Clostridioides (formerly Clostridium) difficile is the most important infectious 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 associated with high morbidity and even fatality. Diagnosis of CDI can be challenging as currently available tests detect either the presence of organism or disease-causing toxin but cannot distinguish colonization from infection. Since colonization can be high in specific pediatric groups, such as infants and young children, biomarkers to aid in accurate diagnosis are urgently needed. Similar to disease in adults, recurrence of CDI in children is common, affecting 20% to 30% of incident cases. Metronidazole has long been considered the mainstay therapy for CDI in children. However, new evidence supports the safety and efficacy of oral vancomycin and fidaxomicin as additional treatment options, whereas fecal microbiota transplantation is gaining popularity for recurrent infection. Recent advancements in our understanding of emerging epidemiologic trends and management of CDI unique to children are highlighted in this review. Despite encouraging therapeutic advancements, there remains a pressing need to optimize CDI therapy in children, particularly as it pertains to severe and recurrent disease.

Clostridioides (formerly Clostridium) difficile is a spore-forming, anaerobic, Gram-positive bacillus first described in 1935 from the stool of healthy newborn infants.1  Initially named Bacillus difficile, because it was difficult to isolate, with time this pathogen has also proven difficult to treat and control. Most C. difficile bacteria secrete toxin A and toxin B. The presence of toxin B appears to be the primary cause of diarrhea, and sufficient by itself to induce disease, whereas other virulence factors, such as binary toxin, surface layer protein, and biofilm formation, may contribute to colonization, transmission, or disease severity.

C. difficile has risen to become a leading cause of healthcare-associated infection among adult populations in the United States, resulting in an estimated half-million attributable cases each year, leading to 223 900 people requiring hospital care and 12 800 deaths.2,3  CDI prolongs hospital stay and increases healthcare expenses, costing the US healthcare system at least 1 billion dollars annually.3  The US Centers for Disease Control and Prevention (CDC) have identified C. difficile as an urgent threat, listing it as one of the top 5 drug-resistant pathogens in need of aggressive action.3 

Although CDI is more common and tends to be more severe in older adults, the burden of C. difficile to children is also significant (Fig 1). C. difficile is the most important infectious cause of antibiotic-associated diarrhea in children, contributing approximately 20 000 infections in the United States each year.4,5  Rates of CDI in children have increased over the past 3 decades and the rate of pediatric hospitalizations resulting from or complicated by C. difficile increased 57% from 1997 to 2006.5,6  There is some more recent indication of leveling rates, particularly in the inpatient setting.2,7,8  Changes in testing, diagnosis, treatment and strain type can all contribute to fluctuations in incidence. Nonetheless, CDI-associated hospitalizations cost 1.6 times more than non-CDI associated hospitalizations in children, and CDI adds an additional 4 days to length of stay for pediatric hospitalizations.9  Broader, less well-described indirect consequences of CDI on child health also bear consideration, including educational consequences from lost days of schooling and economic impact from missed parental days of work.10 

FIGURE 1

The impact of Clostridioides difficile infection in children.

FIGURE 1

The impact of Clostridioides difficile infection in children.

Close modal

The Infectious Diseases Society of America (IDSA) and the Society for Healthcare Epidemiology of America (SHEA) updated CDI clinical practice guidelines in 2017, including, for the first time, specific recommendations regarding CDI diagnosis and therapeutic considerations in children.11  Our understanding of CDI in children has continued to grow since the release of these guidelines. This review summarizes recent updates pertaining to the epidemiology, diagnosis, and management of CDI in children, with a principle focus on developments over the past 5 years.

The incidence of CDI as a cause of diarrhea in children has increased over time, though the exact magnitude of this problem is challenging to quantify given limitations in diagnosis.4,12,13  CDI is classified as healthcare-associated (onset more than 3 days after hospitalization) or community-associated (onset within 3 days of hospitalization provided no documented overnight stay in a healthcare facility in the preceding 12 weeks). CDI, considered mostly a healthcare-associated infection in adults, contrasts as most commonly community-associated in children, nonetheless community-associated CDI rates have increased in both adults and children.2,4  Population-based surveillance conducted by the CDC Emerging Infections Program estimates the incidence of community-associated CDI in children was as high as 25.8 per 100 000 in 2019, accounting for 75% of cases, compared with 9.0 per 100 000 reported as healthcare-associated (Fig 1).12  Diversion of public health resources during the coronavirus disease 2019 pandemic has since led to delays in subsequent surveillance estimates.14 

The global impact of CDI in children is even harder to quantify given even wider variations in diagnostic approach.15  Regardless, high rates of CDI in children are reported worldwide. A recent cross-sectional study conducted among children presenting with acute gastroenteritis at a tertiary children’s hospital in Zhejiang, China, for example, reported 14.3% of cases solely attributed to CDI.16  Whereas, C. difficile was detected from 19.7% of children being evaluated for diarrhea in Perth, Australia.16,17 

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.1921  The frequency remains as high as 22% in toddlers 1 to 2 years of age before declining to approach rates seen in healthy adults of 1% to 3%.11  This finding is consistent even among resource-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 incidence varies by country.22 

For reasons unknown, infants appear almost universally protected from clinical disease despite these high colonization rates, even in the presence of toxin producing strains. Only rare cases of CDI in infants are reported,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 the lack of expression of receptors for binding C. difficile toxins, based on limited animal data from the intestines of newborn rabbits, however further corroboration in humans is required to substantiate this observation.24  Although protection from passive transplacental transfer of maternal antibody is also unlikely the explanation based on maternal cord blood sampling, colonization may be important for acquired protection later in life, as natural immunization against toxin A and B does seem to occur following colonization, irrespective of feeding method, though the durability of this protection is unknown.25 

Beyond infancy and toddlerhood, other specific pediatric populations are predisposed to higher rates of colonization, including those with inflammatory bowel disease, cystic fibrosis, and cancer as well as transplant recipients.2630  Up to one-quarter of hospitalized children may be asymptomatically colonized31  and one-third or more of pediatric oncology patients may have asymptomatic C. difficile colonization on admission. In these older age groups, colonization serves as a risk factor for subsequent infection as CDI is more common in children with chronic medical conditions. For example, nearly all clinical isolates from pediatric oncology patients diagnosed with CDI during hospitalization were identical to their baseline admission isolates by pulsed-field gel electrophoresis analysis; the caveat here being that diarrhea in children with cancer can be multifactorial, and it may not always be possible to clinically distinguish the primary etiology.18 

Antibiotic exposure is the most important predisposing factor to the development of CDI.3234  Diarrhea in general is a common side effect of antibiotics, occurring in more than one third of patients who receive them, with mechanisms such as osmotic effect and promotility contributing. Antibiotics may specifically predispose to CDI through disruptions in intestinal microbial composition and metabolism, a state frequently referred to as dysbiosis. Through complex interactions, the distorted microbial gut community can selectively promote survival of C. difficile.35  For example, enterococci are highly abundant in the stools of children with CDI. Enterococci may produce fermentable amino acids, such as leucine and ornithine, whereas depleting arginine, leading to metabolic alterations in the gut that may further promote CDI.36  Any antibiotic class can potentially predispose to CDI and onset can occur within days to weeks of exposure. In particular, third generation cephalosporins, clindamycin, fluoroquinolones, and amoxicillin-clavulanate are strong associations.33,37  Several other factors that predispose to CDI are described, including administration of proton pump inhibitors, tube feeds, healthcare facility exposure, prolonged hospitalization, and contact with a person infected with C. difficile.32,33 

Given that CDI is mostly community-associated in children, recent attention has focused on identifying specific risk factors for this setting. In a large case-control study conducted by the Kaiser-Permanente system, risk factors for community-associated CDI in children included non-Hispanic ethnicity, antibiotic exposure to amoxicillin-clavulanate, cephalosporins or clindamycin within the previous 12 weeks, a previous positive C. difficile test within 6 months, and increased health care visits within the last year.4  However, traditional risk factors may not always be present and 13.6% of children lacked any identifiable risk factor in another multisite case-control study of younger children with community-associated CDI.34 

