. In a large retrospective
spective review, and further studies are still needed.74,75 review of FMT in 335 patients performed at 18 pediatric
Prolonged use of metronidazole, which can be associated centers between 2014 and 2017, 81% of children were
with neurotoxicity, should be avoided. successfully treated, increasing to 87% with repeat FMT.
Predictors of successful outcome included use of fresh
Recurrent CDI donor stool over frozen stool, delivery by endoscopy, ab-
For the first recurrence, the 2017 IDSA and SHEA guidelines sence of a feeding tube, and lower number of recurrences
recommend retreatment can be with oral metronidazole or of CDI before FMT. Just over 5% experienced an adverse

PEDIATRICS Volume 152, number 3, September 2023 7


Downloaded from http://publications.aap.org/pediatrics/article-pdf/doi/10.1542/peds.2023-062307/1523499/peds.2023-062307.pdf
by guest
TABLE 3 Strengths and Limitations of Current C. difficile Therapies in Children
Therapeutic Strengths Limitations
Metronidazole  Established experience  Broad spectrum gut microbial activity with
recurrence risk;
 systemically absorbed;
 neurotoxic with prolonged or repeated use
Vancomycin  Established experience;  Broad spectrum gut microbial activity with
 not systemically absorbed recurrence risk;
 promotes resistant pathogens such as
vancomycin resistant enterococci
Fidaxomicin  Narrow spectrum gut activity;  Limited experience with use in children;
 not systemically absorbed  expensive;
 adverse effects of pyrexia, abdominal pain,
emesis, diarrhea, constipation, elevation in
liver enzyme activity and rash reported
Bezlotoxumab  No systemic activity;  Insufficient data in children;
 no affect on gut dysbiosis  caution in those with congestive heart failure;
 short-lived protection
Fecal microbiota transplantation  May help to restore gut microbial dysbiosis  Difficult to standardize;
 no long term safety data on consequences of
altering child microbiome yet available;
 risk of transmission of donor derived
infections or conditions

reaction related to FMT, including diarrhea, pain, and live purified Firmicutes bacterial spores from healthy donors.
bloating. There were 2 severe adverse events deemed re- In a trial of adults at risk for rCDI, lower recurrence rates at
lated to FMT reported, including an aspiration pneumo- 8 weeks after treatment were found compared with placebo.89
nia event post procedure and admission for emesis or These developments represent important steps forward toward
diarrhea from inflammatory bowel disease relapse in standardization of FMT, although they are several more steps
another.79 further away from potentially being available options for use in
Despite these overall encouraging results, long term un- children. Ridinilazole, a novel narrow spectrum antimicrobial;90
anticipated consequences of FMT in children remain un- ibezapolstat, a DNA polymerase inhibitor;91 and MGB-BP-3, a
known. Changes in the microbiome have been implicated to novel topoisomerase inhibitor, are other C. difficile therapeutics
contribute to autoimmune, metabolic, and psychiatric dis- advancing through later stages of clinical development.92
eases, hence any treatment that involves alterations in the All currently available C. difficile therapies have limita-
microbiome at such an early age warrants further long- tions to use (Table 3). Even established therapies such as
term investigation.80–82 The FDA has also issued several vancomycin and metronidazole can increase the risk of
safety alerts regarding potential transmission of multidrug recurrence, disrupt the gut microbiota, induce resistance,
resistant organisms, severe acute respiratory syndrome co- and promote proliferation of multidrug resistant bacteria,
ronavirus 2 virus and Mpox virus by FMT, highlighting the while none of the upcoming treatments are currently be-
risk of transmission of emerging transmissible pathogens ing studied in children as of yet.93–99 Characteristics of
posed by FMT.82–85 Published societal guidelines for chil- an ideal C. difficile therapy include a safe and effective
dren help to address screening and treatment but chal-
agent that can be administered directly to the gut with
lenges around standardization and preparation of stool
minimal influence on local microbial composition or sys-
persist.86 FMT is considered investigational, hence regula-
temic effect and provision of enduring protection. Promis-
tions around administration apply.87
ing therapies that can provide direct neutralizing activity
Upcoming Treatments using amoeba, yeast, or bacteria vehicles are under very
early stages of exploration.
Rebyota (RBX2660) is an FMT product administered rectally
as a single dose of commercially prepared stool from healthy
screened donors. Higher treatment success with absence of PREVENTION
diarrhea within 8 weeks was shown by clinical trial when Prevention of CDI is an important goal to protect chil-
compared with placebo, leading to FDA approval for adults dren and limit healthcare costs. In the absence of a safe
in December 2022.88 Following closely behind in development and effective vaccine, prevention strategies rely on a 2-
is SER-109, an oral microbiome biotherapeutic composed of pronged approach of prevention of spread of C. difficile

