<em>Clostridioides difficile</em> Infection: Updates on Epidemiologic Patterns, Diagnostic Tools, and Treatment Modalities
PDF
Cite
Share
Request
REVIEW
P: 10-10
January 2023

Clostridioides difficile Infection: Updates on Epidemiologic Patterns, Diagnostic Tools, and Treatment Modalities

Mediterr J Infect Microb Antimicrob 2023;12(1):10-10
1. Cairo University Faculty of Medicine, Department of Microbiology and Immunology, Egypt
No information available.
No information available
PDF
Cite
Share
Request

Summary

Clostridioides difficile is the predominant worldwide etiology of healthcare-associated diarrhea. Furthermore, C. difficile infections (CDIs) have been designated by the Centers for Disease Control and Prevention as an urgent threat, which is the highest of all threat levels. Throughout the years, epidemiologic surveillance efforts and infection prevention measures have been focused on combating healthcare-associated CDIs. Nonetheless, the incidence of community-associated infection is currently witnessing an upsurge. In the meantime, insufficient clinicians’ awareness, inadequate frequency of testing, or a suboptimal diagnostic scheme can result in underdiagnosis with the subsequent pervasion of CDIs. Another factor contributing to the escalating morbidities is the scarcity of anti-clostridial therapeutics that can tackle notorious re-infections and relapses. Altogether, these factors warrant raising awareness about epidemiologic patterns, diagnostic algorithms, and the updated treatment regimens for CDI.

Introduction

Clostridioides difficile infection (CDI) is a cardinal cause of infectious diarrhea and one of the most prevalent healthcare-associated infections globally[1]. It inflicts approximately 8 in 100,000 individuals annually; in the hospital setting, it inflicts 4-8 of 1000 patients[2]. In the United States, the financial toll of CDIs hits up to 7 billion dollars annually, as CDIs prolong the hospital stay by 2.8-10.4 days[1, 3], with a burden exceeding $42,000 for each patient[3].

The disease is caused by the activity of three toxins produced by C. difficile (enterotoxin A, cytotoxin B, and binary toxin)[5]. Through glucosyltransferase action, toxins A and B inactivate GTPases, the regulators of cytoplasmic F-actin. Meanwhile, the binary toxin is an ADP-ribosyltransferase that catalyzes actin depolymerization[6]. Together, the toxins disrupt the cytoskeleton and tight junctions between gut epithelial cells[7]. Such a disruption provokes an inflammatory response with the liberation of cytokines and leukotrienes, eventually triggering diarrhea and pseudomembranous colitis[8, 9]. Additionally, C. difficile spores are extremely resistant structures that promote feco-oral transmission and cause relapses in those who temporarily recover[10].

In recent years, massive worldwide efforts have been conducted to curb healthcare-associated CDIs. However, one of the hurdles has been the emergence of hypervirulent strains including ribotype 027 that produce exceedingly large amounts of toxins[11]. Moreover, CDIs are no longer restricted to healthcare settings. Although initially believed to be a nosocomial pathogen, increasing proof reveals that CDI is acquired in the community[12], where it accounts for 37% of all CDIs[13].

Despite numerous attempts at competent prevention and effective treatments, only a few drugs are approved for the treatment of CDIs, mainly fidaxomicin, vancomycin, and metronidazole. A challenge in employing either vancomycin or metronidazole is that both agents perturb the colonic microflora. Concerning metronidazole, a further restraint is that it is totally absorbed from the intestine, leaving only trace amounts at the infection site. These shortcomings contribute to the high rate of treatment failure and relapse[14]. Fidaxomicin, approved by the Food and Drug Administration (FDA) in 2011, has an enhanced profile of its narrow spectrum and oral bioavailability[15]. Nonetheless, the treatment outcome is still far from satisfactory[16].

Consequently, in the 2019 report on antimicrobial resistance, the Centers for Disease Control and Prevention continued to designate CDIs as an urgent health threat, the highest of threat levels, with nearly 13,000 deaths in US hospitals annually[17].

Considering the previous data, the purpose of this article is to spotlight the updates in the epidemiologic features of C. difficile as well as to delineate the diagnostic approaches and novel treatment algorithms for CDIs.

Epidemiology of CDIs

C. difficile colonizes the intestinal tract mainly through feco-oral transmission and contact with contaminated surfaces[18]. The spectrum of CDIs comprises asymptomatic carriage, mild diarrhea, and life-threatening fulminant disease with sepsis, toxic megacolon, and pancolitis that may end in colectomy[12]. Even after recovery from initial CDI, patients remain at risk of recurrence. Approximately 18-35% of individuals treated for CDI experience one or more episodes within 2-8 weeks of initial CDI[19]. The subsequent episode can be classified as either a “relapse” with the same strain or a “reinfection” with a new C. difficile strain[20].

Of note, the mortality rate of CDIs reaches 12.6%[21] and is remarkably higher in patients with inflammatory bowel disease and those residing in intensive care units (ICUs)[22].

1. Healthcare-associated CDIs (HA-CDIs)

In US hospitals, C. difficile represents the most frequently reported healthcare-associated pathogen (15% of all infections with a reported pathogen)[23], at an estimated burden of 462,000 cases of CDIs in 2017[24]. Moreover, data from 28 hospitals in the US imply that CDIs have replaced methicillin-resistant Staphylococcus aureus as the most frequent etiology of healthcare-associated infections, ranking third behind urinary catheter-related infections and surgical site infections[25].

Compared with hospitalized individuals without CDIs, those with CDIs as a secondary diagnosis experience a threefold longer hospital stay, have a 3.5-fold rise in hospital expenditure, and are six times as likely to die[26].

On the contrary, a meta-analysis of data from 111 studies demonstrated that the pooled CDI prevalence rate in the Middle East is 10.2% among patients with diarrhea[27]. Another meta-analysis of 85 studies from developing countries revealed a 15% prevalence of CDIs among patients with diarrhea[28]. Meanwhile, data from Egypt demonstrated the prevalence of C. difficile in 13.7% of inpatients suffering from diarrhea[29]. Another Egyptian study revealed that C. difficile was isolated from pediatric and adult inpatients with diarrhea at a rate of 17.9% and 27%, respectively[30].

