Fact-checked by Grok 2 weeks ago

Clostridioides difficile

Clostridioides difficile (formerly known as Clostridium difficile) is a Gram-positive, , spore-forming bacterium that causes Clostridioides difficile infection (CDI), a potentially life-threatening condition characterized by and resulting from the release of toxins that damage the intestinal mucosa. The infection typically arises when antibiotics disrupt the normal , allowing C. difficile spores to germinate, colonize the colon, and produce toxins A (TcdA) and B (TcdB), which inactivate , leading to cytoskeletal disruption, increased , and . CDI is a leading cause of healthcare-associated infections worldwide, with an estimated 500,000 cases annually (as of 2015), contributing to approximately 29,000 deaths each year (as of 2015). The incidence rose significantly in the early , from about 139,000 hospitalized cases in 2000 to over 336,000 in 2009, although it has since declined, driven by the emergence of hypervirulent strains such as BI/NAP1/027 and increasing community-acquired infections, which account for up to 41% of cases in some studies. Globally, the burden is substantial, with higher rates in regions like and , exacerbated by factors such as the due to heightened use. Symptoms of CDI range from mild to severe, including watery (often more than three times daily), abdominal cramps, fever, and loss of appetite, progressing in fulminant cases to pseudomembranous , , or . Transmission occurs primarily through the fecal-oral route, with resilient spores contaminating surfaces in healthcare settings and surviving standard disinfectants, making hand hygiene with soap and water essential over alcohol-based sanitizers. Risk factors prominently include recent exposure (increasing risk up to 10-fold), advanced age (≥65 years), prolonged hospitalization, and immunocompromised states. Diagnosis relies on detecting toxins or toxin genes via nucleic acid amplification tests (NAATs) or enzyme immunoassays for glutamate dehydrogenase (GDH), while treatment involves discontinuing inciting antibiotics and administering targeted therapies like oral vancomycin or fidaxomicin, with fecal microbiota transplantation for recurrent episodes. Prevention strategies emphasize antimicrobial stewardship, contact precautions, thorough environmental cleaning with sporicidal agents, and vigilant infection control in healthcare facilities.

Taxonomy and nomenclature

Taxonomy

Clostridioides difficile is a Gram-positive, strictly , spore-forming classified within the family Peptostreptococcaceae of the order Clostridiales in the phylum . This placement reflects its rod-shaped morphology and ability to produce endospores, characteristics typical of many , though its phylogenetic position distinguishes it from the core group. The species was first described in 1935 by Hall and O'Toole as , isolated from the stool of a healthy newborn and named for the challenges in its cultivation. In 2016, it was reclassified into the newly proposed genus Clostridioides based on molecular evidence demonstrating its divergence from the restricted genus , which is now limited to and closely related species sharing high 16S rRNA gene sequence similarities (>97%) and robust phenotypic coherence. This taxonomic revision was justified by phylogenetic analyses showing that C. difficile clusters separately, with average nucleotide identity values below 60% and DNA-DNA hybridization estimates under 25% relative to the type species, C. butyricum. The genus Clostridioides comprises two species: Clostridioides difficile and Clostridioides mangenotii. Evolutionary relationships of C. difficile have been elucidated through 16S rRNA gene sequencing and whole-genome comparisons, revealing its closest relative as Clostridioides mangenotii with 94.7% 16S rRNA similarity. Other relatives include Paeniclostridium sordellii and , all co-occurring in the Peptostreptococcaceae family and sharing ecological niches in the mammalian gut, though C. difficile's lineage shows distinct adaptations for sporulation and toxin production. No significant taxonomic revisions have occurred since 2016, with the genus Clostridioides firmly established in subsequent phylogenetic frameworks.

Etymology and pronunciation

The genus name Clostridioides is derived from klōstēr (κλωστήρ), meaning "spindle," combined with the suffix -oeidēs (οειδής), meaning "resembling" or "like," and referencing the established Clostridium; this reflects the bacterium's rod-shaped, spindle-like morphology similar to other clostridia. The species epithet difficile is the neuter form of the Latin adjective difficilis, translating to "difficult," a nod to the organism's fastidious growth requirements that complicated its initial isolation and cultivation. Clostridioides difficile was originally described in 1935 by Ivan C. Hall and Elizabeth O'Toole as Bacillus difficilis, isolated from the intestinal contents of healthy newborn infants during studies of neonatal gut flora. It was reclassified as Clostridium difficile in 1938 due to its anaerobic, spore-forming characteristics aligning with the Clostridium genus. In 2016, phylogenetic and genomic analyses prompted its transfer to the newly proposed genus Clostridioides to distinguish it from true clostridia in the family Peptostreptococcaceae, establishing the current binomial nomenclature. The standard pronunciation is /klɒˌstrɪdiˈɔɪdiːz dɪˈfɪsɪleɪ/ in and /kləˌstrɪdiˈɔɪdiːz dɪˈfɪsɪl/ in , with the stressed on the "oy" and the on the second of "difficile." It is commonly abbreviated and referred to as C. difficile or "C. diff" (/siː ˈdɪf/), though mispronunciations like /klɒˈstrɪdiəz/ (omitting the "oy") or French-influenced /di.fiˈsil/ for the name occasionally occur in non-specialist contexts.