8 SHIRLEY et al
Downloaded from http://publications.aap.org/pediatrics/article-pdf/doi/10.1542/peds.2023-062307/1523499/peds.2023-062307.pdf
by guest
TABLE 4 Key Points of C. difficile Infection in Children
Key Points
 Clostridioides difficile infection (CDI) is an important cause of antibiotic-associated diarrhea and healthcare-associated infection in children.
 A multistep approach is currently recommended for diagnosis
 Newer treatment options for CDI in children include oral vancomycin and fidaxomicin
 Recurrence of CDI is common and presents a therapeutic challenge; fecal microbiome transplantation, an exploratory therapy to restore distorted
microbial communities, is becoming more commonly used in children
 More child focused investigation is warranted to improve the burden of CDI in children

spores and prevention of factors that promote progres- Prevention of Disease


sion to disease in those with established colonization. Exposure to antibiotics is the most important modifiable
risk factor for development of CDI. Antibiotic stewardship
Prevention of Transmission programs that target reduction of inappropriate use in
adults have proven helpful in reducing CDI rates, though
In healthcare settings, attention to infection control practi-
comparable data in children are needed.105 Stewardship
ces and facility cleaning may help to limit the spread of
efforts are also needed in the outpatient setting, with at
C. difficile. As alcohol-based hand sanitizers do not inactivate
least 30% of outpatient antibiotic prescriptions esti-
spores, exercising meticulous hand hygiene with soap and
mated to be unnecessary.7 Probiotics have been used as
water after caring for a patient with CDI is recommended.11
a preventative measure for C. difficile in an attempt to
Contact isolation includes the donning of gloves and gowns
restore some of the microbial disruption caused by anti-
by healthcare workers, with dedicated use of private patient
biotics, the benefit of which is still debated. A large sys-
rooms when available. Gloves have been shown to reduce
tematic review reported the risk of CDI was lowered
the risk of hand contamination when caring for patients 60% with probiotics. Conversely, in a recent trial of hos-
with CDI.100 Computer-simulated modeling suggests screen- pitalized children who received antibiotics randomized
ing asymptomatic children on admission may decrease hos- to receive Lactobacillus reuteri (2 × 108 CFU) or placebo
pital onset CDI by 28.5%, but incorporating universal twice daily for the duration of antibiotic treatment, no
screening as part of a multi-intervention bundle could prove difference in prevention of antibiotic associated diar-
logistically prohibitive for some institutions given the high rhea was found.106 The wide heterogeneity in formula-
rates of colonization in some pediatric populations coupled tions, dose, and optimal timing casts uncertainty on the
with pediatric bed shortages.101 generalizability of probiotics for CDI prevention. The
As spores remain hardy in the environment surviving 2017 IDSA and SHEA guidelines state there is insuffi-
months or longer, environmental cleaning with approved cient evidence at this time to recommend use for pri-
sporicidal disinfectants is advised, as staying in a hospital mary prevention of CDI11 Probiotics are generally well
room previously occupied by a patient with CDI increases tolerated, though caution with use in severely immuno-
risk of the current occupant.102 Ultraviolet disinfection has compromised or debilitated patients is needed and the
become a widely used and promising adjunctive measure.103 cost to families must also be factored if used.107
The American Academy of Pediatrics Committee on Infec- Other novel preventative approaches aimed at pro-
tious diseases advises that children with CDI be excluded moting colonization resistance by binding concomitantly
from child care settings until stools are contained within the administered antibiotics, such as activated charcoal and
diaper or the child is continent and stool frequency is no the oral b-lactamase ribaxamase, are under explora-
more than 2 stools above normal frequency.104 tion.108,109 Demonstration of protection against disease

TABLE 5 Research priorities for improving outcomes in children with C. difficile infection
Diagnosis Development of diagnostic biomarkers to improve the performance of currently available testing strategies in order to more
accurately differentiate infection from colonization
Therapy Development of C. difficile targeted therapies that:
 can be delivered locally to the gut and with limited systemic effects
 have narrow spectrum activity to limit intestinal dysbiosis
 maintain long lasting effect to decrease recurrence and
 are also effective for treatment of severe disease
Prevention Development of safe and effective C. difficile vaccines or other preventative measures that promote resistance to C. difficile
colonization