Transmission of C. difficile occurs fecal-orally through ingestion of spores, including by contact with contaminated surfaces in the environment. Whole genome sequencing indicates that transmission of C. difficile among symptomatic children within healthcare settings may be less common than in adult populations. Only about 12% of healthcare-associated transmission events could be linked to another symptomatic patient at a medical center that implemented contact isolation for all patients with diarrhea.38  Many children with healthcare associated CDI are already colonized with C. difficile at the time of admission.18  Infants, young children, and household contacts with active CDI may serve as possible sources of community-associated C. difficile exposure.18,39,40  Potential transmission from companion animals has also been implicated.41 

Ribotyping, a molecular technique often used for surveillance purposes, characterizes C. difficile strains based on specific differences in ribosomal RNA. C. difficile ribotype 106 has emerged as the most common strain in the United States, superseding the hypervirulent ribotype 027 strain (also known as BI/NAP1), which was previously the most common.12  In 2018, ribotype 106 accounted for about 16% of community-associated and 12% of healthcare-associated infections in the United States.12  Ribotype 106 is also the most common strain in the pediatric population.42  Globally, ribotype 014 (ST2, ST13, ST49/clade 1) appears to be common.43 

C. difficile causes a wide spectrum of disease severity, ranging from asymptomatic carriage all the way to life threatening colitis. Diarrhea is thought to primarily result from toxin-mediated intestinal epithelial cytotoxicity and inflammation. Most symptomatic children will have mild-to-moderate diarrhea. Severe CDI, defined by leukocytosis (≥15, 000 cells/mm3) or renal impairment (serum creatinine >1.5 mg/dL) in adults, is poorly defined in children, necessitating clinical judgement to diagnose. Severe complications of CDI in children include development of pseudomembranous colitis, pneumatosis intestinalis, toxic megacolon, perforation, peritonitis, and shock with multisystem failure.44  Severe CDI is less common in children than adults, but does occur, and up to 8% of will develop severe disease.45  By other estimates, up to 0.1% to 1.2% may require colectomy, and all-cause mortality in children with CDI is about 2% to 4%.7  Imaging and endoscopic findings of CDI are nonspecific. Disease in children is often cecocolonic or colonic. In severe cases, abdominal imaging may show colonic wall thickening, colonic dilation or free air in the case of perforation (Fig 2).

FIGURE 2

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

FIGURE 2

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

Close modal

Recurrent CDI (rCDI) is as common in children as adults, occurring in about 20% to 30% of cases despite treatment.11,46,47  Recurrence is defined as new onset of CDI symptoms with a positive C. difficile specific laboratory test following an incident episode in the previous 2 to 8 weeks. Ongoing dysbiosis perpetuates survival and proliferation of C. difficile, leading to relapse or reinfection.48  Risk factors for recurrence include the presence of comorbidities such as cancer, IBD, medical technology dependence, recent surgery, and antibiotic exposure.49  The risk of additional recurrent episodes increase with each recurrence. Interestingly, nearly one-fourth of children referred for fecal microbiota transplantation (FMT) because of rCDI at one institution were found to have an alternative diagnosis, such as constipation or overflow diarrhea or inflammatory bowel disease, emphasizing the need for careful consideration of alternative causes of recurrent loose stools when faced with this dilemma.50 

The diagnosis of CDI in children is challenging, and detection does not always equate to diagnosis. There are several commercial tests available for diagnosis that either detect the presence of C. difficile organism or toxin production, but each has limitations in application and there is no gold standard (Table 1). Tests that detect the organism, such as nucleic acid amplification tests (NAATs), are highly sensitive but do not distinguish colonization from infection. Tests that detect toxin, such as enzyme immunoassays, are more specific but lack sufficient sensitivity for diagnosis. Detection of toxin does not correlate with severity of symptoms in children nor can stool toxin concentration reliably distinguish carriage from infection.51  A prospective study of asymptomatic children with cancer, cystic fibrosis, or inflammatory bowel disease in which 21% were colonized with C. difficile demonstrated that toxin detection did not differ when compared with a cohort of children with symptomatic CDI.52  Hence, correct interpretation of C. difficile test results ultimately requires careful consideration of patient factors. Consequences of misinterpretation include unnecessary antibiotic exposure, potential drug adverse effects, and delay in diagnosis of the true cause of diarrhea.

TABLE 1

Summary of Clostridioides difficile Testinga

TestTargetStrengthsLimitations
Tests to detect presence of C. difficile organism 
GDH EIA Detects C. difficile-specific enzyme • High sensitivity;
• rapid, takes hours to complete;
• inexpensive 
• Low specificitya;
• does not distinguish toxigenic from nontoxigenic strains 
NAATs Detects genes for C. difficile toxin (toxin A/B) • High sensitivity;
• good specificity;
• can be rapid 
• Lower positive predictive value;
• does not detect presence of toxin 
Toxigenic culture Detects toxin producing C. difficile strains • High sensitivity;
• high specificity;
• allows for antimicrobial susceptibility testing 
• Technically complex to perform;
• takes days to complete;
• used mostly as reference for 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;
• rapid, takes hours to complete;
• inexpensive 
• Low sensitivity (variable but as low as 35%)111  
CCNA Demonstrates free toxin B • High sensitivity;
• high specificity 
• Technically complex to perform;
• takes day to complete;
• used mostly as reference for validation or in research settings 
TestTargetStrengthsLimitations
Tests to detect presence of C. difficile organism 
GDH EIA Detects C. difficile-specific enzyme • High sensitivity;
• rapid, takes hours to complete;
• inexpensive 
• Low specificitya;
• does not distinguish toxigenic from nontoxigenic strains 
NAATs Detects genes for C. difficile toxin (toxin A/B) • High sensitivity;
• good specificity;
• can be rapid 
• Lower positive predictive value;
• does not detect presence of toxin 
Toxigenic culture Detects toxin producing C. difficile strains • High sensitivity;
• high specificity;
• allows for antimicrobial susceptibility testing 
• Technically complex to perform;
• takes days to complete;
• used mostly as reference for 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;
• rapid, takes hours to complete;
• inexpensive 
• Low sensitivity (variable but as low as 35%)111  
CCNA Demonstrates free toxin B • High sensitivity;
• high specificity 
• Technically complex to perform;
• 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 diagnostic approach.11 

CCNA, cell culture cytotoxicity neutralization assay; EIA, enzyme immunoassay; GDH, Glutamate dehydrogenase; NAATs, nucleic acid amplification tests.

Adherence to several strategies of diagnostic stewardship can further improve the predictive value of testing. First, diarrheal stool should preferentially only be collected from patients with unexplained and new-onset diarrhea, defined as 3 or more unformed stools within a 24 hour period.11  An exception to this would be in patients with ileus or toxic megacolon suspected to be secondary to severe CDI. Second, testing should be avoided when a more likely cause of diarrhea, such as laxative use, is apparent. Following similar reasoning, other more likely causes should be evaluated before testing children under 2 years of age, and testing should generally be avoided in infants less than 1 year of age, given the high rates of asymptomatic colonization in these young age groups. In a multicenter investigation of children under 2 years of age, C. difficile detection by polymerase chain reaction was twice as common in asymptomatic children (28%) as in those presenting with acute gastroenteritis (14%), exemplifying the worth of incorporating age stratification in testing approach.53  Third, prolonged carriage can occur despite treatment, so repeat testing for proof of test of cure is not helpful.54 