In low- and medium-income populations, CDIs are likely underreported owing to inadequate awareness and limited diagnostic resources[31]. Available data indicate that CDIs are, nonetheless, a significant etiology of diarrhea in low-resource settings[28, 32].

On the contrary, certain factors increase the risk of HA-CDIs. Prior intake of antimicrobials, namely, cephalosporins, clindamycin, and less commonly fluoroquinolones, is a frequent risk factor[33]. CDIs are also more encountered in older patients and those with comorbid events, e.g., inflammatory bowel disease[34] and low vitamin D levels[35]. Diminished CD4 cell counts in HIV infection is an additional risk determinant[36]. Patients with a long stay in healthcare facilities are also at risk, probably because of excessive exposure to C. difficile via contact with those who are colonized[37]. Finally, the consumption of gastric acid suppressants[38] and certain nonsteroidal anti-inflammatory agents have been linked to CDIs[39, 40].

Consequently, many countries have endeavored to launch protocols and guidelines to constrain CDIs in the acute care setting. These protocols included environmental cleaning, contact precautions, case detection, and antibiotic stewardship[41, 42].

2. Community-associated CDIs (CA-CDIs)

According to the 2018 guidelines by the Society for Healthcare Epidemiology of America, CDIs are considered CA-CDIs if the patient had diarrhea onset in the community or within 48 h after hospitalization and had not been discharged from a healthcare facility in the previous three months[43].

Although formerly categorized as exclusively nosocomial, growing evidence shows that person-to-person transmission causes only <25% of CDIs, advocating the hypothesis that food might be a source of spore ingestion in humans[44].

A study proposed that C. difficile exists in various animals including livestock (cows, pigs, sheep, goats, and chickens), domestic animals (dogs and cats), and horses[45]. Furthermore, C. difficile has been recovered from retail meat products (beef, pork, and turkey), seafood (salmon and shrimp), and vegetables (lettuce, ginger, onions, carrots, potatoes, and spinach). Spores can be propagated among humans and food via the airborne route, direct contact, rodents, birds, arthropods, or fecal contamination during slaughtering[46].

Interestingly, CA-CDIs exhibit a predilection to younger people, with a substantial percentage of them (36%) reporting no antibiotic intake three months before diagnosis[47].

Khanna et al.[48] reported that patients with CA-CDIs were younger than those with HA-CDIs (median age of 50 versus 72). Other studies have revealed 21-38% of patients with CA-CDIs without antibiotic intake compared with 6-20.3% in those with HA-CDIs[49, 50].

Additionally, patients with CA-CDIs have lower mortality rates than those with HA-CDIs[24, 51]. Contrarily, a retrospective study from France revealed that CA-CDIs were weakly associated with more severe disease than HA-CDIs; however, no difference in mortality was found[52].

3. Epidemic Ribotypes

Different typing approaches have been used to study the epidemiology of CDIs. While serotyping was used previously, polymerase chain reaction (PCR) ribotyping is now considered the gold standard typing method. Of note, C. difficile ribotypes display diversity in their regional prevalence and epidemic potential. The most commonly identified ribotypes are depicted in Table 1[53].

Table 1: Characteristic features of common epidemic ribotypes of C. difficile[53]

4. Coronavirus Disease-2019 and CDIs

Efforts have been ongoing to assess CDI trends during the coronavirus disease-2019 pandemic. Published data so far revealed no rise in the rate of CDIs despite the upsurge in antibiotic consumption throughout the pandemic[54, 55]. Some centers have even reported a decline in HA-CDIs[56, 57].

A group of factors have plausibly contributed to such a decline. Among them has been the commitment to infection control approaches, such as higher compliance to hand hygiene and isolation precautions[57, 58].

Diagnosis

An accurate and timely diagnosis of CDIs necessitates both clinical manifestations and a positive laboratory test[59]. A fundamental clinical manifestation is diarrhea, which is defined as loose stools plus a frequency of ≥3 stools over 24 or fewer hours[60]. Severe ileus, where diarrhea comes to a halt, leukocytosis, and high creatinine are significant and should draw ample attention[61].

When a patient suffers from loose stools and has other risk factors for CDIs in the absence of another possibility, e.g., diarrheagenic agents, a fecal specimen should be procured for laboratory testing to assess the possibility of CDIs[59]. At present, no single stool test represents a reference standard for CDI diagnoses[62].

However, historically, the laboratory gold standard for diagnosing CDI was toxigenic culture (TC)[63]. This entailed the culture of C. difficile from the stool and then testing the retrieved isolates to demonstrate their ability to produce toxins. Although TC exhibited >95% sensitivity, its utility has been hindered by the lengthy turnaround duration (3-5 days), rendering it inconvenient for routine diagnosis. In addition, TC alone often yields false-positive results because of nontoxigenic strains[64]. Owing to these limitations, TC is traditionally employed as a reference method rather than a diagnostic tool.

Before 2009, laboratory identification of C. difficile was performed by a two-step process through the detection of glutamate dehydrogenase (GDH) by enzyme immunoassay (EIA) followed by EIA for toxins A and B. While GDH EIAs have a high sensitivity reaching >90%, they cannot distinguish active infection from asymptomatic colonization, resulting in a relatively low specificity[59, 65]. To account for this limitation, positive GDH EIA can be a screening tool to be ensued by toxin A/B EIA, which has a lower sensitivity (51-63%) but higher specificity (91-100%)[59]. In combination, GDH EIA screening ensued by toxin A/B EIA permits a sensitive, specific, and practical method for CDI diagnosis.

Subsequently, in 2009, the US FDA approved the first nucleic acid amplification test (NAAT) for C. difficile[59, 63]. The amplification of C. difficile DNA is carried out via PCR, which detects the genes encoding A and B toxins; however, it cannot distinguish between pathogen presence versus toxin production. Although NAAT cost is higher than those of other approaches, it has been widely allocated as the preferred diagnostic tool owing to its high sensitivity (Table 2), rapid turnaround time, and single-step strategy[59].

Table 2: Performance of the available tests for the diagnosis of C. difficile infections[59, 64, 66]

After many centers accredited NAAT as the sole method of diagnosing CDIs, US hospitals began to witness remarkable uptrends in CDIs, with a concomitant uptick in the consumption of anticlostridial therapeutics. Retrospective studies comparing the incidence of CDIs before and after NAAT implementation demonstrated a >50% leap in HA-CDIs[67]. Further evaluation of NAAT elucidated that despite being highly sensitive, it lacks sufficient specificity to distinguish colonization from active infection. Hence, the use of NAAT alone carries the potential of overdiagnosing CDIs by detecting asymptomatic carriers. Furthermore, NAAT can remain positive in >50% of patients after completing their treatment, compounding the challenge of interpreting results in those with prior infection[68].