PEDIATRICS Volume 152, number 3, September 2023 9


Downloaded from http://publications.aap.org/pediatrics/article-pdf/doi/10.1542/peds.2023-062307/1523499/peds.2023-062307.pdf
by guest
by antibodies to toxin supports ongoing efforts to de- 9. Mehrotra P, Jang J, Gidengil C, Sandora TJ. Attributable cost of
velop a vaccine, but both toxin based and nontoxin Clostridium difficile infection in pediatric patients. Infect Control
based candidates are much further away from use in Hosp Epidemiol. 2017;38(12):1472–1477
children.110 10. Lurienne L, Bandinelli PA, Galvain T, Coursel CA, Oneto C, Feuerstadt
P. Perception of quality of life in people experiencing or having ex-
CONCLUSIONS perienced a Clostridioides difficile infection: a US population sur-
vey. J Patient Rep Outcomes. 2020;4(1):14
Recent progress made in our understanding of the epidemi-
ology, risk, and management of CDI in children allows us to 11. McDonald LC, Gerding DN, Johnson S, et al. Clinical practice
improve upon pediatric considerations in the diagnosis and guidelines for Clostridium difficile infection in adults and chil-
treatment of this common infection (Table 4). Much of our dren: 2017 update by the Infectious Diseases Society of America
(IDSA) and Society for Healthcare Epidemiology of America
current practice is still adapted from generalizing findings
(SHEA). Clin Infect Dis. 2018;66(7):e1–e48
from adult studies, however significant knowledge gaps re-
main. Prioritizing translational research needs (Table 5) will 12. Centers for Disease Control and Prevention. Healthcare-Associ-
allow us to make further advances to improve the outcomes ated Infections - Community Interface (HAIC) Clostridioides diffi-
of children affected by CDI. cile infections. Available at: https://www.cdc.gov/hai/eip/haicviz.
html. Accessed December 8, 2022
13. Pant C, Deshpande A, Gilroy R, Olyaee M, Donskey CJ. Rising inci-
dence of Clostridium difficile related discharges among hospital-
ABBREVIATIONS
ized children in the United States. Infect Control Hosp Epidemiol.
CDC: Centers for Disease Control and Prevention 2016;37(1):104–106
CDI: Clostridioides difficile infection
14. Centers for Disease Control and Prevention . COVID-19: U.S. impact
FDA: Food and Drug Administration
on antimicrobial resistance, special report 2022. Available at:
FMT: fecal microbiota transplantation https://www.cdc.gov/drugresistance/pdf/covid19-impact-report-508.
NAAT: nucleic acid amplification pdf. Accessed May 23, 2023
rCDI: recurrent Clostridioides difficile infection
15. Lees EA, Carrol ED, Ellaby NAF, et al. Characterization of circulating
Clostridium difficile strains, host response and intestinal micro-
biome in hospitalized children with diarrhea. Pediatr Infect Dis J.
REFERENCES 2020;39(3):221–228
1. Hall IC, O’Toole E. Intestinal flora in new-born infants: with a de- 16. Shuai H, Bian Q, Luo Y, et al. Molecular characteristics of Clostrid-
scription of a new pathogenic anaerobe, bacillus difficilis. Am J
ium difficile in children with acute gastroenteritis from Zhejiang.
Dis Child. 1935;49(2):390–402
BMC Infect Dis. 2020;20(1):343
2. Guh AY, Mu Y, Winston LG, et al; Emerging Infections Program Clos-
17. Perumalsamy S, Lim SC, Riley TV. Clostridioides (Clostridium) diffi-
tridioides difficile Infection Working Group. Trends in U.S. burden
cile isolated from paediatric patients in Western Australia 2019-
of Clostridioides difficile infection and outcomes. N Engl J Med.
2020. Pathology. 2022;54(4):460–465
2020;382(14):1320–1330
18. Al-Rawahi GN, Al-Najjar A, McDonald R, et al. Pediatric oncology
3. Centers for Disease Control and Prevention. Antibiotic resistance
and stem cell transplant patients with healthcare-associated Clos-
threats in the United States, 2019. Available at: www.cdc.gov/
DrugResistance/Biggest-Threats.html. Accessed April 7, 2023 tridium difficile infection were already colonized on admission.
Pediatr Blood Cancer. 2019;66(5):e27604
4. Miranda-Katz M, Parmar D, Dang R, Alabaster A, Greenhow TL. Epi-
demiology and risk factors for community associated Clostri- 19. Tougas SR, Lodha N, Vandermeer B, et al. Prevalence of detection of
dioides difficile in children. J Pediatr. 2020;221:99–106 Clostridioides difficile among asymptomatic children: a systematic
review and meta-analysis. JAMA Pediatr. 2021;175(10):e212328
5. Lessa FC, Mu Y, Bamberg WM, et al. Burden of Clostridium difficile
infection in the United States. N Engl J Med. 2015;372(9):825–834 20. Pichler K, Bausenhardt B, Huhulescu S, et al. Impact and time
course of Clostridium difficile colonization in very low birth
6. Zilberberg MD, Tillotson GS, McDonald C. Clostridium difficile in-
weight infants. Neonatology. 2018;114(2):149–154
fections among hospitalized children, United States, 1997-2006.
Emerg Infect Dis. 2010;16(4):604–609 21. Ferraris L, Couturier J, Eckert C, et al. Carriage and colonization
of C. difficile in preterm neonates: a longitudinal prospective
7. Edwards PT, Thurm CW, Hall M, et al. Clostridioides difficile infec-
study. PLoS One. 2019;14(2):e0212568
tion in hospitalized pediatric patients: comparisons of epidemiol-
ogy, testing, and treatment from 2013 to 2019. J Pediatr. 22. Brennhofer SA, Rogawski McQuade ET, Liu J, Guerrant RL, Platts-
2023;252:111–116.e1 Mills JA, Warren CA. Clostridioides difficile colonization among
very young children in resource-limited settings. Clin Microbiol In-
8. Silva A, Du T, Choi KB, et al; CNISP C. difficile working group. Epide-
miology of primary and recurrent healthcare-associated and com- fect. 2022;28(7):996–1002
munity-associated pediatric Clostridioides difficile infection in 23. Xie X, Xiang B, Wu Y, Zhao Y, Wang Q, Jiang X. Infant progressive
Canada, 2015-2020. J Pediatric Infect Dis Soc. 2023;12(4):222–225 colonic stenosis caused by antibiotic-related Clostridium difficile