Microbiology

Genome

The genome of Clostridioides difficile consists of a single circular approximately 4.3 million base pairs () in length, with a G+C content of about 28.8%. This structure encodes roughly 3,800 to 4,000 protein-coding genes, reflecting the bacterium's adaptation as an capable of spore formation and toxin production. The lacks a second or large extrachromosomal elements in the reference , though smaller plasmids can occur in some isolates. A defining feature is the pathogenicity locus (PaLoc), a 19.6-kilobase (kb) region containing genes for the major factors, including tcdA and tcdB, which encode the large glucosyltransferase s TcdA and TcdB, respectively. The PaLoc also includes accessory genes such as tcdR, encoding a sigma-70-like that positively regulates expression by binding upstream promoters; tcdC, a negative that inhibits TcdR activity; and tcdE, which codes for a holin-like protein aiding release. Additionally, a separate 6.2-kb cdt locus encodes the binary CDT, comprising cdtA and cdtB subunits that ADP-ribosylate , disrupting the , along with cdtR, a promoting expression under specific conditions. These loci are absent or disrupted in non-toxigenic strains, underscoring their role in . The first complete genome sequence was published in 2006 for the multidrug-resistant strain 630, revealing the bacterium's mosaic architecture and paving the way for . By 2025, over 19,000 C. difficile genomes had been sequenced and deposited in public databases like , enabling analyses of evolutionary dynamics and strain diversity. These efforts uncovered , including up to 10% of the genome composed of prophages and transposons, as well as CRISPR-Cas systems (primarily type I-B) that provide adaptive immunity against phages and plasmids. Genetic plasticity in C. difficile is driven by high recombination rates, facilitated by integrases and resolvases associated with mobile elements, allowing rapid acquisition of antibiotic resistance and toxin variants. Prophages, such as those in the φCD27 family, integrate into the and promote , while plasmids (e.g., pCD6.8, ~6.8 kb) carry resistance genes like ermB for . This variability contributes to the pathogen's adaptability, with recombination hotspots near loci enabling diversification across clades.

Epigenome

The epigenome of Clostridioides difficile is characterized by patterns that serve as regulatory switches, primarily involving N6-methyladenine (6mA) modifications. A key mechanism is phase-variable mediated by conserved methyltransferases, such as the solitary Type II methyltransferase CamA (locus tag CD630_2758 in strain 630), which specifically targets the CAAAAA motif. This enzyme is present in all sequenced C. difficile genomes (over 300) and across 36 analyzed isolates, contributing to epigenomic diversity while operating independently of restriction endonucleases in most cases. Multiple restriction-modification (R-M) systems are prevalent, with an average of 3.5 Type II systems per , where patterns influence approximately 20% of these systems by modulating accessibility and expression without altering the DNA sequence. A seminal 2019 study utilizing single-molecule real-time (SMRT) sequencing revealed that these patterns enable reversible switches in , with non-methylated CAAAAA sites enriched in promoter regions, binding sites, and transcription start sites. Inactivation of camA resulted in a approximately 50% reduction in sporulation efficiency, primarily through delayed activation of the early sporulation σF, highlighting 's role in developmental timing. Similarly, variable at orthologous CAAAAA sites overlaps with genes involved in , such as fliZ and fliN, suggesting epigenetic control of flagellar assembly and bacterial dissemination. Although production levels remained unaffected in camA mutants, these findings underscore how facilitates phenotypic heterogeneity without genetic mutations. These epigenetic mechanisms provide C. difficile with adaptive advantages, enabling rapid responses to environmental stresses such as antibiotics. In mouse models of , camA mutants exhibited significantly reduced colonization and persistence in the presence of clindamycin, indicating that methylation supports survival and transmission in perturbed gut microbiomes. Overall, the epigenome's , mapped via SMRT sequencing, positions it as a critical layer of distinct from genomic sequence variations, briefly intersecting with loci to fine-tune expression.

Bacteriophage

Clostridioides difficile is primarily infected by temperate belonging to the families Myoviridae and Siphoviridae within the order . Myoviruses, such as phiCD27 and phiC2, represent the most abundant group, characterized by contractile tails that facilitate host attachment and genome injection. Siphoviruses, including phiCDHM1 and phiSM101-like phages, feature non-contractile tails and are less common but contribute to diversity in environmental isolates. As of 2022, more than 35 distinct C. difficile phage genomes have been sequenced and publicly deposited, with ongoing discoveries suggesting dozens more by 2025, many of which integrate as in bacterial genomes. Several of these phages encode accessory genes, such as tcdE (a holin aiding secretion) or components of the binary (cdtA and cdtB), potentially enhancing host during lysogeny. The lifecycle of C. difficile bacteriophages predominantly follows a temperate , where upon , the phage integrates into the bacterial as a via , often at tRNA or conserved intergenic sites. This lysogenic state allows the phage to replicate passively with the host during , conferring potential benefits like immunity to by similar phages. Induction to the occurs under stress conditions, such as exposure to antibiotics (e.g., or fluoroquinolones), DNA damage, or quorum-sensing signals, triggering prophage excision, virion assembly, and host to release new particles. While most identified phages are temperate, engineered or naturally lytic variants have been isolated and show promise for targeted bacterial killing without lysogeny. Bacteriophages play a key role in the of C. difficile by facilitating through , where phage particles package and transfer bacterial DNA fragments, including virulence-associated genes, between strains. This process contributes to , enabling the spread of factors like antibiotic resistance cassettes or regulatory elements that modulate expression, though direct transduction of the core genes tcdA and tcdB (located in the chromosomal PaLoc) is rare and more commonly achieved via conjugation. induction can also indirectly influence by altering host physiology, such as upregulating production (e.g., TcdB levels increase post-infection in some strains) or promoting formation, thereby enhancing survival in the gut . These interactions underscore phages' contribution to strain variation, with multiple prophages (up to 5–6 per genome) driving adaptive in clinical and environmental isolates. Therapeutic exploitation of C. difficile phages focuses on lytic variants or engineered cocktails to selectively decolonize the without disrupting commensal . Preclinical studies, including hamster models of , have demonstrated that phage mixtures (e.g., combining myoviruses like phiCDHM3 and siphoviruses) reduce bacterial loads by over 90% and attenuate toxin-mediated pathology when administered orally post-infection.