Approaches to Mitigate Overdiagnosis

Several key strategies can be employed to improve the appropriate testing of CDIs and distinguish colonization from clinical disease. One of the fundamental interventions is diagnostic stewardship. Patients should not be tested for C. difficile unless they present with clinical features of actual infection, such as an unexplainable and new-onset watery diarrhea (≥3 loose stools in 24 h) in the absence of other causes, e.g., laxative intake or antibacterial/chemotherapy-related diarrhea[69].

At present, the recommended diagnostic algorithm comprises a 2-3-step approach to augment sensitivity and specificity. Acceptable approaches include either GDH detection or NAAT, followed by toxin A/B EIA. Patients are eligible for treatment only if toxin EIA yields a positive result, inferring that they have clinical CDIs rather than mere colonization. In general, employing NAAT alone is not recommended owing to its low positive predictive value[59].

Further, repeating the test for CDIs should be avoided within a week of a negative test result because of the low diagnostic yield. Likewise, patients successfully treated should not undergo a test of cure because the tests remain positive in >60% of patients[47].

In the same context, antimicrobial stewardship is a crucial approach to combat CDIs. To reduce overall antibiotic exposure, prescribers are encouraged to specify the duration of antibacterial therapy. Alternatively, antibacterial therapeutics prescribed for >7 days should be assessed by the antimicrobial stewardship program to evaluate the appropriateness and value of continued therapy. Furthermore, according to the regional epidemiology and C. difficile prevalence, restricting the use of clindamycin, cephalosporins, carbapenems, and fluoroquinolones can be advised to ensure the judicious usage of broad-spectrum antibacterial agents[69].

Specific Prophylaxis Against CDIs

Although a Cochrane review has found that probiotics are effective in preventing CDIs[70], such a result may be insufficient for routine clinical implementation. This in part is due to the use of different probiotic formulations in the trials included in the Cochrane review. In addition, probiotics may delay microbiome reconstitution following antibiotic therapy in addition to concerns about the associated adverse effects[71]. Another prophylactic approach against CDIs is antibiotic administration, where retrospective studies have revealed a 5-30% decrease in CDI occurrence with oral vancomycin prophylaxis in patients on broad-spectrum antibiotics[72, 73]. Another regimen is the co-administration of ribaxamase (a poorly absorbed β-lactamase) when administering broad-spectrum antibiotics. Of note, a phase 2b trial revealed a 2.4% risk reduction of CDI occurrence when administering ribaxamase along with ceftriaxone[74].

Treatment of CDIs

Based on the 2021 guidelines of the Infectious Diseases Society of America, CDIs are best treated with fidaxomicin, while vancomycin is an alternative[75].

In 2021, the European Society of Clinical Microbiology and Infectious Diseases updated its 2014 guidelines for CDI management. An important modification stated that metronidazole is no longer recommended for CDIs if fidaxomicin or vancomycin is available and that fidaxomicin is the preferred agent for both initial CDIs and first recurrence of CDIs[71].

Based on these guidelines, severe CDIs are marked by one of the following criteria at the time of presentation: fever (>38.5 °C), marked leukocytosis (>15×109/L), and high creatinine (>50% above the baseline). Meanwhile, severe complicated CDIs (or fulminant CDIs) occur when one of the following signs is present that needs to be attributed to CDIs: hypotension, septic shock, high lactate levels, ileus, toxic megacolon, perforated bowel, or any fulminant disease course. On the contrary, refractory CDIs are irresponsive CDIs after 3-5 days of the recommended therapy[71].

A summary of the treatment recommendations is depicted in Figure 1. In addition to these interventions, the following measures were recommended in the 2014 treatment guideline[60, 76]:

Figure 1: Suggested treatment algorithm for C. difficile infections[71]
*Vancomycin taper and pulse: 125 mg qid for 14 days, followed by 125 mg bid for 7 days, then 125 mg qd for 7 days, then 125 mg q48h for 7 days, then 125 mg q72h for 7 days
CDIs: C. difficile infections

- Cessation of unnecessary antimicrobials,

- Replacing deficient fluids and electrolytes,

- Avoiding antimotility agents,

- Re-assessment of the use of proton-pump inhibitors.

1. Fidaxomicin and Vancomycin as the Standard of Care

A phase three randomized controlled trial compared fidaxomicin with vancomycin and reported no difference in the cure rates; however, the study revealed a 9.9% reduction in the recurrence risk at four weeks in favor of fidaxomicin (15% vs. 25% reduction)[77].

As it possesses the narrowest activity spectrum, fidaxomicin is less likely to disrupt the gut microbiome[78], which is a merit in the case of CDIs. Of note, fidaxomicin costs much higher than vancomycin, which represents an obstacle against a widespread prescription. Nonetheless, the decreased recurrences and subsequent decline in rehospitalizations partially balance this high cost and may eventually result in cost-effectiveness[79, 80].

2. Bezlotoxumab

This is a monoclonal antibody against toxin B of C. difficile. The addition of bezlotoxumab to the standard of care antibiotics produced the same cure rates but resulted in 10% reduced risk of recurrences in MODIFY-I and II trials[81]. In these trials, 48% of the patients were receiving vancomycin, whereas only 4% were taking fidaxomicin. Thus, the value of combining bezlotoxumab with fidaxomicin is uncertain.

For patients with a history of congestive heart failure, vigilance should be applied when prescribing bezlotoxumab. In these patients, heart failure was more commonly reported compared with the placebo group, and more deaths occurred. Hence, in these vulnerable patients, bezlotoxumab must be reserved for use only when the drug benefits outbalance the risks[71].

3. Intravenous Metronidazole

A retrospective analysis performed on 138 patients admitted to the ICU revealed that adding intravenous metronidazole to oral vancomycin was not associated with a better clinical result in severe nonfulminant CDIs[82]. Contrarily, a retrospective study in patients admitted to the ICU reinforced the suggestion that adding intravenous metronidazole to oral CDI therapy in patients in a critical condition might be of benefit[83]. Such conflicting results necessitate weighing the benefits against the risks for each case.