10 SHIRLEY et al
Downloaded from http://publications.aap.org/pediatrics/article-pdf/doi/10.1542/peds.2023-062307/1523499/peds.2023-062307.pdf
by guest
colitis - a case report and literature review. BMC Pediatr. 2018; 40. Wilcox MH, Mooney L, Bendall R, Settle CD, Fawley WN. A case-control
18(1):320 study of community-associated Clostridium difficile infection.
24. Eglow R, Pothoulakis C, Itzkowitz S, et al. Diminished Clostridium J Antimicrob Chemother. 2008;62(2):388–396
difficile toxin A sensitivity in newborn rabbit ileum is associated 41. Rabold D, Espelage W, Abu Sin M, et al. The zoonotic potential of
with decreased toxin A receptor. J Clin Invest. 1992;90(3):822–829 Clostridium difficile from small companion animals and their
25. Kociolek LK, Espinosa RO, Gerding DN, et al. Natural Clostridioides owners. PLoS One. 2018;13(2):e0193411
difficile toxin immunization in colonized infants. Clin Infect Dis. 42. Kociolek LK, Patel SJ, Shulman ST, Gerding DN. Molecular epidemiology
2020;70(10):2095–2102 of Clostridium difficile infections in children: a retrospective cohort
26. Hourigan SK, Sears CL, Oliva-Hemker M. Clostridium difficile infec- study. Infect Control Hosp Epidemiol. 2015;36(4):445–451
tion in pediatric inflammatory bowel disease. Inflamm Bowel Dis. 43. Perumalsamy S, Riley TV. Molecular epidemiology of Clostridioides
2016;22(4):1020–1025 difficile infections in children. J Pediatric Infect Dis Soc. 2021;
27. Theunissen C, Knoop C, Nonhoff C, et al. Clostridium difficile colitis 10(Supplement_3):S34–S40
in cystic fibrosis patients with and without lung transplantation. 44. Loftus KV, Wilson PM. A curiously rare case of septic shock from
Transpl Infect Dis. 2008;10(4):240–244 Clostridium difficile colitis. Pediatr Emerg Care. 2017;33(8):576–578
28. Dominguez SR, Dolan SA, West K, et al. High colonization rate and 45. Wendt JM, Cohen JA, Mu Y, et al. Clostridium difficile infection
prolonged shedding of Clostridium difficile in pediatric oncology among children across diverse US geographic locations. Pediat-
patients. Clin Infect Dis. 2014;59(3):401–403 rics. 2014;133(4):651–658
29. Ochfeld E, Balmert LC, Patel SJ, Muller WJ, Kociolek LK. Risk factors 46. Nicholson MR, Crews JD, Starke JR, Jiang ZD, DuPont H, Edwards
for Clostridioides (Clostridium) difficile infection following solid K. Recurrent Clostridium difficile infection in children: patient risk
organ transplantation in children. Transpl Infect Dis. 2019;21(5):e13149 factors and markers of intestinal inflammation. Pediatr Infect Dis
30. Duclaux-Loras R, Berthiller J, Ferroni A, et al. Clostridium difficile: J. 2017;36(4):379–383
a frequent infection in children after intestinal transplantation. 47. Orenstein R, Patron RL, Seville MT. Why does Clostridium difficile
Transplantation. 2020;104(1):197–200 infection recur? J Am Osteopath Assoc. 2019;119(5):322–326
31. Leibowitz J, Soma V, Rosen L, Ginocchio C, Rubin L. Similar pro- 48. Chang JY, Antonopoulos DA, Kalra A, et al. Decreased diversity of
portions of stool specimens from hospitalized children with and the fecal microbiome in recurrent Clostridium difficile-associated
without diarrhea test positive for Clostridium difficile. Pediatr In- diarrhea. J Infect Dis. 2008;197(3):435–438
fect Dis J. 2014;34(3):261–266
49. Schwab EM, Wilkes J, Korgenski K, Hersh AL, Pavia AT, Stevens VW.
32. Anjewierden S, Han Z, Foster CB, Pant C, Deshpande A. Risk fac- Risk factors for recurrent Clostridium difficile infection in pediat-
tors for Clostridium difficile infection in pediatric inpatients: a ric inpatients. Hosp Pediatr. 2016;6(6):339–344
meta-analysis and systematic review. Infect Control Hosp Epide-
50. Ruan W, Kellermayer R. Alternative diagnoses in pediatric fecal
miol. 2019;40(4):420–426
microbiota transplant referral patients. J Pediatr Gastroenterol
33. Adams DJ, Eberly MD, Rajnik M, Nylund CM. Risk factors for com- Nutr. 2021;72(5):693–696
munity-associated Clostridium difficile infection in children. J Pe-
51. Sandora TJ, Williams DN, Daugherty K, et al. Stool toxin concentra-
diatr. 2017;186:105–109
tion does not distinguish Clostridioides difficile infection from col-
34. Weng MK, Adkins SH, Bamberg W, et al. Risk factors for commu- onization in children less than 3 years of age. J Pediatric Infect
nity-associated Clostridioides difficile infection in young children. Dis Soc. 2022;11(10):454–458
Epidemiol Infect. 2019;147:e172
52. Parnell JM, Fazili I, Bloch SC, et al. Two-step testing for Clostridioides
35. Ramirez J, Guarner F, Bustos Fernandez L, Maruy A, Sdepanian VL, difficile is inadequate in differentiating infection from colonization
Cohen H. Antibiotics as major disruptors of gut microbiota. Front in children. J Pediatr Gastroenterol Nutr. 2021;72(3):378–383
Cell Infect Microbiol. 2020;10:572912
53. Pahud BA, Hassan F, Harrison CJ, et al. Detection of Clostridioides
36. Smith AB, Jenior ML, Keenan O, et al. Enterococci enhance Clostri- difficile by real-time PCR in young children does not predict dis-
dioides difficile pathogenesis. Nature. 2022;611(7937):780–786 ease. Hosp Pediatr. 2020;10(7):555–562
37. Dang R, Alabaster A, Miranda-Katz M, Parmar D, Greenhow TL. 54. Sethi AK, Al-Nassir WN, Nerandzic MM, Bobulsky GS, Donskey CJ.
Impact of antecedent antibiotic usage on community-associated Persistence of skin contamination and environmental shedding of
Clostridioides difficile infection in Pediatrics. Pediatr Infect Dis J. Clostridium difficile during and after treatment of C. difficile in-
2021;40(5):426–428 fection. Infect Control Hosp Epidemiol. 2010;31(1):21–27
38. Kociolek LK, Gerding DN, Espinosa RO, Patel SJ, Shulman ST, Ozer 55. Kelly CR, Fischer M, Allegretti JR, et al. ACG clinical guidelines:
EA. Clostridium difficile Whole Genome Sequencing Reveals Lim- prevention, diagnosis, and treatment of Clostridioides difficile in-
ited Transmission Among Symptomatic Children: A Single-Center fections. Am J Gastroenterol. 2021;116(6):1124–1147
Analysis. Clin Infect Dis. 2018;67(2):229–234 56. Coffin SE, Abanyie F, Bryant K, et al; CDC Prevention Epicenters
39. Chitnis AS, Holzbauer SM, Belflower RM, et al. Epidemiology of Program. Pediatric research priorities in healthcare-associated
community-associated Clostridium difficile infection, 2009 through infections and antimicrobial stewardship. Infect Control Hosp Epi-
2011. JAMA Intern Med. 2013;173(14):1359–1367 demiol. 2021;42(5):519–522