The recommended diagnostic approach has evolved with time from toxin-based assays to NAAT-based testing and currently to multistep testing. As the best-performing approach for diagnosis, the 2017 IDSA and SHEA guidelines recommend using a stool toxin test as part of a 2-step algorithm (Fig 3), unless institutions have developed other agreed upon clinical and laboratory criteria for test submission, in which case detection of toxigenic C. difficile by NAAT alone is considered acceptable.11  More recently in 2021, the American College of Gastroenterologists developed guidelines for the preferred management of CDI in adults, which are similar but differ slightly to the 2017 IDSA and SHEA guidelines by recommending use of testing algorithms that include a highly sensitive and a highly specific testing modality to distinguish colonization from active infection.55  Despite societal guidance, testing practices for diagnosis of C. difficile in children varies considerably, suggesting further opportunities to improve diagnosis exist. Judicious testing of C. difficile ranks among one of the top 12 high-priority research topics in healthcare-associated infections and antimicrobial stewardship by the CDC.56,57  Incorporation of clinical decision support around test ordering in electronic health records and preauthorization of testing are additional strategies successfully implemented by some institutions to improve appropriate test ordering.58  Gaining in popularity, the use of multiplex gastrointestinal polymerase chain reaction panels that include C. difficile targets may unintentionally increase inappropriate testing, so some laboratories preferentially suppress C. difficile results from the panel unless specifically requested, as another way to limit over testing in populations with low prevalence of CDI.59 

FIGURE 3

A laboratory testing approach for diagnosis of Clostridioides difficile infection in children.a

aBased on the 2017 IDSA and SHEA guideline, which recommends a 2-step algorithm, including s stool toxin test (ie, glutamate dehydrogenase EIA plus Toxin EIA, arbitrated by NAAT or NAAT plus toxin EIA) as the best diagnostic approach.11  Discordant results require further clinical 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.

FIGURE 3

A laboratory testing approach for diagnosis of Clostridioides difficile infection in children.a

aBased on the 2017 IDSA and SHEA guideline, which recommends a 2-step algorithm, including s stool toxin test (ie, glutamate dehydrogenase EIA plus Toxin EIA, arbitrated by NAAT or NAAT plus toxin EIA) as the best diagnostic approach.11  Discordant results require further clinical 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.

Close modal

The search for novel stool biomarkers to improve diagnosis is an ongoing and pressing need. Measurement of fecal metabolites,60  proinflammatory cytokines,61  and volatile organic compounds62  all hold promise as potential future makers.

The management of CDI is determined by the number and severity of episodes. Children who do not respond to targeted treatment within 5 days should be re-evaluated for other causes. There is currently insufficient evidence to support probiotics as an alternative to antimicrobial therapy for CDI treatment.63 

For symptomatic children, initial management should include discontinuation of the inciting antibiotic, when possible, to limit further intestinal microbial disruption. Current antimicrobial therapies targeted against the initial CDI episode of mild or moderate severity include oral metronidazole and oral vancomycin (Table 2). Metronidazole, which has been a mainstay treatment of pediatric CDI, was removed as a recommended first line choice for adults in the 2017 IDSA and SHEA guidelines.11  Vancomycin and fidaxomicin are now recommended as preferred first line therapies in adults based on improved clinical cure rates and lower recurrence risk.11,6467  High quality data to support treatment guideline recommendation in children were lacking at the time, however postguideline shifts in prescribing practice with decreased use of metronidazole and increased use of oral vancomycin in children with CDI have been observed.7,68 

TABLE 2

Summary of Therapies for Children With Clostridioides difficile Infection^

PresentationDrug#RouteDoseFrequencyDuration
Initial infection 
Nonsevere Metronidazole* or PO 7.5 mg/kg per dose (max 500 mg) TID or QID 10 d 
 Vancomycin PO 10 mg/kg per dose (max 125 mg) QID 10 d 
Severea or fulminant Vancomycin PO or PR 10 mg/kg per dose (max 500 mgbQID 10 d 
 ± Metronidazole IV 10 mg/kg per dose (max 500 mg) TID 10 d 
Recurrent infection 
First recurrence Metronidazole* or PO 7.5 mg/kg per dose (max 500 mg) TID or QID 10 d 
 Vancomycin PO 10 mg/kg per dose (max 125 mg) QID 10 d 
Second or subsequent recurrencec Vancomycin or PO 10 mg/kg per dose (max 125 mg) QID 10 d (if not used previously) 
Vancomycin tapered and pulsed or PO 10 mg/kg per dose (max 125 mg) QID 10–14 d, then BID x 7 d, then once daily × 7 d, then every 2–3 d for 2–8 wk 
Vancomycin; followed by rifaximin chaser or PO 10 mg/kg per dose (max 125 mg); no pediatric dosing for rifaximin′ (max 400 mg) QID; TID 10 d; 20 d 
 FMT under IND — — — — 
PresentationDrug#RouteDoseFrequencyDuration
Initial infection 
Nonsevere Metronidazole* or PO 7.5 mg/kg per dose (max 500 mg) TID or QID 10 d 
 Vancomycin PO 10 mg/kg per dose (max 125 mg) QID 10 d 
Severea or fulminant Vancomycin PO or PR 10 mg/kg per dose (max 500 mgbQID 10 d 
 ± Metronidazole IV 10 mg/kg per dose (max 500 mg) TID 10 d 
Recurrent infection 
First recurrence Metronidazole* or PO 7.5 mg/kg per dose (max 500 mg) TID or QID 10 d 
 Vancomycin PO 10 mg/kg per dose (max 125 mg) QID 10 d 
Second or subsequent recurrencec Vancomycin or PO 10 mg/kg per dose (max 125 mg) QID 10 d (if not used previously) 
Vancomycin tapered and pulsed or PO 10 mg/kg per dose (max 125 mg) QID 10–14 d, then BID x 7 d, then once daily × 7 d, then every 2–3 d for 2–8 wk 
Vancomycin; followed by rifaximin chaser or PO 10 mg/kg per dose (max 125 mg); no pediatric dosing for rifaximin′ (max 400 mg) QID; TID 10 d; 20 d 
 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. +/– 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.

A retrospective, comparative effectiveness study of 192 children hospitalized with nonsevere CDI has since shown that those treated with vancomycin had earlier resolution of symptoms (86.3%) compared with those treated with metronidazole (71.1%) by day 5, whereas recurrence was similar.69  The recent landmark SUNSHINE study represents the first clinical trial conducted in children with CDI. In this phase 3 trial, 142 children with CDI were age-stratified and randomized to receive either oral fidaxomicin or vancomycin for 10 days. Children had to have negative rotavirus testing results to be eligible, and children with inflammatory bowel disease were excluded.70  No difference in clinical response at the end of therapy was shown, however at 30 days after therapy, the rate of sustained response was higher with fidaxomcin (68%) compared with vancomycin (50%), with an adjusted treatment difference of 18.8% (95% confidence interval 1.5%–35.3%). Children under 2 years of age were included, so it is unclear if other etiologies contributed to diarrhea in younger participants.71  Regardless, based on these positive results, in 2020 the US Food and Drug Administration (FDA) expanded approval of fidaxomicin for treatment of CDI to include children 6 months of age and older. These 2 studies open up new therapeutic options in children.

Based on limited data extrapolated from care in adults, children with more severe disease are treated with oral vancomycin, which can be administered via gastric feeding tube in intubated children. Intravenous metronidazole is often added on, though it is unclear whether colonic concentrations of metronidazole are adequate in this situation. There are no optimal treatments for fulminant disease. When complications, such as hypotension, shock, ileus, or toxic megacolon arise, multiple interventions are instituted simultaneously in hopes of improving outcomes. When ileus is present, administration of vancomycin via retention enema can be attempted. Surgical intervention, in the form of either organ-preserving diverting loop ileostomy or more invasive colectomy or FMT are other deliberations.11,72,73  Further studies are needed to improve outcomes from severe disease.

The optimal management of children who require treatment with other concomitant antibiotics beyond the recommended duration of CDI targeted therapy is unknown. In practice, some prolong the course of CDI treatment, though relapse rates were similar in adults who received extended CDI therapy beyond 14 days in retrospective review, and further studies are still needed.74,75  Prolonged use of metronidazole, which can be associated with neurotoxicity, should be avoided.