Strains

Clostridioides difficile strains are classified using methods such as ribotyping, which identifies variations in the 16S-23S rRNA intergenic spacer region, and (MLST), which analyzes seven housekeeping genes to assign sequence types (STs). ribotyping has been the most widely adopted globally for , revealing over 300 ribotypes, while MLST groups strains into clades, with ST1 associated with ribotype 027 (RT027) and ST11 with RT078. Key toxigenic strains include RT001 and RT106, both producing toxins A and B (A+B+ profile) and belonging to MLST clade 1; RT001 is historically prevalent in , while RT106 has emerged as a dominant type in recent years. RT017, common in , features an A-B+ toxin profile due to a deletion in the A and is linked to severe in that region. The hypervirulent RT027 (also known as BI/NAP1/027, ) produces elevated levels of toxins A, B, and binary (CDT), often with fluoroquinolone due to mutations like Thr82Ile in gyrA. RT078 (ST11) similarly produces CDT and is associated with community and animal sources. An emerging hypervirulent strain, RT955 (), reported in the UK from late 2023 to 2025, shows increased severity, , and genetic similarity to RT027. Virulence varies significantly among strains; hypervirulent types like RT027 and RT955 produce higher quantities, leading to more severe and higher recurrence rates, compounded by resistance profiles such as fluoroquinolone insensitivity. In contrast, non-toxigenic strains, which lack genes, primarily facilitate and may competitively inhibit toxigenic strains, reducing risk in carriers. Globally, RT027 dominated infections in and before the 2010s, driving epidemics, but its prevalence declined post-2010 due to enhanced infection control and reduced fluoroquinolone use. By 2025, shifts have occurred, with RT106 becoming prominent in and parts of , while RT017 persists in and RT955 emerges in the UK.

Pathogenesis and virulence

Role as a human pathogen

Clostridioides difficile acts as an opportunistic pathogen in humans, exploiting disruptions in the normal , most commonly following therapy. It is the leading cause of healthcare-associated infectious and accounts for approximately 20-30% of all -associated cases. Furthermore, it serves as the primary etiologic agent of pseudomembranous , a severe inflammatory condition characterized by the formation of pseudomembranes on the colonic mucosa. The pathogen's virulence is primarily driven by two large glucosyltransferase s: A (TcdA), an enterotoxin that triggers fluid and , and B (TcdB), a potent cytotoxin that disrupts the of intestinal epithelial cells. About 20% of C. difficile strains also produce the binary CDT (Clostridium difficile ), which ADP-ribosylates and may enhance the severity of when co-expressed with TcdA and TcdB. In addition, the bacterium's ability to form highly resistant endospores allows for prolonged environmental survival, facilitating and reinfection in susceptible hosts. C. difficile was first recognized as a in 1978, when studies linked its toxins to cases of antibiotic-associated pseudomembranous previously attributed to other causes. The term "" (CDI) was formalized in the 2010 clinical practice guidelines issued by the Infectious Diseases Society of America (IDSA) and the Society for Healthcare Epidemiology of America (SHEA), reflecting updated understanding of its and management. Although C. difficile exhibits zoonotic potential with isolation from various animal species, foods, and environmental sources, humans represent the principal reservoir for toxigenic strains responsible for clinical disease.

Antibiotic resistance

Clostridioides difficile exhibits both intrinsic and acquired resistance to multiple antibiotics, contributing to its persistence in healthcare settings and complicating treatment of associated infections. Intrinsic resistance to beta-lactams is mediated by chromosomal genes encoding beta-lactamases, such as CDD-1 and CDD-2, which hydrolyze the beta-lactam ring, rendering penicillins and cephalosporins ineffective. Similarly, intrinsic resistance to clindamycin and other macrolide-lincosamide-streptogramin B (MLSB) antibiotics occurs through ribosomal methylation via the erm(B) gene, which modifies the 23S rRNA target site and is often carried on mobile genetic elements like Tn5398. These mechanisms allow C. difficile to survive exposure to antibiotics commonly used in clinical practice. Acquired resistance has emerged particularly to fluoroquinolones, driven by point mutations in genes such as gyrA (e.g., Thr82Ile) and gyrB, which alter the drug's and are prevalent in strains. Efflux pumps, including AcdP (encoded by CD2068) and Mef variants like mefH paired with msr(D), actively expel antibiotics such as , tetracyclines, and fluoroquinolones from the cell, conferring multidrug efflux capabilities. Ribosomal protection proteins encoded by ermB further enhance resistance by preventing drug binding to the . Resistance linked to the binary toxin, often co-occurring in hypervirulent isolates, involves genetic elements that promote overall multidrug tolerance, though direct mechanistic ties remain under investigation. The evolution of resistance in C. difficile is exemplified by hypervirulent strains like PCR ribotype 027 (RT027), which display multidrug resistance profiles encompassing fluoroquinolones, , and tetracyclines, facilitated by and selective pressure from use. Recent studies from 2023–2025 indicate emerging reduced susceptibility to in some isolates, attributed to mutations in the vanGCD operon (e.g., Ser313Phe in CD) and murG, leading to elevated minimum inhibitory concentrations (MICs) that challenge standard therapies, though many global isolates remain susceptible. These adaptations highlight the pathogen's genomic plasticity in response to antimicrobial exposure. Antibiotic resistance in C. difficile significantly impacts clinical outcomes, contributing to recurrence rates exceeding 20% following initial treatment, as resistant strains evade eradication and persist in the gut microbiota. The selective pressure exerted by broad-spectrum antibiotics, such as cephalosporins and clindamycin, not only promotes the proliferation of resistant C. difficile but also disrupts host microbiota, facilitating recurrent infections. This underscores the need for stewardship to mitigate resistance evolution.