4. Intravenous Tigecycline

A retrospective study performed in a single center compared intravenous tigecycline as monotherapy with oral vancomycin plus intravenous metronidazole in severe CDIs. The study demonstrated a higher cure rate (76%) with tigecycline than with vancomycin plus metronidazole (53%)[84]. On the contrary, another study revealed that combining tigecycline with vancomycin had no added benefit[85]. A recent review based on retrospective observational research deduced that tigecycline can be a potential therapeutic against severe CDIs[2].

5. Other Agents

Rifaximin is an oral antibiotic of the same class as rifampin. In the management of mild and moderate CDIs, it has demonstrated noninferiority to available therapies, with the advantages of being poorly absorbed from the intestine, having slight adverse reactions, and a surprisingly minimal effect on the colonic microbiome. Nonetheless, clinical studies have displayed a resistance rate of 29-48.9%[86].

In addition, ridinilazole is potentially an antibiotic of interest for treating CDIs. In a phase 2 randomized controlled trial, it proved its superiority over vancomycin in attaining sustained cure[87].

Interestingly, some drugs have been experimented in mice and yielded a noticeable anticlostridial activity, e.g., auranofin, an FDA-approved antirheumatic agent, exhibited a reduction in C. difficile sporulation and toxin secretion in mice[88]. Other therapeutics that were experimented in mice include doxapram (a breathing stimulant), amoxapine (an antidepressant), and trifluoperazine (an antipsychotic). Nonetheless, the mechanism by which they achieved anticlostridial action is obscure[89].

Meanwhile, antivirulence therapy has drawn remarkable interest in combating antibiotic-resistant infections. Such agents are neither bacteriostatic nor bactericidal; they primarily attenuate bacterial virulence rather than its growth[90, 91]. Thus, there is a low tendency to provoke pathogen resistance, in addition to the advantage of minimally affecting the gut microflora[92, 93]. Baicalin, a flavone glycoside in the herb Scutellaria baicalensis, has been investigated in vitro against C. difficile with promising results[94].

On the other hand, the development of surotomycin and cadazolid was terminated after failing to prove noninferiority in randomized controlled trials[95, 96].

6. Fecal Microbiota Transplantation

Fecal microbiota transplantation (FMT) has gained a foothold in treating multiple recurrent CDIs. An observational cohort study reported an 87% cure rate in one month for severe complicated refractory CDIs[97]. Considering the high mortality associated with the surgical treatment of CDIs and the fact that some patients are unfit for surgery, FMT can play an important role in patients with refractory severe complicated CDIs for whom surgery is unfeasible, provided that a cautious risk assessment is made on a case-by-case basis. The expert team should also discuss intravenous antibiotics pre- and post-FMT, depending on the patients underlying condition and follow-up parameters[71].

Conclusion

Clostridioides difficile infections still represent costly and potentially life-threatening infections, especially in older vulnerable patients. Further, CDIs are now observable in community-dwelling younger, healthier populations.

To attain the dual target of reducing infection rates and lowering the risk of resistance, it is pivotal to distinguish colonization from clinical CDIs through judicious testing algorithms. Meanwhile, experimenting with novel treatment modalities directed against CDIs has become a worldwide priority owing to the high disease incidence, recurrence, mortality, and scarce therapeutic options. Agents that are less amenable to invoke resistance and/or alter gut microflora represent plausible approaches and can provide a novel prospect in impeding CDIs.

In summary, CDIs still pose a significant menace, and a multifaceted approach is warranted to curb this infection. Both diagnostic and antimicrobial stewardship must be accompanied by state-of-the-art education on appropriate testing and treatment modalities.

Ethics

Peer-review: Externally peer-reviewed.

Financial Disclosure: The author declared that this study received no financial support.