For the first recurrence, the 2017 IDSA and SHEA guidelines recommend retreatment can be with oral metronidazole or oral vancomycin in children (Table 2). Pulse-tapered vancomycin regimens or vancomycin course followed by rifaximin chaser can be prescribed for second or subsequent recurrences after that. Secondary prophylaxis with oral vancomycin while receiving systemic antibiotics may reduce the risk of recurrent CDI in the highest risk pediatric patients with an established history of CDI.76,77  Bezlotoxumab, an antitoxin B monoclonal antibody providing modest risk reduction, is currently recommended in adults at high risk of recurrence.64,78  Results of the phase 3 trial in children are expected soon.

FMT, which involves transfer of stool from a healthy donor to the gastrointestinal tract of a recipient with rCDI has gained popularity as a potential way to restore disrupted intestinal microbial composition. Traditionally, donor stool is transferred via endoscopy or enteric capsules for treatment, but enema may be an option if the other methods are unavailable. In a large retrospective review of FMT in 335 patients performed at 18 pediatric centers between 2014 and 2017, 81% of children were successfully treated, increasing to 87% with repeat FMT. Predictors of successful outcome included use of fresh donor stool over frozen stool, delivery by endoscopy, absence of a feeding tube, and lower number of recurrences of CDI before FMT. Just over 5% experienced an adverse reaction related to FMT, including diarrhea, pain, and bloating. There were 2 severe adverse events deemed related to FMT reported, including an aspiration pneumonia event post procedure and admission for emesis or diarrhea from inflammatory bowel disease relapse in another.79 

Despite these overall encouraging results, long term unanticipated consequences of FMT in children remain unknown. Changes in the microbiome have been implicated to contribute to autoimmune, metabolic, and psychiatric diseases, hence any treatment that involves alterations in the microbiome at such an early age warrants further long-term investigation.8082  The FDA has also issued several safety alerts regarding potential transmission of multidrug resistant organisms, severe acute respiratory syndrome coronavirus 2 virus and Mpox virus by FMT, highlighting the risk of transmission of emerging transmissible pathogens posed by FMT.8285  Published societal guidelines for children help to address screening and treatment but challenges around standardization and preparation of stool persist.86  FMT is considered investigational, hence regulations around administration apply.87 

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 diarrhea within 8 weeks was shown by clinical trial when compared with placebo, leading to FDA approval for adults in December 2022.88  Following closely behind in development is SER-109, an oral microbiome biotherapeutic composed of live purified Firmicutes bacterial spores from healthy donors. In a trial of adults at risk for rCDI, lower recurrence rates at 8 weeks after treatment were found compared with placebo.89  These developments represent important steps forward toward standardization of FMT, although they are several more steps further away from potentially being available options for use in children. Ridinilazole, a novel narrow spectrum antimicrobial;90  ibezapolstat, a DNA polymerase inhibitor;91  and MGB-BP-3, a novel topoisomerase inhibitor, are other C. difficile therapeutics advancing through later stages of clinical development.92 

All currently available C. difficile therapies have limitations to use (Table 3). Even established therapies such as vancomycin and metronidazole can increase the risk of recurrence, disrupt the gut microbiota, induce resistance, and promote proliferation of multidrug resistant bacteria, while none of the upcoming treatments are currently being studied in children as of yet.9399  Characteristics of an ideal C. difficile therapy include a safe and effective agent that can be administered directly to the gut with minimal influence on local microbial composition or systemic effect and provision of enduring protection. Promising therapies that can provide direct neutralizing activity using amoeba, yeast, or bacteria vehicles are under very early stages of exploration.

TABLE 3

Strengths and Limitations of Current C. difficile Therapies in Children

TherapeuticStrengthsLimitations
Metronidazole • Established experience • Broad spectrum gut microbial activity with recurrence risk;
• systemically absorbed;
• neurotoxic with prolonged or repeated use 
Vancomycin • Established experience;
• not systemically absorbed 
• Broad spectrum gut microbial activity with recurrence risk;
• promotes resistant pathogens such as vancomycin resistant enterococci 
Fidaxomicin • Narrow spectrum gut activity;
• not systemically absorbed 
• Limited experience with use in children;
• expensive;
• adverse effects of pyrexia, abdominal pain, emesis, diarrhea, constipation, elevation in liver enzyme activity and rash reported 
Bezlotoxumab • No systemic activity;
• no affect on gut dysbiosis 
• Insufficient data in children;
• 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 
TherapeuticStrengthsLimitations
Metronidazole • Established experience • Broad spectrum gut microbial activity with recurrence risk;
• systemically absorbed;
• neurotoxic with prolonged or repeated use 
Vancomycin • Established experience;
• not systemically absorbed 
• Broad spectrum gut microbial activity with recurrence risk;
• promotes resistant pathogens such as vancomycin resistant enterococci 
Fidaxomicin • Narrow spectrum gut activity;
• not systemically absorbed 
• Limited experience with use in children;
• expensive;
• adverse effects of pyrexia, abdominal pain, emesis, diarrhea, constipation, elevation in liver enzyme activity and rash reported 
Bezlotoxumab • No systemic activity;
• no affect on gut dysbiosis 
• Insufficient data in children;
• 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 

Prevention of CDI is an important goal to protect children and limit healthcare costs. In the absence of a safe and effective vaccine, prevention strategies rely on a 2-pronged approach of prevention of spread of C. difficile spores and prevention of factors that promote progression to disease in those with established colonization.

In healthcare settings, attention to infection control practices and facility cleaning may help to limit the spread of C. difficile. As alcohol-based hand sanitizers do not inactivate spores, exercising meticulous hand hygiene with soap and water after caring for a patient with CDI is recommended.11  Contact isolation includes the donning of gloves and gowns by healthcare workers, with dedicated use of private patient rooms when available. Gloves have been shown to reduce the risk of hand contamination when caring for patients with CDI.100  Computer-simulated modeling suggests screening asymptomatic children on admission may decrease hospital onset CDI by 28.5%, but incorporating universal screening as part of a multi-intervention bundle could prove logistically prohibitive for some institutions given the high rates of colonization in some pediatric populations coupled with pediatric bed shortages.101 

As spores remain hardy in the environment surviving months or longer, environmental cleaning with approved sporicidal disinfectants is advised, as staying in a hospital room previously occupied by a patient with CDI increases risk of the current occupant.102  Ultraviolet disinfection has become a widely used and promising adjunctive measure.103 

The American Academy of Pediatrics Committee on Infectious diseases advises that children with CDI be excluded from child care settings until stools are contained within the diaper or the child is continent and stool frequency is no more than 2 stools above normal frequency.104 

Exposure to antibiotics is the most important modifiable risk factor for development of CDI. Antibiotic stewardship programs that target reduction of inappropriate use in adults have proven helpful in reducing CDI rates, though comparable data in children are needed.105  Stewardship efforts are also needed in the outpatient setting, with at least 30% of outpatient antibiotic prescriptions estimated to be unnecessary.7  Probiotics have been used as a preventative measure for C. difficile in an attempt to restore some of the microbial disruption caused by antibiotics, the benefit of which is still debated. A large systematic review reported the risk of CDI was lowered 60% with probiotics. Conversely, in a recent trial of hospitalized children who received antibiotics randomized to receive Lactobacillus reuteri (2 × 108 CFU) or placebo twice daily for the duration of antibiotic treatment, no difference in prevention of antibiotic associated diarrhea was found.106  The wide heterogeneity in formulations, dose, and optimal timing casts uncertainty on the generalizability of probiotics for CDI prevention. The 2017 IDSA and SHEA guidelines state there is insufficient evidence at this time to recommend use for primary prevention of CDI11  Probiotics are generally well tolerated, though caution with use in severely immunocompromised or debilitated patients is needed and the cost to families must also be factored if used.107 

Other novel preventative approaches aimed at promoting colonization resistance by binding concomitantly administered antibiotics, such as activated charcoal and the oral β-lactamase ribaxamase, are under exploration.108,109  Demonstration of protection against disease by antibodies to toxin supports ongoing efforts to develop a vaccine, but both toxin based and nontoxin based candidates are much further away from use in children.110 

Recent progress made in our understanding of the epidemiology, risk, and management of CDI in children allows us to improve upon pediatric considerations in the diagnosis and treatment of this common infection (Table 4). Much of our current practice is still adapted from generalizing findings from adult studies, however significant knowledge gaps remain. Prioritizing translational research needs (Table 5) will allow us to make further advances to improve the outcomes of children affected by CDI.