Pathophysiology

Clostridioides difficile, a Gram-positive, spore-forming anaerobe, initiates through the of its resilient spores in the disrupted intestinal environment, primarily following antibiotic-induced . Antibiotics such as clindamycin or fluoroquinolones deplete protective commensal , reducing microbial diversity and creating a niche for C. difficile overgrowth by eliminating competitors that inhibit spore and vegetative . This elevates levels of primary acids like taurocholate in the , as gut microbes normally convert them to secondary acids (e.g., deoxycholate) that suppress C. difficile growth; taurocholate acts as a potent germinant, binding to the SpoVA in the spore's inner to calcium release and cortex , converting dormant spores to metabolically active vegetative cells. is further potentiated by co-germinants such as , which interact with pseudoproteases like CspC to sense environmental cues, enabling rapid of the colon without tissue invasion. Once vegetatively growing, C. difficile produces two major exotoxins, A (TcdA) and B (TcdB), which are glucosyls that inactivate Rho (RhoA, Rac1, Cdc42) by adding glucose moieties, disrupting dynamics in colonic epithelial cells. TcdA primarily induces fluid secretion and mucosal inflammation by glucosylation of Rho proteins in enterocytes, leading to loss of tight junctions, increased permeability, and release that recruits neutrophils; TcdB, more potent in , causes similar glucosylation but targets a broader range of Rho isoforms, resulting in cell rounding, , and barrier disruption. A subset of hypervirulent strains also secretes binary CDT (Clostridium difficile ), an ADP-ribosyl that modifies G-, depolymerizing and impairing while enhancing TcdA/TcdB adhesion to host cells via alterations. These -mediated effects are purely extracellular and non-invasive, relying on (e.g., via family receptors for TcdB) to deliver the glucosyl domain into the . Pathological progression begins with toxin-induced epithelial damage, progressing from superficial erosions to full-thickness mucosal injury and formation of pseudomembranes—adherent plaques composed of , , inflammatory cells, and cellular debris that coalesce into a characteristic "" appearance on the colonic surface. In mild cases, this manifests as localized , but unchecked activity exacerbates and , potentially leading to severe complications like , where colonic dilation (>6 cm) and systemic toxicity arise from massive fluid shifts, bacterial translocation, and storms. The process is self-perpetuating in dysbiotic conditions, as ongoing imbalance sustains vegetative proliferation and production, amplifying tissue destruction without direct bacterial invasion.

Immune response

The innate immune response to Clostridioides difficile infection begins with recognition of bacterial flagella by Toll-like receptor 5 (TLR5), which signals through the adaptor protein MyD88 to activate and, to a lesser extent, MAPK pathways in epithelial cells, promoting an early proinflammatory state. This TLR5-MyD88 axis is crucial for restoring intestinal innate defenses disrupted by antibiotics, as demonstrated in mouse models where TLR5 stimulation ameliorates inflammation and prevents mortality from C. difficile colonization. Additionally, C. difficile toxins A and B induce IL-8 production from intestinal epithelial cells and monocytes, recruiting to the site of infection and amplifying local inflammation. Toxins also activate the and pyrin in macrophages and epithelial cells, leading to caspase-1-mediated processing and release of IL-1β, which drives neutrophil influx and contributes to mucosal injury during acute infection. Adaptive immunity against C. difficile involves Th17 cells, which differentiate in response to and produce IL-17 and to enhance barrier integrity and protect the from . IL-17 correlates with disease severity in humans and promotes recruitment of neutrophils and , while supports epithelial repair and restricts expansion in mouse models of colitis-associated . Circulating IgG antibodies specific to toxins A and B neutralize toxin activity, reducing recurrence risk by preventing toxin-mediated damage; levels of toxin-specific IgG post- predict protection against reinfection. Regulatory T cells (Tregs) mitigate excessive by suppressing Th17 responses and IL-17 , balancing immunity to limit tissue destruction without compromising clearance. Dysregulation of the exacerbates C. difficile outcomes, with overactive innate and adaptive signaling—particularly excessive IL-1β and IL-17—causing collateral tissue damage, barrier disruption, and worsened in animal models. In elderly hosts, age-related impairs adaptive responses, including reduced Th17 differentiation and toxin-specific IgG production, leading to higher susceptibility, severe disease, and recurrence rates. Recurrent cases often show persistent Treg dysfunction and diminished responses, failing to sustain protective . Insights from vaccine development highlight immune correlates of protection, such as bezlotoxumab, a targeting B (TcdB), which neutralizes cytotoxicity, reduces , and prevents recurrence by preserving thymic function and limiting extraintestinal damage in preclinical models. Recent 2024 clinical trials, including those for adjuvanted vaccines like GSK's candidate and multivalent mRNA-LNP platforms, demonstrate robust induction of -neutralizing IgG and Th17-associated cytokines (IL-17/) as key correlates, with AS01-adjuvanted formulations eliciting superior systemic and mucosal responses in healthy adults.