References

1Lanzas C, Jara M, Tucker R, Curtis S; CDC Modeling Infectious Diseases in Healthcare Program (MinD). A review of epidemiological models of Clostridioides difficile transmission and control (2009-2021). Anaerobe. 2022;74:102541.
2Kechagias KS, Chorepsima S, Triarides NA, Falagas ME. Tigecycline for the treatment of patients with Clostridium difficile infection: an update of the clinical evidence. Eur J Clin Microbiol Infect Dis. 2020;39:1053-8.
3Zhang S, Palazuelos-Munoz S, Balsells EM, Nair H, Chit A, Kyaw MH. Cost of hospital management of Clostridium difficile infection in United States-a meta-analysis and modelling study. BMC Infect Dis. 2016;16:447.
4Scott RD. The Direct Medical Costs of Healthcare-Associated Infections in U.S. Hospitals and the Benefits of Prevention. Last Accessed Date: October 9, 2022. Available from: https://www.cdc.gov/hai/pdfs/hai/scott_costpaper. pdf
5Loo VG, Bourgault AM, Poirier L, Lamothe F, Michaud S, Turgeon N, Toye B, Beaudoin A, Frost EH, Gilca R, Brassard P, Dendukuri N, Béliveau C, Oughton M, Brukner I, Dascal A. Host and pathogen factors for Clostridium difficile infection and colonization. N Engl J Med. 2011;365:1693-703.
6Gerding DN, Johnson S, Rupnik M, Aktories K. Clostridium difficile binary toxin CDT: mechanism, epidemiology, and potential clinical importance. Gut Microbes. 2014;5:15-27.
7Keel MK, Songer JG. The comparative pathology of Clostridium difficile-associated disease. Vet Pathol. 2006;43:225-40.
8Sunenshine RH, McDonald LC. Clostridium difficile-associated disease: new challenges from an established pathogen. Cleve Clin J Med. 2006;73:187-97.
9Hookman P, Barkin JS. Clostridium difficile-associated infection, diarrhea and colitis. World J Gastroenterol. 2009;15:1554-80.
10Burns DA, Heap JT, Minton NP. Clostridium difficile spore germination: an update. Res Microbiol. 2010;161:730-4.
11Gerding DN, Young VB and Donskey CJ. Clostridioides difficile (formerly Clostridium difficile) infection. In: Bennett JE, Dolin R, Blaser MJ (eds). Mandell, Douglas, and Bennett’s principles and practice of infectious diseases. Philadelphia, PA: Elsevier, 2020.
12De Roo AC, Regenbogen SE. Clostridium difficile Infection: An Epidemiology Update. Clin Colon Rectal Surg. 2020;33:49-57.
13Durovic A, Widmer AF, Tschudin-Sutter S. New insights into transmission of Clostridium difficile infection-narrative review. Clin Microbiol Infect. 2018;24:483-92.
14Kelly CP, LaMont JT. Clostridium difficile--more difficult than ever. N Engl J Med. 2008;359:1932-40.
15Cruz MP. Fidaxomicin (Dificid), a novel oral macrocyclic antibacterial agent for the treatment of Clostridium difficile-associated diarrhea in adults. P T. 2012;37:278-81.
16AbdelKhalek A, Mohammad H, Mayhoub AS, Seleem MN. Screening for potent and selective anticlostridial leads among FDA-approved drugs. J Antibiot (Tokyo). 2020;73:392-409.
17CDC, Antibiotic Resistance Threats in the United States, 2019, U.S. Department of Health and Human Services, CDC, Atlanta, GA, 2019. Last Accessed Date: October 11, 2022. Available from: https://www.cdc.gov/drugresistance/biggest-threats.html
18Roberts K, Smith CF, Snelling AM, Kerr KG, Banfield KR, Sleigh PA, Beggs CB. Aerial dissemination of Clostridium difficile spores. BMC Infect Dis. 2008;8:7.
19McDonald LC, Coignard B, Dubberke E, Song X, Horan T, Kutty PK; Ad Hoc Clostridium difficile Surveillance Working Group. Recommendations for surveillance of Clostridium difficile-associated disease. Infect Control Hosp Epidemiol. 2007;28:140-5.
20Lessa FC, Mu Y, Bamberg WM, Beldavs ZG, Dumyati GK, Dunn JR, Farley MM, Holzbauer SM, Meek JI, Phipps EC, Wilson LE, Winston LG, Cohen JA, Limbago BM, Fridkin SK, Gerding DN, McDonald LC. Burden of Clostridium difficile infection in the United States. N Engl J Med. 2015;372:825-34.
21Caupenne A, Ingrand P, Ingrand I, Forestier E, Roubaud-Baudron C, Gavazzi G, Paccalin M. Acute Clostridioides difficile Infection in Hospitalized Persons Aged 75 and Older: 30-Day Prognosis and Risk Factors for Mortality. J Am Med Dir Assoc. 2020;21:110-4.
22Tariq R, Law CCY, Khanna S, Murthy S, McCurdy JD. The Impact of Clostridium difficile Infection on Mortality in Patients With Inflammatory Bowel Disease: A Systematic Review and Meta-Analysis. J Clin Gastroenterol. 2019;53:127-33.
23Magill SS, O’Leary E, Janelle SJ, Thompson DL, Dumyati G, Nadle J, Wilson LE, Kainer MA, Lynfield R, Greissman S, Ray SM, Beldavs Z, Gross C, Bamberg W, Sievers M, Concannon C, Buhr N, Warnke L, Maloney M, Ocampo V, Brooks J, Oyewumi T, Sharmin S, Richards K, Rainbow J, Samper M, Hancock EB, Leaptrot D, Scalise E, Badrun F, Phelps R, Edwards JR; Emerging Infections Program Hospital Prevalence Survey Team. Changes in Prevalence of Health Care-Associated Infections in U.S. Hospitals. N Engl J Med. 2018;379:1732-44.
24Guh AY, Mu Y, Winston LG, Johnston H, Olson D, Farley MM, Wilson LE, Holzbauer SM, Phipps EC, Dumyati GK, Beldavs ZG, Kainer MA, Karlsson M, Gerding DN, McDonald LC; Emerging Infections Program Clostridioides difficile Infection Working Group. Trends in U.S. Burden of Clostridioides difficile Infection and Outcomes. N Engl J Med. 2020;382:1320-30.
25Miller BA, Chen LF, Sexton DJ, Anderson DJ. Comparison of the burdens of hospital-onset, healthcare facility-associated Clostridium difficile Infection and of healthcare-associated infection due to methicillin-resistant Staphylococcus aureus in community hospitals. Infect Control Hosp Epidemiol. 2011;32:387-90.
26Mullane K. Fidaxomicin in Clostridium difficile infection: latest evidence and clinical guidance. Ther Adv Chronic Dis. 2014;5:69-84.