PEDIATRICS Volume 152, number 3, September 2023 11


Downloaded from http://publications.aap.org/pediatrics/article-pdf/doi/10.1542/peds.2023-062307/1523499/peds.2023-062307.pdf
by guest
57. Kociolek LK, Kutty PK, Polgreen PM, Beekmann SE. Healthcare 71. Kociolek LK. Moving the needle on Clostridioides difficile research
provider diagnostic testing practices for identification of Clos- in children: a major step forward but challenges remain. Clin In-
tridioides (Clostridium) difficile in children: an Emerging fect Dis. 2020;71(10):2589–2591
Infections Network survey. Infect Control Hosp Epidemiol. 72. Neal MD, Alverdy JC, Hall DE, Simmons RL, Zuckerbraun BS. Divert-
2019;40(3):276–280 ing loop ileostomy and colonic lavage: an alternative to total ab-
58. Tran NN, Mills JP, Zimmerman C, et al. Incorporating preauthoriza- dominal colectomy for the treatment of severe, complicated
tion into antimicrobial stewardship pharmacist workflow reduces Clostridium difficile associated disease. Ann Surg. 2011;254(3):
Clostridioides difficile and gastrointestinal panel testing. Infect 423–427, discussion 427–429
Control Hosp Epidemiol. 2020;41(10):1136–1141 73. Fashandi AZ, Martin AN, Wang PT, et al. An institutional compari-
59. Cotter JM, Thomas J, Birkholz M, et al. Impact of multiplex testing son of total abdominal colectomy and diverting loop ileostomy
on the identification of pediatric Clostridiodes difficile. J Pediatr. and colonic lavage in the treatment of severe, complicated Clos-
2020;218:157–165.e3 tridium difficile infections. Am J Surg. 2017;213(3):507–511
60. Zhou P, Zhou N, Shao L, et al. Diagnosis of Clostridium difficile in- 74. Fitzpatrick F, Safdar N, van Prehn J, Tschudin-Sutter S. How can pa-
fection using an UPLC-MS based metabolomics method. Metabolo- tients with Clostridioides difficile infection on concomitant antibiotic
mics. 2018;14(8):102 treatment be best managed? Lancet Infect Dis. 2022;22(11):e336–e340
61. Galvez JAV, Pollock NR, Alonso CD, et al. Stool interleukin-1b dif- 75. Kaki R, Brooks A, Main C, Jayaratne P, Mertz D. Does extending
ferentiates Clostridioides difficile infection from asymptomatic Clostridium difficile treatment in patients who are receiving con-
carriage and non-C.difficile infection diarrhea. Clin Infect Dis. comitant antibiotics reduce the rate of relapse? The Internet
2023;76(3):e1467–e1475 Journal of Infectious Diseases. 2016;15(1)
62. Patel M, Fowler D, Sizer J, Walton C. Faecal volatile biomarkers of 76. Bao H, Lighter J, Dubrovskaya Y, et al. Oral vancomycin as second-
Clostridium difficile infection. PLoS One. 2019;14(4):e0215256 ary prophylaxis for Clostridioides difficile infection. Pediatrics.
63. Ofosu A. Clostridium difficile infection: a review of current and 2021;148(2):e2020031807
emerging therapies. Ann Gastroenterol. 2016;29(2):147–154 77. Shin JH, Chaplin AS, Hays RA, et al. Outcomes of a multidisciplin-
64. Johnson S, Lavergne V, Skinner AM, et al. Clinical practice ary clinic in evaluating recurrent Clostridioides difficile infection
guideline by the Infectious Diseases Society of America (IDSA) patients for fecal microbiota transplant: a retrospective cohort
and Society for Healthcare Epidemiology of America (SHEA): analysis. J Clin Med. 2019;8(7):1036
2021 focused update guidelines on management of Clostri- 78. Gerding DN, Kelly CP, Rahav G, et al. Bezlotoxumab for prevention