Possible role in colon cancer

Emerging research has proposed that Clostridioides difficile may contribute to colorectal cancer (CRC) development through its toxins, which induce chronic inflammation and DNA damage in colonic tissues. Specifically, toxins TcdA and TcdB disrupt the epithelial barrier by targeting tight junction proteins such as claudins and occludins, leading to increased permeability and persistent low-grade inflammation that activates oncogenic pathways like NF-κB and STAT3. Additionally, the binary toxin CDT has been linked to genotoxicity, causing DNA damage in intestinal epithelial cell lines by disrupting microtubule assembly and promoting double-strand breaks. These hypotheses stem from observations that chronic C. difficile exposure creates a pro-tumorigenic microenvironment, though direct causation in humans remains unproven. Studies from 2018 to 2025 have provided evidence of elevated C. difficile abundance in CRC tumors and its association with accelerated tumorigenesis. Metagenomic analyses of CRC patient biopsies have detected higher levels of C. difficile DNA and toxins in tumor tissues compared to healthy controls, with one epidemiological study reporting a 2.7-fold increased CRC risk following C. difficile infection. In mouse models, chronic colonization with patient-derived toxigenic C. difficile strains in germ-free ApcMin/+ mice significantly increased tumor burden, with tumor counts rising by up to 3-fold in the presence of toxin-producing strains. These effects were toxin-dependent, as non-toxigenic mutants failed to promote tumor growth. Mechanistically, C. difficile toxins foster oncogenesis by generating (ROS) via TcdB, which upregulates Wnt/β-catenin signaling and induces in colonic stem cells, while CDT exacerbates genotoxic stress. This leads to epithelial barrier disruption, allowing microbial translocation and shifts that reduce short-chain production while elevating secondary bile acids, both of which favor . from these processes recruits IL-17-producing immune cells, further amplifying tumor progression in preclinical models. However, no direct causal has been established in cohorts. Controversies persist regarding whether C. difficile acts as a primary driver or merely an opportunist in dysbiotic environments predisposed to . While animal and data support a promotional role, human studies largely show correlations, such as enriched C. difficile in metastatic , without isolating it from confounding alterations or exposures. The variability in production across strains complicates interpretations, and longitudinal clinical trials are needed to clarify .

Epidemiology

Transmission

Clostridioides difficile is transmitted primarily through the fecal-oral route, with its infectious spores serving as the key vehicle for spread between hosts. These spores are shed in the of infected or colonized individuals and can contaminate hands, surfaces, , and medical equipment, facilitating indirect . Once ingested, the spores resist acid and germinate in the colon, leading to infection. The durability of C. difficile spores significantly contributes to environmental persistence and transmission. Spores can survive on dry surfaces such as floors, bed rails, and toilets for up to five months, remaining viable under diverse conditions including , , and many disinfectants. Notably, they are resistant to alcohol-based hand sanitizers, which fail to inactivate them, underscoring the importance of soap-and-water handwashing in prevention efforts. In healthcare settings, where approximately 50% of cases are healthcare-associated, transmission is amplified by contaminated healthcare worker hands (detected in up to 50% of interactions with infected patients) and shared equipment like commodes and thermometers. Community-acquired infections have risen notably, now accounting for approximately 50% of cases, often linked to prior outpatient exposure or unrecognized environmental contact. Environmental reservoirs beyond healthcare include , , sediments, and , as well as various animals such as , pets, and , which may harbor toxigenic strains. Foodborne transmission remains rare but has been documented, particularly in and processed meats during studies in the , where viable spores were isolated from retail samples at rates up to 5%. Asymptomatic carriers play a critical role in sustaining transmission chains, with carriage rates of 5-15% among hospitalized patients serving as a major source of spore shedding into the environment. These individuals, often comprising 10-20% of admissions, can unknowingly disseminate spores via fecal contamination, contributing to both healthcare-associated and community outbreaks without developing symptoms themselves.

Host range

Clostridioides difficile primarily infects humans, where it is a leading cause of antibiotic-associated , particularly affecting vulnerable populations such as infants and the elderly. In infants, an immature with limited microbial diversity allows for high rates of , often exceeding 50% in the first months of life, though is rare due to low production. Among the elderly, age-related decline in diversity and shifts toward pro-inflammatory taxa reduce resistance, increasing susceptibility to symptomatic . The bacterium also colonizes a wide range of animals, establishing it as a potential zoonotic with evidence of interspecies transmission. In food-producing animals, pigs serve as major reservoirs, with prevalence rates up to 43% in piglets and adults, often carrying hypervirulent ribotype 078 (RT078), which is genetically similar to strains causing human infections and suggesting farm-to-human zoonotic spread. , dogs, and are also common hosts; neonatal foals and piglets exhibit high shedding (up to 100%) due to underdeveloped , while adult horses develop severe post-antibiotics. Companion animals like dogs and cats show variable carriage rates (0-38%), with some isolates matching human-associated ribotypes, though direct household transmission appears infrequent. Non-mammalian hosts are rare, but C. difficile has been detected in reptiles and , typically without causing overt disease. In reptiles such as , , and chelonians, isolation rates are low (around 4%), yet toxigenic strains linked to human ribotypes (e.g., RT081) indicate potential risks from captive or wild specimens. Among , prevalence is similarly low (e.g., 4% in shelter samples), confined to species like and wild avians, with shared ribotypes like RT078 and RT002 between birds, animals, and humans. do not serve as natural hosts or vectors; however, houseflies (Musca domestica) can mechanically transmit spores for up to 4 hours after exposure, facilitating environmental spread in settings like farms or hospitals. Across host species, and disease risk are governed by composition, which provides competitive exclusion against C. difficile, and exposure, which universally disrupts this barrier regardless of host. Neonates in both humans and animals are particularly susceptible due to microbiota immaturity, while antibiotics like clindamycin or fluoroquinolones precipitate overgrowth in adults of multiple species.