27Brajerova M, Zikova J, Krutova M. Clostridioides difficile epidemiology in the Middle and the Far East. Anaerobe. 2022;74:102542.
28Curcio D, Cané A, Fernández FA, Correa J. Clostridium difficile-associated Diarrhea in Developing Countries: A Systematic Review and Meta-Analysis. Infect Dis Ther. 2019;8:87-103.
29El-Sokkary RH, Gerges MA, Sharaf HE, Fattah LA, Amer FA, Tash RM, Aamir R, Sherif AM, Hegab MS, Elashkar SSA, Khalil AM, Al-Akad GM, Salah TS. Clostridium difficile occurrence, toxin profile and antibiotic susceptibility: An Egyptian center experience. Egypt J Med Microbiol. 2017;26:31-6.
30Elgendy SG, Aly SA, Fathy R, Deaf EAE, Abu Faddan NH, Abdel Hameed MR. Clinical and microbial characterization of toxigenic Clostridium difficile isolated from antibiotic associated diarrhea in Egypt. Iran J Microbiol. 2020;12:296-304.
31Roldan GA, Cui AX, Pollock NR. Assessing the burden of Clostridium difficile infection in low-and middle-income countries. J Clin Microbiol. 2018;56:e01747-17.
32Balsells E, Shi T, Leese C, Lyell I, Burrows J, Wiuff C, Campbell H, Kyaw MH, Nair H. Global burden of Clostridium difficile infections: a systematic review and meta-analysis. J Glob Health. 2019;9:010407.
33Slimings C, Riley TV. Antibiotics and hospital-acquired Clostridium difficile infection: update of systematic review and meta-analysis. J Antimicrob Chemother. 2014;69:881-91.
34Nguyen GC, Kaplan GG, Harris ML, Brant SR. A national survey of the prevalence and impact of Clostridium difficile infection among hospitalized inflammatory bowel disease patients. Am J Gastroenterol. 2008;103:1443-50.
35Furuya-Kanamori L, Wangdi K, Yakob L, McKenzie SJ, Doi SAR, Clark J, Paterson DL, Riley TV, Clements ACA. 25-Hydroxyvitamin D Concentrations and Clostridium difficile Infection: A Meta-Analysis. JPEN J Parenter Enteral Nutr. 2017;41:890-5.
36Di Bella S, Friedrich AW, García-Almodóvar E, Gallone MS, Taglietti F, Topino S, Galati V, Johnson E, D’Arezzo S, Petrosillo N. Clostridium difficile infection among hospitalized HIV-infected individuals: epidemiology and risk factors: results from a case-control study (2002-2013). BMC Infect Dis. 2015;15:194.
37Ziakas PD, Zacharioudakis IM, Zervou FN, Grigoras C, Pliakos EE, Mylonakis E. Asymptomatic carriers of toxigenic C. difficile in long-term care facilities: a meta-analysis of prevalence and risk factors. PLoS One. 2015;10:e0117195.
38Oshima T, Wu L, Li M, Fukui H, Watari J, Miwa H. Magnitude and direction of the association between Clostridium difficile infection and proton pump inhibitors in adults and pediatric patients: a systematic review and meta-analysis. J Gastroenterol. 2018;53:84-94.
39Permpalung N, Upala S, Sanguankeo A, Sornprom S. Association between nonsteroidal anti-inflammatory drugs and Clostridium difficile-associated diarrhea: A systematic review and meta-analysis. Can J Gastroenterol Hepatol. 2015:16960.
40Novack L, Kogan S, Gimpelevich L, Howell M, Borer A, Kelly CP, Leffler DA, Novack V. Acid suppression therapy does not predispose to Clostridium difficile infection: the case of the potential bias. PLoS One. 2014;9:e110790.
41Centers for Disease Control and Prevention. 2014 National and state healthcare-associated infections progress report. Last Accessed Date: October 14, 2022. Available from: https://www.cdc.gov/hai/data/archive/2014-progress-report.html
42Balsells E, Filipescu T, Kyaw MH, Wiuff C, Campbell H, Nair H. Infection prevention and control of Clostridium difficile: a global review of guidelines, strategies, and recommendations. J Glob Health. 2016;6:020410.
43Fu Y, Luo Y, Grinspan AM. Epidemiology of community-acquired and recurrent Clostridioides difficile infection. Therap Adv Gastroenterol. 2021;14:17562848211016248.
44Rodriguez-Palacios A, Mo KQ, Shah BU, Msuya J, Bijedic N, Deshpande A, Ilic S. Global and Historical Distribution of Clostridioides difficile in the Human Diet (1981-2019): Systematic Review and Meta-Analysis of 21886 Samples Reveal Sources of Heterogeneity, High-Risk Foods, and Unexpected Higher Prevalence Toward the Tropic. Front Med (Lausanne). 2020;7:9.
45Ofori E, Ramai D, Dhawan M, Mustafa F, Gasperino J, Reddy M. Community-acquired Clostridium difficile: epidemiology, ribotype, risk factors, hospital and intensive care unit outcomes, and current and emerging therapies. J Hosp Infect. 2018;99:436-42.
46Knight DR, Riley TV. Genomic delineation of zoonotic origins of Clostridium difficile. Front Public Health. 2019;7:164.
47McDonald LC, Gerding DN, Johnson S, Bakken JS, Carroll KC, Coffin SE, Dubberke ER, Garey KW, Gould CV, Kelly C, Loo V, Shaklee Sammons J, Sandora TJ, Wilcox MH. 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;66:1-48.
48Khanna S, Pardi DS, Aronson SL, Kammer PP, Orenstein R, St Sauver JL, Harmsen WS, Zinsmeister AR. The epidemiology of community-acquired Clostridium difficile infection: a population-based study. Am J Gastroenterol. 2012;107:89-95.
49Chitnis AS, Holzbauer SM, Belflower RM, Winston LG, Bamberg WM, Lyons C, Farley MM, Dumyati GK, Wilson LE, Beldavs ZG, Dunn JR, Gould LH, MacCannell DR, Gerding DN, McDonald LC, Lessa FC. Epidemiology of community-associated Clostridium difficile infection, 2009 through 2011. JAMA Intern Med. 2013;173:1359-67.
50Fawley WN, Davies KA, Morris T, Parnell P, Howe R, Wilcox MH; Clostridium difficile Ribotyping Network (CDRN) Working Group. Enhanced surveillance of Clostridium difficile infection occurring outside hospital, England, 2011 to 2013. Euro Surveill. 2016.
51Alfayyadh M, Collins DA, Tempone S, McCann R, Armstrong PK, Riley TV, Cook A. Recurrence of Clostridium difficile infection in the Western Australian population. Epidemiol Infect. 2019;147:153.