dioides difficile infection in adults. Clin Infect Dis. 2021;73(5): of recurrent Clostridium difficile infection in patients at increased
e1029–e1044 risk for recurrence. Clin Infect Dis. 2018;67(5):649–656
65. Louie TJ, Miller MA, Mullane KM, et al; OPT-80-003 Clinical Study 79. Nicholson MR, Mitchell PD, Alexander E, et al. Efficacy of fecal mi-
Group. Fidaxomicin versus vancomycin for Clostridium difficile in- crobiota transplantation for Clostridium difficile infection in chil-
fection. N Engl J Med. 2011;364(5):422–431 dren. Clin Gastroenterol Hepatol. 2020;18(3):612–619.e1
66. Guery B, Menichetti F, Anttila VJ, et al; EXTEND Clinical Study 80. De Palma G, Lynch MD, Lu J, et al. Transplantation of fecal microbiota
Group. Extended-pulsed fidaxomicin versus vancomycin for Clos- from patients with irritable bowel syndrome alters gut function and
tridium difficile infection in patients 60 years and older (EXTEND): behavior in recipient mice. Sci Transl Med. 2017;9(379):eaaf6397
a randomised, controlled, open-label, phase 3b/4 trial. Lancet In- 81. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gor-
fect Dis. 2018;18(3):296–307 don JI. An obesity-associated gut microbiome with increased ca-
67. Mikamo H, Tateda K, Yanagihara K, et al. Efficacy and safety of fi- pacity for energy harvest. Nature. 2006;444(7122):1027–1031
daxomicin for the treatment of Clostridioides (Clostridium) diffi- 82. Bibiloni R, Mangold M, Madsen KL, Fedorak RN, Tannock GW. The
cile infection in a randomized, double-blind, comparative phase III bacteriology of biopsies differs between newly diagnosed, un-
study in Japan. J Infect Chemother. 2018;24(9):744–752 treated, Crohn’s disease and ulcerative colitis patients. J Med Mi-
68. Stultz JS, Hopp J, Orndahl CM, Omecene NE, Pakyz AL. Changes in crobiol. 2006;55(Pt 8):1141–1149
metronidazole and vancomycin utilization for nonsevere Clostri- 83. US Food and Drug Administration. Safety alert regarding use of
dioides difficile infection among institutions caring for children. fecal microbiota for transplantation and additional safety protec-
Pediatr Infect Dis J. 2021;40(7):634–636 tions pertaining to monkeypox virus. Available at: https://www.
69. Yin J, Kociolek LK, Same RG, Hsu AJ, Amoah J, Tamma PD. Oral fda.gov/vaccines-blood-biologics/safety-availability-biologics/
vancomycin may be associated with earlier symptom resolution safety-alert-regarding-use-fecal-microbiota-transplantation-and-ad-
than metronidazole for hospitalized children with nonsevere Clos- ditional-safety-protections-0#::text=The%20Food%20and%20Drug
tridiodes difficile infections. Open Forum Infect Dis. 2019;6(12): %20Administration,additional%20safety%20protections%20are%20
ofz492 needed. Accessed August 22, 2022
70. Wolf J, Kalocsai K, Fortuny C, et al. Safety and efficacy of fidaxomi- 84. US Food and Drug Administration Safety Communication. Impor-
cin and vancomycin in children and adolescents with Clostri- tant safety alert regarding use of fecal microbiota for transplan-
dioides (Clostridium) difficile infection: a phase 3, multicenter, tation and risk of serious adverse reactions due to transmission
randomized, single-blind clinical trial (SUNSHINE). Clin Infect Dis. of multi-drug resistant organisms. Available at: https://www.fda.
2020;71(10):2581–2588 gov/vaccines-blood-biologics/safety-availability-biologics/important-