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 
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 
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 
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 

Drs Shirley and Moonah conceptualized the review, drafted the manuscript, and reviewed and revised the manuscript; Mr Tornel contributed to the design and helped to draft the manuscript; Dr Warren critically reviewed the manuscript for important intellectual content; and all authors approved the final manuscript as submitted and agree to be accountable for all aspects of the work.

FUNDING: This work was supported by the Hartwell Foundation and the NIH, R01 DK131313 and R34 AI165304. The Hartwell Foundation and the NIH had no role in the design and conduct of the study.

CONFLICT OF INTEREST DISCLOSURES: Dr Warren is a medical advisor for SER-109. The rest of the authors have indicated they have no conflicts of interest relevant to this article to disclose.

CDC

Centers for Disease Control and Prevention

CDI

Clostridioides difficile infection

FDA

Food and Drug Administration

FMT

fecal microbiota transplantation

NAAT

nucleic acid amplification

rCDI

recurrent Clostridioides difficile infection

1
Hall
IC
,
O’Toole
E
.
Intestinal flora in new-born infants: with a description of a new pathogenic anaerobe, bacillus difficilis
.
Am J Dis Child
.
1935
;
49
(
2
):
390
402
2
Guh
AY
,
Mu
Y
,
Winston
LG
, et al;
Emerging Infections Program Clostridioides difficile Infection Working Group
.
Trends in U.S. burden of Clostridioides difficile infection and outcomes
.
N Engl J Med
.
2020
;
382
(
14
):
1320
1330
3
Centers for Disease Control and Prevention
.
Antibiotic resistance threats in the United States, 2019
.
Available at: www.cdc.gov/DrugResistance/Biggest-Threats.html. Accessed April 7, 2023
4
Miranda-Katz
M
,
Parmar
D
,
Dang
R
,
Alabaster
A
,
Greenhow
TL
.
Epidemiology and risk factors for community associated Clostridioides difficile in children
.
J Pediatr
.
2020
;
221
:
99
106
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
6
Zilberberg
MD
,
Tillotson
GS
,
McDonald
C
.
Clostridium difficile infections among hospitalized children, United States, 1997-2006
.
Emerg Infect Dis
.
2010
;
16
(
4
):
604
609
7
Edwards
PT
,
Thurm
CW
,
Hall
M
, et al
.
Clostridioides difficile infection in hospitalized pediatric patients: comparisons of epidemiology, testing, and treatment from 2013 to 2019
.
J Pediatr
.
2023
;
252
:
111
116.e1
8
Silva
A
,
Du
T
,
Choi
KB
, et al;
CNISP C. difficile working group
.
Epidemiology of primary and recurrent healthcare-associated and community-associated pediatric Clostridioides difficile infection in Canada, 2015-2020
.
J Pediatric Infect Dis Soc
.
2023
;
12
(
4
):
222
225
9
Mehrotra
P
,
Jang
J
,
Gidengil
C
,
Sandora
TJ
.
Attributable cost of Clostridium difficile infection in pediatric patients
.
Infect Control Hosp Epidemiol
.
2017
;
38
(
12
):
1472
1477
10
Lurienne
L
,
Bandinelli
PA
,
Galvain
T
,
Coursel
CA
,
Oneto
C
,
Feuerstadt
P
.
Perception of quality of life in people experiencing or having experienced a Clostridioides difficile infection: a US population survey
.
J Patient Rep Outcomes
.
2020
;
4
(
1
):
14
11
McDonald
LC
,
Gerding
DN
,
Johnson
S
, et al
.
Clinical practice guidelines for Clostridium difficile infection in adults and children: 2017 update by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA)
.
Clin Infect Dis
.
2018
;
66
(
7
):
e1
e48
12
Centers for Disease Control and Prevention
.
Healthcare-Associated Infections - Community Interface (HAIC) Clostridioides difficile 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 incidence of Clostridium difficile related discharges among hospitalized children in the United States
.
Infect Control Hosp Epidemiol
.
2016
;
37
(
1
):
104
106
14
Centers for Disease Control and Prevention
.
COVID-19: U.S. impact on antimicrobial resistance, special report 2022
.
15
Lees
EA
,
Carrol
ED
,
Ellaby
NAF
, et al
.
Characterization of circulating Clostridium difficile strains, host response and intestinal microbiome in hospitalized children with diarrhea
.
Pediatr Infect Dis J
.
2020
;
39
(
3
):
221
228
16
Shuai
H
,
Bian
Q
,
Luo
Y
, et al
.
Molecular characteristics of Clostridium difficile in children with acute gastroenteritis from Zhejiang
.
BMC Infect Dis
.
2020
;
20
(
1
):
343
17
Perumalsamy
S
,
Lim
SC
,
Riley
TV
.
Clostridioides (Clostridium) difficile isolated from paediatric patients in Western Australia 2019-2020
.
Pathology
.
2022
;
54
(
4
):
460
465
18
Al-Rawahi
GN
,
Al-Najjar
A
,
McDonald
R
, et al
.
Pediatric oncology and stem cell transplant patients with healthcare-associated Clostridium difficile infection were already colonized on admission
.
Pediatr Blood Cancer
.
2019
;
66
(
5
):
e27604
19
Tougas
SR
,
Lodha
N
,
Vandermeer
B
, et al
.
Prevalence of detection of Clostridioides difficile among asymptomatic children: a systematic review and meta-analysis
.
JAMA Pediatr
.
2021
;
175
(
10
):
e212328
20
Pichler
K
,
Bausenhardt
B
,
Huhulescu
S
, et al
.
Impact and time course of Clostridium difficile colonization in very low birth weight infants
.
Neonatology
.
2018
;
114
(
2
):
149
154
21
Ferraris
L
,
Couturier
J
,
Eckert
C
, et al
.
Carriage and colonization of C. difficile in preterm neonates: a longitudinal prospective study
.
PLoS One
.
2019
;
14
(
2
):
e0212568
22
Brennhofer
SA
,
Rogawski McQuade
ET
,
Liu
J
,
Guerrant
RL
,
Platts-Mills
JA
,
Warren
CA
.
Clostridioides difficile colonization among very young children in resource-limited settings
.
Clin Microbiol Infect
.
2022
;
28
(
7
):
996
1002
23
Xie
X
,
Xiang
B
,
Wu
Y
,
Zhao
Y
,
Wang
Q
,
Jiang
X
.
Infant progressive colonic stenosis caused by antibiotic-related Clostridium difficile colitis - a case report and literature review
.
BMC Pediatr
.
2018
;
18
(
1
):
320
24
Eglow
R
,
Pothoulakis
C
,
Itzkowitz
S
, et al
.
Diminished Clostridium difficile toxin A sensitivity in newborn rabbit ileum is associated with decreased toxin A receptor
.
J Clin Invest
.
1992
;
90
(
3
):
822
829
25
Kociolek
LK
,
Espinosa
RO
,
Gerding
DN
, et al
.
Natural Clostridioides difficile toxin immunization in colonized infants
.
Clin Infect Dis
.
2020
;
70
(
10
):
2095
2102
26
Hourigan
SK
,
Sears
CL
,
Oliva-Hemker
M
.