Incidence and prevalence

Clostridioides difficile infection (CDI) imposes a significant global burden, with an estimated global incidence of 49 cases per 100,000 person-years, equating to approximately 4 million cases annually, with an estimated 500,000 cases annually in the United States alone, contributing to approximately 30,000 deaths each year and a ranging from 6% to 10% among infected individuals. In the / (EU/EEA), healthcare-associated CDI affects nearly 124,000 patients yearly, resulting in about 3,700 deaths, with an incidence rate of approximately 3.7 cases per 10,000 patient-days in hospitals. Epidemiological trends indicate a post-COVID-19 surge in CDI cases, driven by increased antibiotic use during the pandemic, which disrupted gut microbiota and heightened susceptibility. In the , CDI cases rose by 33% from the 2020/2021 period to 2023/2024, reaching 19,239 reported infections in the year ending January 2025. In the , community-onset CDI has been increasing, accounting for over 50% of cases by 2019 and continuing to rise, reflecting a shift from predominantly healthcare-associated infections. Hospitalized elderly individuals over 65 years old face a 10-fold higher risk of CDI compared to younger adults, with this age group representing the majority of severe cases and deaths. In high-burden regions such as Poland's , the 2023 incidence reached 65.1 cases per 100,000 population, underscoring disparities in . Emerging patterns show a growing divide between healthcare-associated and community-acquired CDI, with the latter increasingly driven by non-hospital sources and a diversification beyond traditional hypervirulent strains like ribotype 027, including non-027/078 variants causing severe disease in community settings.

Clinical aspects

Signs and symptoms

Clostridioides difficile infection (CDI) most commonly manifests as watery , often accompanied by abdominal cramping and tenderness, typically beginning 5 to 10 days after initiation of antibiotic therapy, though onset can occur as early as the first day or up to 2 months later. The diarrhea is usually nonbloody but may contain or occult blood, with patients experiencing three or more unformed stools per 24 hours. Additional symptoms can include fever, , loss of appetite, and , particularly in more pronounced cases. In mild CDI, patients remain hemodynamically stable without systemic signs of infection, presenting primarily with the aforementioned and mild abdominal discomfort, often without fever or significant tenderness on examination. Severe CDI, by contrast, involves systemic illness with marked ( count ≥15,000 cells/μL) or elevated serum (≥1.5 mg/dL), alongside , intense , and fever. Endoscopic evaluation in severe cases may reveal pseudomembranous , characterized by yellowish plaques on the colonic mucosa. Fulminant CDI represents a life-threatening progression, featuring or , (absence of bowel sounds), or (colonic dilation >6 cm), often necessitating intensive care admission and carrying a of up to 30–40%. Rare extraintestinal manifestations, such as bacteremia, can occur, particularly in immunocompromised individuals or those with gastrointestinal breaches. Recurrent CDI affects approximately 20–30% of patients within 2 months of initial resolution, presenting with symptoms similar to the primary episode, including watery and , though severity can vary. carriage of toxigenic C. difficile occurs in 3–5% of the general population, without or other symptoms, representing rather than active .

Diagnosis

Diagnosis of Clostridioides difficile infection (CDI) begins with clinical suspicion, typically in patients presenting with new-onset diarrhea—defined as three or more unformed stools in 24 hours—particularly following recent antibiotic exposure or hospitalization, after excluding other potential causes such as laxative use or alternative infections. Testing is not recommended for asymptomatic individuals, as up to 15% of hospitalized patients may carry toxigenic C. difficile without symptoms, leading to unnecessary treatment and increased resistance risks. Laboratory confirmation relies on stool-based tests targeting the presence of toxigenic C. difficile or its toxins. Nucleic acid amplification tests (NAAT), such as PCR, detect the toxin-encoding genes tcdA and tcdB with high sensitivity (typically 95–100%) and specificity (90–100%), providing rapid results within hours; however, they cannot distinguish active infection from colonization. Enzyme immunoassays (EIA) for toxins A and B offer high specificity (around 90–100%) but lower sensitivity (70–90%), making them suitable for confirming toxin production in symptomatic patients. The cell cytotoxicity neutralization assay (CCNA), which detects toxin B's cytopathic effects on cell cultures, serves as the historical gold standard with sensitivity of 80–95% and specificity exceeding 99%, though it is labor-intensive and requires 24–48 hours. Glutamate dehydrogenase (GDH) antigen tests, with sensitivity up to 100%, are often used as initial screens due to their speed but require follow-up with toxin or NAAT confirmation owing to lower specificity (85–95%). Diagnostic algorithms typically employ a two-step approach to balance while minimizing . The Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA) recommend starting with GDH or NAAT screening, followed by toxin EIA for positives; discordant results may be arbitrated by NAAT or . In severe or cases where stool testing is inconclusive or unobtainable, or can visualize characteristic yellow-white pseudomembranes on the colonic mucosa, confirming with near-100% specificity when present, though this is not routine due to procedural risks. Key challenges in CDI diagnosis include differentiating colonization from true infection, as NAAT positivity alone can occur in 5–15% of asymptomatic carriers, potentially leading to overtreatment. Current guidelines, including the 2021 IDSA/SHEA update and 2025 institutional protocols aligned with them, emphasize prioritizing -positive tests (e.g., EIA or ) over NAAT alone to confirm active disease, particularly in low-prevalence settings or non-severe cases. Repeat testing within seven days of a negative result is discouraged unless symptoms worsen significantly, to avoid false negatives from instability in unrefrigerated samples.