52Ogielska M, Lanotte P, Le Brun C, Valentin AS, Garot D, Tellier AC, Halimi JM, Colombat P, Guilleminault L, Lioger B, Vegas H, De Toffol B, Constans T, Bernard L. Emergence of community-acquired Clostridium difficile infection: the experience of a French hospital and review of the literature. Int J Infect Dis. 2015;37:36-41.
53Martínez-Meléndez A, Morfin-Otero R, Villarreal-Treviño L, Baines SD, Camacho-Ortíz A, Garza-González E. Molecular epidemiology of predominant and emerging Clostridioides difficile ribotypes. J Microbiol Methods. 2020;175:105974.
54Laszkowska M, Kim J, Faye AS, Joelson AM, Ingram M, Truong H, Silver ER, May B, Greendyke WG, Zucker J, Lebwohl B, Hur C, Freedberg DE. Prevalence of Clostridioides difficile and Other Gastrointestinal Pathogens in Patients with COVID-19. Dig Dis Sci. 2021;66:4398-405.
55Luo Y, Grinspan LT, Fu Y, Adams-Sommer V, Willey DK, Patel G, Grinspan AM. Hospital-onset Clostridioides difficile infections during the COVID-19 pandemic. Infect Control Hosp Epidemiol. 2021;42:1165-6.
56Ponce-Alonso M, Sáez de la Fuente J, Rincón-Carlavilla A, Moreno-Nunez P, Martínez-García L, Escudero-Sánchez R, Pintor R, García-Fernández S, Cobo J. Impact of the coronavirus disease 2019 (COVID-19) pandemic on nosocomial Clostridioides difficile infection. Infect Control Hosp Epidemiol. 2021;42:406-10.
57Hazel K, Skally M, Glynn E, Foley M, Burns K, O’Toole A, Boland K, Fitzpatrick F. The other ‘C’: Hospital-acquired Clostridioides difficile infection during the coronavirus disease 2019 (COVID-19) pandemic. Infect Control Hosp Epidemiol. 2022;43:540-1.
58Granata G, Bartoloni A, Codeluppi M, Contadini I, Cristini F, Fantoni M, Ferraresi A, Fornabaio C, Grasselli S, Lagi F, Masucci L, Puoti M, Raimondi A, Taddei E, Trapani FF, Viale P, Johnson S, Petrosillo N, On Behalf Of The CloVid Study Group. The Burden of Clostridioides Difficile Infection during the COVID-19 Pandemic: A Retrospective Case-Control Study in Italian Hospitals (CloVid). J Clin Med. 2020;9:3855.
59Crobach MJ, Planche T, Eckert C, Barbut F, Terveer EM, Dekkers OM, Wilcox MH, Kuijper EJ. European Society of Clinical Microbiology and Infectious Diseases: update of the diagnostic guidance document for Clostridium difficile infection. Clin Microbiol Infect. 2016;22(Suppl 4):63-81.
60Debast SB, Bauer MP, Kuijper EJ; European Society of Clinical Microbiology and Infectious Diseases. European Society of Clinical Microbiology and Infectious Diseases: update of the treatment guidance document for Clostridium difficile infection. Clin Microbiol Infect. 2014;20(Suppl 2):1-26.
61Gerding DN, File Jr TM, McDonald LC. Diagnosis and treatment of Clostridium difficile infection (CDI). Infect Dis Clin Pract (Baltim Md). 2016;24:3.
62Peng Z, Ling L, Stratton CW, Li C, Polage CR, Wu B, Tang YW. Advances in the diagnosis and treatment of Clostridium difficile infections. Emerg Microbes Infect. 2018;7:15.
63Burnham CA, Carroll KC. Diagnosis of Clostridium difficile infection: an ongoing conundrum for clinicians and for clinical laboratories. Clin Microbiol Rev. 2013;26:604-30.
64Bartlett JG. Detection of Clostridium difficile infection. Infect Control Hosp Epidemiol. 2010;31(Suppl 1):35-7.
65Carroll KC, Mizusawa M. Laboratory Tests for the Diagnosis of Clostridium difficile. Clin Colon Rectal Surg. 2020;33:73-81.
66Brecher SM, Novak-Weekley SM, Nagy E. Laboratory diagnosis of Clostridium difficile infections: there is light at the end of the colon. Clin Infect Dis. 2013;57:1175-81.
67Moehring RW, Lofgren ET, Anderson DJ. Impact of change to molecular testing for Clostridium difficile infection on healthcare facility-associated incidence rates. Infect Control Hosp Epidemiol. 2013;34:1055-61.
68Abujamel T, Cadnum JL, Jury LA, Sunkesula VC, Kundrapu S, Jump RL, Stintzi AC, Donskey CJ. Defining the vulnerable period for re-establishment of Clostridium difficile colonization after treatment of C. difficile infection with oral vancomycin or metronidazole. PLoS One. 2013;8:e76269.
69Lee HS, Plechot K, Gohil S, Le J. Clostridium difficile: Diagnosis and the Consequence of Over Diagnosis. Infect Dis Ther. 2021;10:687-97.
70Goldenberg JZ, Yap C, Lytvyn L, Lo CK, Beardsley J, Mertz D, Johnston BC. Probiotics for the prevention of Clostridium difficile-associated diarrhea in adults and children. Cochrane Database Syst Rev. 2017;12:CD006095.
71van Prehn J, Reigadas E, Vogelzang EH, Bouza E, Hristea A, Guery B, Krutova M, Norén T, Allerberger F, Coia JE, Goorhuis A, van Rossen TM, Ooijevaar RE, Burns K, Scharvik Olesen BR, Tschudin-Sutter S, Wilcox MH, Vehreschild MJGT, Fitzpatrick F, Kuijper EJ; Guideline Committee of the European Study Group on Clostridioides difficile. European Society of Clinical Microbiology and Infectious Diseases: 2021 update on the treatment guidance document for Clostridioides difficile infection in adults. Clin Microbiol Infect. 2021;27(Suppl 2):1-21.
72Caroff DA, Menchaca JT, Zhang Z, Rhee C, Calderwood MS, Kubiak DW, Yokoe DS, Klompas M. Oral vancomycin prophylaxis during systemic antibiotic exposure to prevent Clostridiodes difficile infection relapses. Infect Control Hosp Epidemiol. 2019;40:662-7.
73Ganetsky A, Han JH, Hughes ME, Babushok DV, Frey NV, Gill SI, Hexner EO, Loren AW, Luger SM, Mangan JK, Martin ME, Smith J, Freyer CW, Gilmar C, Schuster M, Stadtmauer EA, Porter DL. Oral Vancomycin Prophylaxis Is Highly Effective in Preventing Clostridium difficile Infection in Allogeneic Hematopoietic Cell Transplant Recipients. Clin Infect Dis. 2019;68:2003-9.
74Kokai-Kun JF, Roberts T, Coughlin O, Le C, Whalen H, Stevenson R, Wacher VJ, Sliman J. Use of ribaxamase (SYN-004), a β-lactamase, to prevent Clostridium difficile infection in β-lactam-treated patients: a double-blind, phase 2b, randomised placebo-controlled trial. Lancet Infect Dis. 2019;19:487-96.