Clostridium difficile infection in pediatric inflammatory bowel disease
.
Inflamm Bowel Dis
.
2016
;
22
(
4
):
1020
1025
27
Theunissen
C
,
Knoop
C
,
Nonhoff
C
, et al
.
Clostridium difficile colitis in cystic fibrosis patients with and without lung transplantation
.
Transpl Infect Dis
.
2008
;
10
(
4
):
240
244
28
Dominguez
SR
,
Dolan
SA
,
West
K
, et al
.
High colonization rate and prolonged shedding of Clostridium difficile in pediatric oncology patients
.
Clin Infect Dis
.
2014
;
59
(
3
):
401
403
29
Ochfeld
E
,
Balmert
LC
,
Patel
SJ
,
Muller
WJ
,
Kociolek
LK
.
Risk factors for Clostridioides (Clostridium) difficile infection following solid organ transplantation in children
.
Transpl Infect Dis
.
2019
;
21
(
5
):
e13149
30
Duclaux-Loras
R
,
Berthiller
J
,
Ferroni
A
, et al
.
Clostridium difficile: a frequent infection in children after intestinal transplantation
.
Transplantation
.
2020
;
104
(
1
):
197
200
31
Leibowitz
J
,
Soma
V
,
Rosen
L
,
Ginocchio
C
,
Rubin
L
.
Similar proportions of stool specimens from hospitalized children with and without diarrhea test positive for Clostridium difficile
.
Pediatr Infect Dis J
.
2014
;
34
(
3
):
261
266
32
Anjewierden
S
,
Han
Z
,
Foster
CB
,
Pant
C
,
Deshpande
A
.
Risk factors for Clostridium difficile infection in pediatric inpatients: a meta-analysis and systematic review
.
Infect Control Hosp Epidemiol
.
2019
;
40
(
4
):
420
426
33
Adams
DJ
,
Eberly
MD
,
Rajnik
M
,
Nylund
CM
.
Risk factors for community-associated Clostridium difficile infection in children
.
J Pediatr
.
2017
;
186
:
105
109
34
Weng
MK
,
Adkins
SH
,
Bamberg
W
, et al
.
Risk factors for community-associated Clostridioides difficile infection in young children
.
Epidemiol Infect
.
2019
;
147
:
e172
35
Ramirez
J
,
Guarner
F
,
Bustos Fernandez
L
,
Maruy
A
,
Sdepanian
VL
,
Cohen
H
.
Antibiotics as major disruptors of gut microbiota
.
Front Cell Infect Microbiol
.
2020
;
10
:
572912
36
Smith
AB
,
Jenior
ML
,
Keenan
O
, et al
.
Enterococci enhance Clostridioides difficile pathogenesis
.
Nature
.
2022
;
611
(
7937
):
780
786
37
Dang
R
,
Alabaster
A
,
Miranda-Katz
M
,
Parmar
D
,
Greenhow
TL
.
Impact of antecedent antibiotic usage on community-associated Clostridioides difficile infection in Pediatrics
.
Pediatr Infect Dis J
.
2021
;
40
(
5
):
426
428
38
Kociolek
LK
,
Gerding
DN
,
Espinosa
RO
,
Patel
SJ
,
Shulman
ST
,
Ozer
EA
.
Clostridium difficile Whole Genome Sequencing Reveals Limited Transmission Among Symptomatic Children: A Single-Center Analysis
.
Clin Infect Dis
.
2018
;
67
(
2
):
229
234
39
Chitnis
AS
,
Holzbauer
SM
,
Belflower
RM
, et al
.
Epidemiology of community-associated Clostridium difficile infection, 2009 through 2011
.
JAMA Intern Med
.
2013
;
173
(
14
):
1359
1367
40
Wilcox
MH
,
Mooney
L
,
Bendall
R
,
Settle
CD
,
Fawley
WN
.
A case-control study of community-associated Clostridium difficile infection
.
J Antimicrob Chemother
.
2008
;
62
(
2
):
388
396
41
Rabold
D
,
Espelage
W
,
Abu Sin
M
, et al
.
The zoonotic potential of Clostridium difficile from small companion animals and their owners
.
PLoS One
.
2018
;
13
(
2
):
e0193411
42
Kociolek
LK
,
Patel
SJ
,
Shulman
ST
,
Gerding
DN
.
Molecular epidemiology of Clostridium difficile infections in children: a retrospective cohort study
.
Infect Control Hosp Epidemiol
.
2015
;
36
(
4
):
445
451
43
Perumalsamy
S
,
Riley
TV
.
Molecular epidemiology of Clostridioides difficile infections in children
.
J Pediatric Infect Dis Soc
.
2021
;
10
(
Supplement_3
):
S34
S40
44
Loftus
KV
,
Wilson
PM
.
A curiously rare case of septic shock from Clostridium difficile colitis
.
Pediatr Emerg Care
.
2017
;
33
(
8
):
576
578
45
Wendt
JM
,
Cohen
JA
,
Mu
Y
, et al
.
Clostridium difficile infection among children across diverse US geographic locations
.
Pediatrics
.
2014
;
133
(
4
):
651
658
46
Nicholson
MR
,
Crews
JD
,
Starke
JR
,
Jiang
ZD
,
DuPont
H
,
Edwards
K
.
Recurrent Clostridium difficile infection in children: patient risk factors and markers of intestinal inflammation
.
Pediatr Infect Dis J
.
2017
;
36
(
4
):
379
383
47
Orenstein
R
,
Patron
RL
,
Seville
MT
.
Why does Clostridium difficile infection recur?
J Am Osteopath Assoc
.
2019
;
119
(
5
):
322
326
48
Chang
JY
,
Antonopoulos
DA
,
Kalra
A
, et al
.
Decreased diversity of the fecal microbiome in recurrent Clostridium difficile-associated diarrhea
.
J Infect Dis
.
2008
;
197
(
3
):
435
438
49
Schwab
EM
,
Wilkes
J
,
Korgenski
K
,
Hersh
AL
,
Pavia
AT
,
Stevens
VW
.
Risk factors for recurrent Clostridium difficile infection in pediatric inpatients
.
Hosp Pediatr
.
2016
;
6
(
6
):
339
344
50
Ruan
W
,
Kellermayer
R
.
Alternative diagnoses in pediatric fecal microbiota transplant referral patients
.
J Pediatr Gastroenterol Nutr
.
2021
;
72
(
5
):
693
696
51
Sandora
TJ
,
Williams
DN
,
Daugherty
K
, et al
.
Stool toxin concentration does not distinguish Clostridioides difficile infection from colonization in children less than 3 years of age
.
J Pediatric Infect Dis Soc
.
2022
;
11
(
10
):
454
458
52
Parnell
JM
,
Fazili
I
,
Bloch
SC
, et al
.
Two-step testing for Clostridioides difficile is inadequate in differentiating infection from colonization in children
.
J Pediatr Gastroenterol Nutr
.
2021
;
72
(
3
):
378
383
53
Pahud
BA
,
Hassan
F
,
Harrison
CJ
, et al
.
Detection of Clostridioides difficile by real-time PCR in young children does not predict disease
.
Hosp Pediatr
.
2020
;
10
(
7
):
555
562
54
Sethi
AK
,
Al-Nassir
WN
,
Nerandzic
MM
,
Bobulsky
GS
,
Donskey
CJ
.
Persistence of skin contamination and environmental shedding of Clostridium difficile during and after treatment of C. difficile infection
.
Infect Control Hosp Epidemiol
.
2010
;
31
(
1
):
21
27
55
Kelly
CR
,
Fischer
M
,
Allegretti
JR
, et al
.
ACG clinical guidelines: prevention, diagnosis, and treatment of Clostridioides difficile infections
.
Am J Gastroenterol
.
2021
;
116
(
6
):
1124
1147
56
Coffin
SE
,
Abanyie
F
,
Bryant
K
, et al;
CDC Prevention Epicenters Program
.
Pediatric research priorities in healthcare-associated infections and antimicrobial stewardship
.
Infect Control Hosp Epidemiol
.
2021
;
42
(
5
):
519
522
57
Kociolek
LK
,
Kutty
PK
,
Polgreen
PM
,
Beekmann
SE
.
Healthcare provider diagnostic testing practices for identification of Clostridioides (Clostridium) difficile in children: an Emerging Infections Network survey