Management

Treatment

The primary treatment for an initial episode of non-fulminant () involves antibiotics that target the pathogen while minimizing disruption to the . , administered at 200 mg orally twice daily for 10 days, is recommended as the preferred first-line therapy due to its lower recurrence rate compared to alternatives, with clinical trials demonstrating sustained response rates of approximately 70% at one month post-treatment. Alternatively, oral at 125 mg four times daily for 10 days is used, particularly when is unavailable, achieving initial cure rates exceeding 85% in randomized controlled trials. For mild, non-severe cases in low-risk patients, oral 500 mg three times daily for 10 days may be considered when preferred agents are inaccessible, though its use has declined due to emerging resistance concerns and inferior outcomes in comparative studies. For recurrent CDI, remains the preferred option, with the standard 10-day regimen or an extended-pulsed regimen (200 mg twice daily for 5 days, followed by once daily on alternate days for days 21–25) reducing recurrence by up to 50% relative to in phase 3 trials. tapered and pulsed regimens (e.g., 125 mg four times daily for 10–14 days, then twice daily for 7 days, once daily for 7 days, and every 2–3 days for 2–8 weeks) serve as an alternative for first recurrences, with observational data showing recurrence prevention in about 60% of cases. For patients with multiple recurrences (≥2 episodes), fecal microbiota transplantation (FMT) is recommended, achieving cure rates of 80–90% in meta-analyses of over 40 studies involving more than 1,000 patients, typically delivered via after a course of antibiotics to clear C. difficile. Adjunctive therapies enhance outcomes in high-risk recurrent cases. Bezlotoxumab, a against B, is administered as a single 10 mg/kg intravenous during treatment, reducing recurrence risk by 40% in phase 3 trials of over 2,600 patients with prior episodes. Approved microbiome-based products include Rebyota (fecal , live-jslm), a single following antibiotics for recurrent prevention, with phase 3 data showing 70.6% treatment success at 8 weeks compared to 57.5% with . Vowst (fecal spores, live-brpk), an oral capsule regimen of four capsules daily for three days post-antibiotics, similarly prevents recurrence in 88% of recipients versus 60% with in pivotal trials. Supportive measures are integral to management. Discontinuing the inciting antibiotic is prioritized to allow microbiota recovery, as this alone resolves up to 20% of mild cases without further intervention, per cohort studies. For fulminant CDI (characterized by hypotension, shock, ileus, or megacolon), high-dose oral or nasogastric vancomycin (500 mg four times daily) combined with intravenous metronidazole (500 mg every 8 hours) is used, with surgical intervention such as subtotal colectomy considered for refractory toxic megacolon, improving survival from <30% to over 50% in severe cases. Emerging therapies include investigational modulators and targeted antimicrobials. As of 2025, Idorsia's synthetic glycan-based candidate for C. difficile, covering over 90% of circulating strains, has advanced in early clinical development following positive phase 1 data. , using bacteriophages specific to C. difficile ribotypes, shows promise in preclinical models and early-phase studies for reducing production without broad disruption.

Prevention

Prevention of Clostridioides difficile infection (CDI) primarily focuses on infection control measures in healthcare settings, where the majority of cases occur due to transmission via contaminated surfaces and hands. Key strategies include isolating patients with suspected or confirmed CDI and implementing contact precautions, such as wearing gloves and gowns, to limit spread. Hand hygiene with and water is emphasized over alcohol-based sanitizers because soap effectively removes C. difficile spores, which are resistant to . Environmental cleaning using sporicidal agents like (bleach) or hydrogen peroxide-based disinfectants is crucial for terminal room disinfection, as these agents inactivate spores on high-touch surfaces. Antibiotic stewardship programs are essential to reduce CDI risk by limiting unnecessary prescriptions and restricting high-risk antibiotics such as clindamycin, cephalosporins, and fluoroquinolones, which disrupt the and promote C. difficile overgrowth. These programs, recommended by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (), involve prospective audit and feedback to clinicians, resulting in decreased CDI incidence in hospitals implementing them. The 2022 SHEA compendium update recommends targeted interventions such as enhanced environmental cleaning and to reduce CDI incidence in healthcare settings. Probiotics, particularly , have been investigated as adjuncts to prevent in patients receiving antibiotics, with some trials showing reduced risk of healthcare-onset when administered prophylactically. However, major guidelines, including those from the American College of Gastroenterology, do not routinely recommend for primary or secondary prevention due to inconsistent evidence and potential risks in immunocompromised patients. For high-risk individuals, such as those with prior CDI episodes, bezlotoxumab—a against toxin B—can prevent recurrence when administered during treatment, reducing recurrence rates by about 10% compared to in clinical trials. Experimental s targeting C. difficile toxins, such as Pfizer's PF-06425090 evaluated in the phase 3 CLOVER trial, have shown potential in reducing severity and duration despite not meeting primary efficacy endpoints for prevention. As of 2025, no is licensed, but ongoing explores multivalent approaches, including mRNA-based platforms. Public health efforts include through the CDC's Emerging Infections Program (EIP), which tracks CDI incidence and trends to inform prevention policies, revealing shifts toward more community-associated cases. Reducing community spread involves education on hygiene practices, such as prompt handwashing and avoiding overuse in outpatient settings. Guidelines emphasize stewardship, including the judicious use of narrow-spectrum antibiotics, to support health and prevent CDI, though microbiome restoration strategies are not yet standardized for routine prevention.