75Johnson S, Lavergne V, Skinner AM, Gonzales-Luna AJ, Garey KW, Kelly CP, Wilcox MH. 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;73:1029-44.
76Surawicz CM, Brandt LJ, Binion DG, Ananthakrishnan AN, Curry SR, Gilligan PH, McFarland LV, Mellow M, Zuckerbraun BS. Guidelines for diagnosis, treatment, and prevention of Clostridium difficile infections. Am J Gastroenterol. l 2013;108:478-98.
77Louie TJ, Miller MA, Mullane KM, Weiss K, Lentnek A, Golan Y, Gorbach S, Sears P, Shue YK; OPT-80-003 Clinical Study Group. Fidaxomicin versus vancomycin for Clostridium difficile infection. N Engl J Med. 2011;364:422-31.
78Tannock GW, Munro K, Taylor C, Lawley B, Young W, Byrne B, Emery J, Louie T. A new macrocyclic antibiotic, fidaxomicin (OPT-80), causes less alteration to the bowel microbiota of Clostridium difficile-infected patients than does vancomycin. Microbiology. 2010;156:3354-9.
79Marković V, Kostić M, Iličković I, Janković SM. Cost-Effectiveness Comparison of Fidaxomicin and Vancomycin for Treatment of Clostridium difficile Infection: A Markov Model Based on Data from a South West Balkan Country in Socioeconomic Transition. Value Health Reg Issues. 2014;4:87-94.
80Chen J, Gong CL, Hitchcock MM, Holubar M, Deresinski S, Hay JW. Cost-effectiveness of bezlotoxumab and fidaxomicin for initial Clostridioides difficile infection. Clin Microbiol Infect. 2021;27:1448-54.
81Wilcox MH, Gerding DN, Poxton IR, Kelly C, Nathan R, Birch T, Cornely OA, Rahav G, Bouza E, Lee C, Jenkin G, Jensen W, Kim YS, Yoshida J, Gabryelski L, Pedley A, Eves K, Tipping R, Guris D, Kartsonis N, Dorr MB; MODIFY I and MODIFY II Investigators. Bezlotoxumab for Prevention of Recurrent Clostridium difficile Infection. N Engl J Med. 2017;376:305-17.
82Vega AD, Heil EL, Blackman AL, Banoub M, Kristie Johnson J, Leekha S, Claeys KC. Evaluation of Addition of Intravenous Metronidazole to Oral Vancomycin Therapy in Critically Ill Patients with Non-Fulminant Severe Clostridioides difficile Infection. Pharmacotherapy. 2020;40:398-407.
83Rokas KE, Johnson JW, Beardsley JR, Ohl CA, Luther VP, Williamson JC. The Addition of Intravenous Metronidazole to Oral Vancomycin is Associated With Improved Mortality in Critically Ill Patients With Clostridium difficile Infection. Clin Infect Dis. 2015;61:934-41.
84Gergely Szabo B, Kadar B, Szidonia Lenart K, Dezsenyi B, Kunovszki P, Fried K, Kamotsay K, Nikolova R, Prinz G. Use of intravenous tigecycline in patients with severe Clostridium difficile infection: a retrospective observational cohort study. Clin Microbiol Infect. 2016;22:990-5.
85Manea E, Sojo-Dorado J, Jipa RE, Benea SN, Rodríguez-Baño J, Hristea A. The role of tigecycline in the management of Clostridium difficile infection: a retrospective cohort study. Clin Microbiol Infect. 2018;24:180-4.
86Ng QX, Loke W, Foo NX, Mo Y, Yeo WS, Soh AYS. A systematic review of the use of rifaximin for Clostridium difficile infections. Anaerobe. 2019;55:35-9.
87Vickers RJ, Tillotson GS, Nathan R, Hazan S, Pullman J, Lucasti C, Deck K, Yacyshyn B, Maliakkal B, Pesant Y, Tejura B, Roblin D, Gerding DN, Wilcox MH; CoDIFy study group. Efficacy and safety of ridinilazole compared with vancomycin for the treatment of Clostridium difficile infection: a phase 2, randomised, double-blind, active-controlled, non-inferiority study. Lancet Infect Dis. 2017;17:735-44.
88Hutton ML, Pehlivanoglu H, Vidor CJ, James ML, Thomson MJ, Lyras D. Repurposing auranofin as a Clostridioides difficile therapeutic. J Antimicrob Chemother. 2020;75:409-17.
89Andersson JA, Peniche AG, Galindo CL, Boonma P, Sha J, Luna RA, Savidge TC, Chopra AK, Dann SM. New Host-Directed Therapeutics for the Treatment of Clostridioides difficile Infection. mBio. 2020;11:e00053-20.
90Rasko DA, Sperandio V. Anti-virulence strategies to combat bacteria-mediated disease. Nat Rev Drug Discov. 2010;9:117-28.
91Khodaverdian V, Pesho M, Truitt B, Bollinger L, Patel P, Nithianantham S, Yu G, Delaney E, Jankowsky E, Shoham M. Discovery of antivirulence agents against methicillin-resistant Staphylococcus aureus. Antimicrob Agents Chemother. 2013;57:3645-52.
92Cegelski L, Marshall GR, Eldridge GR, Hultgren SJ. The biology and future prospects of antivirulence therapies. Nat Rev Microbiol. 2008;6:17-27.
93Mellbye B, Schuster M. The sociomicrobiology of antivirulence drug resistance: a proof of concept. mBio. 2011;2:e00131-11.
94Pellissery AJ, Vinayamohan PG, Venkitanarayanan K. In vitro antivirulence activity of baicalin against Clostridioides difficile. J Med Microbiol. 2020;69:631-9.
95Boix V, Fedorak RN, Mullane KM, Pesant Y, Stoutenburgh U, Jin M, Adedoyin A, Chesnel L, Guris D, Larson KB, Murata Y. Primary Outcomes From a Phase 3, Randomized, Double-Blind, Active-Controlled Trial of Surotomycin in Subjects With Clostridium difficile Infection. Open Forum Infect Dis. 2017;4:ofw275.
96Gerding DN, Cornely OA, Grill S, Kracker H, Marrast AC, Nord CE, Talbot GH, Buitrago M, Gheorghe Diaconescu I, Murta de Oliveira C, Preotescu L, Pullman J, Louie TJ, Wilcox MH. Cadazolid for the treatment of Clostridium difficile infection: results of two double-blind, placebo-controlled, non-inferiority, randomised phase 3 trials. Lancet Infect Dis. 2019;19:265-74.
97Fischer M, Sipe B, Cheng YW, Phelps E, Rogers N, Sagi S, Bohm M, Xu H, Kassam Z. Fecal microbiota transplant in severe and severe-complicated Clostridium difficile: A promising treatment approach. Gut Microbes. 2017;8:289-302.
Article is only available in PDF format. Show PDF
2024 ©️ Galenos Publishing House