.
Infect Control Hosp Epidemiol
.
2019
;
40
(
3
):
276
280
58
Tran
NN
,
Mills
JP
,
Zimmerman
C
, et al
.
Incorporating preauthorization into antimicrobial stewardship pharmacist workflow reduces Clostridioides difficile and gastrointestinal panel testing
.
Infect Control Hosp Epidemiol
.
2020
;
41
(
10
):
1136
1141
59
Cotter
JM
,
Thomas
J
,
Birkholz
M
, et al
.
Impact of multiplex testing on the identification of pediatric Clostridiodes difficile
.
J Pediatr
.
2020
;
218
:
157
165.e3
60
Zhou
P
,
Zhou
N
,
Shao
L
, et al
.
Diagnosis of Clostridium difficile infection using an UPLC-MS based metabolomics method
.
Metabolomics
.
2018
;
14
(
8
):
102
61
Gálvez
JAV
,
Pollock
NR
,
Alonso
CD
, et al
.
Stool interleukin-1β differentiates Clostridioides difficile infection from asymptomatic carriage and non-C.difficile infection diarrhea
.
Clin Infect Dis
.
2023
;
76
(
3
):
e1467
e1475
62
Patel
M
,
Fowler
D
,
Sizer
J
,
Walton
C
.
Faecal volatile biomarkers of Clostridium difficile infection
.
PLoS One
.
2019
;
14
(
4
):
e0215256
63
Ofosu
A
.
Clostridium difficile infection: a review of current and emerging therapies
.
Ann Gastroenterol
.
2016
;
29
(
2
):
147
154
64
Johnson
S
,
Lavergne
V
,
Skinner
AM
, et al
.
Clinical practice guideline by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA): 2021 focused update guidelines on management of Clostridioides difficile infection in adults
.
Clin Infect Dis
.
2021
;
73
(
5
):
e1029
e1044
65
Louie
TJ
,
Miller
MA
,
Mullane
KM
, et al;
OPT-80-003 Clinical Study Group
.
Fidaxomicin versus vancomycin for Clostridium difficile infection
.
N Engl J Med
.
2011
;
364
(
5
):
422
431
66
Guery
B
,
Menichetti
F
,
Anttila
VJ
, et al;
EXTEND Clinical Study Group
.
Extended-pulsed fidaxomicin versus vancomycin for Clostridium difficile infection in patients 60 years and older (EXTEND): a randomised, controlled, open-label, phase 3b/4 trial
.
Lancet Infect Dis
.
2018
;
18
(
3
):
296
307
67
Mikamo
H
,
Tateda
K
,
Yanagihara
K
, et al
.
Efficacy and safety of fidaxomicin for the treatment of Clostridioides (Clostridium) difficile infection in a randomized, double-blind, comparative phase III study in Japan
.
J Infect Chemother
.
2018
;
24
(
9
):
744
752
68
Stultz
JS
,
Hopp
J
,
Orndahl
CM
,
Omecene
NE
,
Pakyz
AL
.
Changes in metronidazole and vancomycin utilization for nonsevere Clostridioides difficile infection among institutions caring for children
.
Pediatr Infect Dis J
.
2021
;
40
(
7
):
634
636
69
Yin
J
,
Kociolek
LK
,
Same
RG
,
Hsu
AJ
,
Amoah
J
,
Tamma
PD
.
Oral vancomycin may be associated with earlier symptom resolution than metronidazole for hospitalized children with nonsevere Clostridiodes difficile infections
.
Open Forum Infect Dis
.
2019
;
6
(
12
):
ofz492
70
Wolf
J
,
Kalocsai
K
,
Fortuny
C
, et al
.
Safety and efficacy of fidaxomicin and vancomycin in children and adolescents with Clostridioides (Clostridium) difficile infection: a phase 3, multicenter, randomized, single-blind clinical trial (SUNSHINE)
.
Clin Infect Dis
.
2020
;
71
(
10
):
2581
2588
71
Kociolek
LK
.
Moving the needle on Clostridioides difficile research in children: a major step forward but challenges remain
.
Clin Infect Dis
.
2020
;
71
(
10
):
2589
2591
72
Neal
MD
,
Alverdy
JC
,
Hall
DE
,
Simmons
RL
,
Zuckerbraun
BS
.
Diverting loop ileostomy and colonic lavage: an alternative to total abdominal colectomy for the treatment of severe, complicated Clostridium difficile associated disease
.
Ann Surg
.
2011
;
254
(
3
):
423
427
,
discussion 427–429
73
Fashandi
AZ
,
Martin
AN
,
Wang
PT
, et al
.
An institutional comparison of total abdominal colectomy and diverting loop ileostomy and colonic lavage in the treatment of severe, complicated Clostridium difficile infections
.
Am J Surg
.
2017
;
213
(
3
):
507
511
74
Fitzpatrick
F
,
Safdar
N
,
van Prehn
J
,
Tschudin-Sutter
S
.
How can patients with Clostridioides difficile infection on concomitant antibiotic treatment be best managed?
Lancet Infect Dis
.
2022
;
22
(
11
):
e336
e340
75
Kaki
R
,
Brooks
A
,
Main
C
,
Jayaratne
P
,
Mertz
D
.
Does extending Clostridium difficile treatment in patients who are receiving concomitant antibiotics reduce the rate of relapse?
The Internet Journal of Infectious Diseases
.
2016
;
15
(
1
)
76
Bao
H
,
Lighter
J
,
Dubrovskaya
Y
, et al
.
Oral vancomycin as secondary prophylaxis for Clostridioides difficile infection
.
Pediatrics
.
2021
;
148
(
2
):
e2020031807
77
Shin
JH
,
Chaplin
AS
,
Hays
RA
, et al
.
Outcomes of a multidisciplinary clinic in evaluating recurrent Clostridioides difficile infection patients for fecal microbiota transplant: a retrospective cohort analysis
.
J Clin Med
.
2019
;
8
(
7
):
1036
78
Gerding
DN
,
Kelly
CP
,
Rahav
G
, et al
.
Bezlotoxumab for prevention of recurrent Clostridium difficile infection in patients at increased risk for recurrence
.
Clin Infect Dis
.
2018
;
67
(
5
):
649
656
79
Nicholson
MR
,
Mitchell
PD
,
Alexander
E
, et al
.
Efficacy of fecal microbiota transplantation for Clostridium difficile infection in children
.
Clin Gastroenterol Hepatol
.
2020
;
18
(
3
):
612
619.e1
80
De Palma
G
,
Lynch
MD
,
Lu
J
, et al
.
Transplantation of fecal microbiota from patients with irritable bowel syndrome alters gut function and behavior in recipient mice
.
Sci Transl Med
.
2017
;
9
(
379
):
eaaf6397
81
Turnbaugh
PJ
,
Ley
RE
,
Mahowald
MA
,
Magrini
V
,
Mardis
ER
,
Gordon
JI
.
An obesity-associated gut microbiome with increased capacity for energy harvest
.
Nature
.
2006
;
444
(
7122
):
1027
1031
82
Bibiloni
R
,
Mangold
M
,
Madsen
KL
,
Fedorak
RN
,
Tannock
GW
.
The bacteriology of biopsies differs between newly diagnosed, untreated, Crohn’s disease and ulcerative colitis patients
.
J Med Microbiol
.
2006
;
55
(
Pt 8
):
1141
1149
83
US Food and Drug Administration
.
Safety alert regarding use of fecal microbiota for transplantation and additional safety protections pertaining to monkeypox virus
.
84
US Food and Drug Administration Safety Communication
.
Important safety alert regarding use of fecal microbiota for transplantation and risk of serious adverse reactions due to transmission of multi-drug resistant organisms
.
85
US Food and Drug Administration
.
Fecal microbiota for transplantation: safety alert - regarding additional safety protections pertaining to SARS-CoV-2 and COVID-19