12 SHIRLEY et al
Downloaded from http://publications.aap.org/pediatrics/article-pdf/doi/10.1542/peds.2023-062307/1523499/peds.2023-062307.pdf
by guest
safety-alert-regarding-use-fecal-microbiota-transplantation-and-risk-- 98. Baines SD, O’Connor R, Freeman J, et al. Emergence of reduced
serious-adverse. Accessed June 13, 2019 susceptibility to metronidazole in Clostridium difficile. J Antimi-
85. US Food and Drug Administration. Fecal microbiota for transplantation: crob Chemother. 2008;62(5):1046–1052
safety alert - regarding additional safety protections pertaining to 99. Martin H, Willey B, Low DE, et al. Characterization of Clostridium
SARS-CoV-2 and COVID-19. Available at: https://www.fda.gov/safety/ difficile strains isolated from patients in Ontario, Canada, from
medical-product-safety-information/fecal-microbiota-transplantation- 2004 to 2006. J Clin Microbiol. 2008;46(9):2999–3004
safety-alert-regarding-additional-safety-protections-pertaining. Accessed 100. Landelle C, Verachten M, Legrand P, Girou E, Barbut F, Brun-Buis-
March 23, 2020
son C. Contamination of healthcare workers’ hands with Clostrid-
86. Davidovics ZH, Michail S, Nicholson MR, et al; FMT Special Interest ium difficile spores after caring for patients with C. difficile
Group of the North American Society of Pediatric Gastroenterology infection. Infect Control Hosp Epidemiol. 2014;35(1):10–15
Hepatology, Nutrition, the European Society for Pediatric Gastroen-
101. Barker AK, Scaria E, Alagoz O, Sethi AK, Safdar N. Reducing C. difficile
terology Hepatology, Nutrition. Fecal microbiota transplantation for
recurrent Clostridium difficile infection and other conditions in chil- in children: an agent-based modeling approach to evaluate interven-
dren: a joint position paper from the North American Society for tion effectiveness. Infect Control Hosp Epidemiol. 2020;41(5):522–530
Pediatric Gastroenterology, Hepatology, and Nutrition and the Euro- 102. Sood G, Truelove S, Dougherty G, et al. Clostridioides difficile in-
pean Society for Pediatric Gastroenterology, Hepatology, and Nutri- fection (CDI) in a previous room occupant predicts CDI in subse-
tion. J Pediatr Gastroenterol Nutr. 2019;68(1):130–143 quent room occupants across different hospital settings. Am J
87. Food and Drug Administration. Enforcement policy regarding in- Infect Control. 2022;50(12):1352–1354
vestigational new drug requirements for use of fecal microbiota 103. Steele M, Hurtado RR, Rychlik K, et al. Impact of an automated
for transplantation to treat Clostridioides difficile infection not re- multiple emitter whole-room ultraviolet-C disinfection system on
sponsive to standard therapies. Available at: https://www.fda.gov/ hospital acquired infections: a quasi-experimental study. Am J In-
media/86440/download. Accessed November 15, 2022 fect Control. 2021;49(9):1200–1203
88. Khanna S, Assi M, Lee C, et al. Efficacy and Safety of RBX2660 in 104. Kimberlin DW, Barnett ED, Lynfield R, Sawyer MH; Committee on
PUNCH CD3, a Phase III, Randomized, Double-Blind, Placebo-Controlled
Infectious Diseases, American Academy of Pediatrics. Red Book:
Trial with a Bayesian Primary Analysis for the Prevention of Recurrent
2021–2024 Report of the Committee on Infectious Diseases.
Clostridioides difficile Infection. Drugs. 2022;82(15):1527–1538
Itasca, IL: American Academy of Pediatrics; 2021