.
86
Davidovics
ZH
,
Michail
S
,
Nicholson
MR
, et al;
FMT Special Interest Group of the North American Society of Pediatric Gastroenterology Hepatology, Nutrition, the European Society for Pediatric Gastroenterology Hepatology, Nutrition
.
Fecal microbiota transplantation for recurrent Clostridium difficile infection and other conditions in children: a joint position paper from the North American Society for Pediatric Gastroenterology, Hepatology, and Nutrition and the European Society for Pediatric Gastroenterology, Hepatology, and Nutrition
.
J Pediatr Gastroenterol Nutr
.
2019
;
68
(
1
):
130
143
87
Food and Drug Administration
.
Enforcement policy regarding investigational new drug requirements for use of fecal microbiota for transplantation to treat Clostridioides difficile infection not responsive to standard therapies
.
Available at: https://www.fda.gov/media/86440/download. Accessed November 15, 2022
88
Khanna
S
,
Assi
M
,
Lee
C
, et al
.
Efficacy and Safety of RBX2660 in PUNCH CD3, a Phase III, Randomized, Double-Blind, Placebo-Controlled Trial with a Bayesian Primary Analysis for the Prevention of Recurrent Clostridioides difficile Infection
.
Drugs
.
2022
;
82
(
15
):
1527
1538
89
Feuerstadt
P
,
Louie
TJ
,
Lashner
B
, et al
.
SER-109, an oral microbiome therapy for recurrent Clostridioides difficile infection
.
N Engl J Med
.
2022
;
386
(
3
):
220
229
90
Carlson
TJ
,
Endres
BT
,
Bassères
E
,
Gonzales-Luna
AJ
,
Garey
KW
.
Ridinilazole for the treatment of Clostridioides difficile infection
.
Expert Opin Investig Drugs
.
2019
;
28
(
4
):
303
310
91
Garey
KW
,
McPherson
J
,
Dinh
AQ
, et al
.
Efficacy, safety, pharmacokinetics, and microbiome changes of ibezapolstat in adults with Clostridioides difficile infection: a phase 2a multicenter clinical trial
.
Clin Infect Dis
.
2022
;
75
(
7
):
1164
1170
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
.
2022
;
8
(
12
):
2552
2563
93
Džunková
M
,
D’Auria
G
,
Xu
H
, et al
.
The monoclonal antitoxin antibodies (Actoxumab-Bezlotoxumab) treatment facilitates normalization of the gut microbiota of mice with Clostridium difficile infection
.
Front Cell Infect Microbiol
.
2016
;
6
:
119
94
Kelly
CP
,
LaMont
JT
.
Clostridium difficile--more difficult than ever
.
N Engl J Med
.
2008
;
359
(
18
):
1932
1940
95
O’Horo
JC
,
Jindai
K
,
Kunzer
B
,
Safdar
N
.
Treatment of recurrent Clostridium difficile infection: a systematic review
.
Infection
.
2014
;
42
(
1
):
43
59
96
Bossù
G
,
Di Sario
R
,
Argentiero
A
,
Esposito
S
.
Antimicrobial prophylaxis and modifications of the gut microbiota in children with cancer
.
Antibiotics (Basel)
.
2021
;
10
(
2
):
152
97
Huang
H
,
Weintraub
A
,
Fang
H
,
Nord
CE
.
Antimicrobial resistance in Clostridium difficile
.
Int J Antimicrob Agents
.
2009
;
34
(
6
):
516
522
98
Baines
SD
,
O’Connor
R
,
Freeman
J
, et al
.
Emergence of reduced susceptibility to metronidazole in Clostridium difficile
.
J Antimicrob Chemother
.
2008
;
62
(
5
):
1046
1052
99
Martin
H
,
Willey
B
,
Low
DE
, et al
.
Characterization of Clostridium difficile strains isolated from patients in Ontario, Canada, from 2004 to 2006
.
J Clin Microbiol
.
2008
;
46
(
9
):
2999
3004
100
Landelle
C
,
Verachten
M
,
Legrand
P
,
Girou
E
,
Barbut
F
,
Brun-Buisson
C
.
Contamination of healthcare workers’ hands with Clostridium difficile spores after caring for patients with C. difficile infection
.
Infect Control Hosp Epidemiol
.
2014
;
35
(
1
):
10
15
101
Barker
AK
,
Scaria
E
,
Alagoz
O
,
Sethi
AK
,
Safdar
N
.
Reducing C. difficile in children: an agent-based modeling approach to evaluate intervention effectiveness
.
Infect Control Hosp Epidemiol
.
2020
;
41
(
5
):
522
530
102
Sood
G
,
Truelove
S
,
Dougherty
G
, et al
.
Clostridioides difficile infection (CDI) in a previous room occupant predicts CDI in subsequent room occupants across different hospital settings
.
Am J Infect Control
.
2022
;
50
(
12
):
1352
1354
103
Steele
M
,
Hurtado
RR
,
Rychlik
K
, et al
.
Impact of an automated multiple emitter whole-room ultraviolet-C disinfection system on hospital acquired infections: a quasi-experimental study
.
Am J Infect Control
.
2021
;
49
(
9
):
1200
1203
104
Kimberlin
DW
,
Barnett
ED
,
Lynfield
R
,
Sawyer
MH
;
Committee on Infectious Diseases
,
American Academy of Pediatrics
.
Red Book: 2021–2024 Report of the Committee on Infectious Diseases
.
Itasca, IL
:
American Academy of Pediatrics
;
2021
105
Shea
KM
,
Hobbs
ALV
,
Jaso
TC
, et al
.
Effect of a health care system respiratory fluoroquinolone restriction program to alter utilization and impact rates of Clostridium difficile infection
.
Antimicrob Agents Chemother
.
2017
;
61
(
6
):
e00125
e00117
106
Kołodziej
M
,
Szajewska
H
.
Lactobacillus reuteri DSM 17938 in the prevention of antibiotic-associated diarrhoea in children: a randomized clinical trial
.
Clin Microbiol Infect
.
2019
;
25
(
6
):
699
704
107
Goldenberg
JZ
,
Yap
C
,
Lytvyn
L
, et al
.
Probiotics for the prevention of Clostridium difficile-associated diarrhea in adults and children
.
Cochrane Database Syst Rev
.
2017
;
12
(
12
):
CD006095
108
Vehreschild
MJGT
,
Ducher
A
,
Louie
T
, et al
.
An open randomized multicentre phase 2 trial to assess the safety of DAV132 and its efficacy to protect gut microbiota diversity in hospitalized patients treated with fluoroquinolones
.
J Antimicrob Chemother
.
2022
;
77
(
4
):
1155
1165
109
Kokai-Kun
JF
,
Le
C
,
Trout
K
, et al
.
Ribaxamase, an orally administered β-Lactamase, diminishes changes to acquired antimicrobial resistance of the gut resistome in patients treated with ceftriaxone
.
Infect Drug Resist
.
2020
;
13
:
2521
2535
110
Riley
TV
,
Lyras
D
,
Douce
GR
.
Status of vaccine research and development for Clostridium difficile
.
Vaccine
.
2019
;
37
(
50
):
7300
7306
111
Luna
RA
,
Boyanton
BL
Jr
,
Mehta
S
, et al
.
Rapid stool-based diagnosis of Clostridium difficile infection by real-time PCR in a children’s hospital
.
J Clin Microbiol
.
2011
;
49
(
3
):
851
857
112
DIFICID
.
(Fidaxomicin) [package insert]
.
Whitehouse Station, NJ
:
Merck
;
2020
This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License (https://creativecommons.org/licenses/by-nc-nd/4.0/), which permits noncommercial, distribution, and reproduction in any medium, provided the original author and source are credited.