89. Feuerstadt P, Louie TJ, Lashner B, et al. SER-109, an oral micro-
105. Shea KM, Hobbs ALV, Jaso TC, et al. Effect of a health care system
biome therapy for recurrent Clostridioides difficile infection. N
Engl J Med. 2022;386(3):220–229 respiratory fluoroquinolone restriction program to alter utiliza-
tion and impact rates of Clostridium difficile infection. Antimicrob
90. Carlson TJ, Endres BT, Basseres E, Gonzales-Luna AJ, Garey KW. Agents Chemother. 2017;61(6):e00125–e00117
Ridinilazole for the treatment of Clostridioides difficile infection.
Expert Opin Investig Drugs. 2019;28(4):303–310 106. Kołodziej M, Szajewska H. Lactobacillus reuteri DSM 17938 in the
prevention of antibiotic-associated diarrhoea in children: a ran-
91. Garey KW, McPherson J, Dinh AQ, et al. Efficacy, safety, pharmaco-
domized clinical trial. Clin Microbiol Infect. 2019;25(6):699–704
kinetics, and microbiome changes of ibezapolstat in adults with
Clostridioides difficile infection: a phase 2a multicenter clinical 107. Goldenberg JZ, Yap C, Lytvyn L, et al. Probiotics for the prevention
trial. Clin Infect Dis. 2022;75(7):1164–1170 of Clostridium difficile-associated diarrhea in adults and children.
Cochrane Database Syst Rev. 2017;12(12):CD006095
92. Hind C, Clifford M, Woolley C, et al. Insights into the spectrum of
activity and mechanism of action of MGB-BP-3. ACS Infect Dis. 108. Vehreschild MJGT, Ducher A, Louie T, et al. An open randomized multi-
2022;8(12):2552–2563 centre phase 2 trial to assess the safety of DAV132 and its efficacy
to protect gut microbiota diversity in hospitalized patients treated
93. Dzunkova M, D’Auria G, Xu H, et al. The monoclonal antitoxin anti-
with fluoroquinolones. J Antimicrob Chemother. 2022;77(4):1155–1165
bodies (Actoxumab-Bezlotoxumab) treatment facilitates normaliza-
tion of the gut microbiota of mice with Clostridium difficile 109. Kokai-Kun JF, Le C, Trout K, et al. Ribaxamase, an orally adminis-
infection. Front Cell Infect Microbiol. 2016;6:119 tered b-Lactamase, diminishes changes to acquired antimicrobial
resistance of the gut resistome in patients treated with ceftriax-
94. Kelly CP, LaMont JT. Clostridium difficile–more difficult than ever.
one. Infect Drug Resist. 2020;13:2521–2535
N Engl J Med. 2008;359(18):1932–1940
95. O’Horo JC, Jindai K, Kunzer B, Safdar N. Treatment of recurrent Clostrid- 110. Riley TV, Lyras D, Douce GR. Status of vaccine research and devel-
ium difficile infection: a systematic review. Infection. 2014;42(1):43–59 opment for Clostridium difficile. Vaccine. 2019;37(50):7300–7306

96. Bossu G, Di Sario R, Argentiero A, Esposito S. Antimicrobial pro- 111. Luna RA, Boyanton BL Jr, Mehta S, et al. Rapid stool-based diag-
phylaxis and modifications of the gut microbiota in children with nosis of Clostridium difficile infection by real-time PCR in a child-
cancer. Antibiotics (Basel). 2021;10(2):152 ren’s hospital. J Clin Microbiol. 2011;49(3):851–857
97. Huang H, Weintraub A, Fang H, Nord CE. Antimicrobial resistance in 112. DIFICID. (Fidaxomicin) [package insert]. Whitehouse Station, NJ:
Clostridium difficile. Int J Antimicrob Agents. 2009;34(6):516–522 Merck; 2020

PEDIATRICS Volume 152, number 3, September 2023 13


Downloaded from http://publications.aap.org/pediatrics/article-pdf/doi/10.1542/peds.2023-062307/1523499/peds.2023-062307.pdf
by guest

You might also like