References

  1. [1]
    Comprehensive review of Clostridium difficile infection - NIH
    This paper reviews recent research on CDI's epidemiological characteristics, risk factors, diagnosis, treatment, and preventionMissing: authoritative | Show results with:authoritative
  2. [2]
    Clostridioides (Clostridium) Difficile Colitis - Medscape Reference
    May 8, 2025 · C difficile colitis results from a disruption of the normal bacterial flora of the colon, colonization with C difficile, and release of toxins that cause ...Missing: authoritative sources
  3. [3]
    About C. diff - CDC
    Dec 18, 2024 · C. diff is a germ that causes diarrhea and colitis (an inflammation of the colon) and can be life-threatening. C. diff can affect anyone.Missing: authoritative | Show results with:authoritative
  4. [4]
    Reclassification of Clostridium difficile as Clostridioides ... - PubMed
    Based on 16S rRNA gene sequence analysis, the closest relative of Clostridium difficile is Clostridium mangenotii with a 94.7% similarity value.
  5. [5]
    Taxonomy browser (Clostridioides difficile) - NCBI
    THE NCBI Taxonomy database allows browsing of the taxonomy tree, which contains a classification of organisms.
  6. [6]
    Proposal to restrict the genus Clostridium Prazmowski to ... - PubMed
    Numerous phylogenetic studies, principally based on sequencing of the 16S rRNA gene, indicate that the genus Clostridium should be restricted to Clostridium ...Missing: DDH | Show results with:DDH
  7. [7]
    Genome-Based Comparison of Clostridioides difficile: Average ... - NIH
    Feb 14, 2018 · The closest neighbor of C. difficile on this tree is Paeniclostridium sordellii. Their close relationship was also demonstrated with a 16S rRNA ...
  8. [8]
    Genus: Clostridioides - LPSN
    Name: Clostridioides Lawson et al. · Category: Genus · Proposed as: gen. · Etymology: Clos. · Gender, pronunciation: neuter (stem: Clostridioid-), klos-tri-di-o-I- ...
  9. [9]
    Etymologia: Clostridium difficile - PMC - NIH
    The species name difficile is a form of the Latin adjective difficilis because when first identified (by Hall and O'Toole in 1935), the organism was difficult ...
  10. [10]
    Species: Bacillus difficilis - LPSN
    Original publication: Hall IC, O'Toole E. Intestinal flora in newborn infants with a description of a new pathogenic anaerobe, Bacillus difficilis. American ...
  11. [11]
    Definition of Clostridium difficile - NCI Dictionary of Cancer Terms
    Clostridium difficile. Listen to pronunciation. (klah-STRIH-dee-um dih-FIH-sih-lee).<|control11|><|separator|>
  12. [12]
    Diversity and Evolution in the Genome of Clostridium difficile - PMC
    Encompassing a 19.6-kb region of the chromosome, the PaLoc has received significant attention, as it contains the genes encoding the major virulence factors ...
  13. [13]
    Molecular Basis of TcdR-Dependent Promoter Activity for Toxin ...
    Apr 23, 2023 · The pathogenicity locus (PaLoc) of Clostridioides difficile. Four TcdR-dependent promoters are located before the genes tcdR, tcdA, and tcdB.
  14. [14]
    CdtR Regulates TcdA and TcdB Production in Clostridium difficile
    The TcdB, TcdA and binary toxins produced by C. difficile are encoded in two genomically distinct loci: TcdB and TcdA in the Pathogenicity Locus (PaLoc) and ...
  15. [15]
    Mechanisms of Clostridioides difficile glucosyltransferase toxins and ...
    PaLoc is the primary pathogenic locus present in all toxic isolates, while CdtLoc is present only in a minority of PCR ribotypes (Awad et al., 2014). TcdA and ...
  16. [16]
    Global insights into the genome dynamics of Clostridioides difficile ...
    Jan 14, 2025 · Methods: A total of 19,711 C. difficile genomes were retrieved from GenBank. Prokka was used for genome annotation, and multi-locus sequence ...
  17. [17]
    CRISPR Diversity and Microevolution in Clostridium difficile - NIH
    CRISPR-Cas immune systems provide adaptive resistance against invasive genetic elements such as phages (Barrangou et al. 2007) and plasmids (Marraffini and ...Missing: Clostridioides | Show results with:Clostridioides
  18. [18]
  19. [19]
    Clostridioides difficile phage biology and application - PMC - NIH
    In a typical phage replication cycle, a virion first binds to a receptor on the surface of a susceptible host and subsequently injects its genetic material ...Missing: plasticity | Show results with:plasticity<|control11|><|separator|>
  20. [20]
    Diverse Temperate Bacteriophage Carriage in Clostridium difficile ...
    difficile phages have been fully sequenced and annotated. Three belong to the Myoviridae (phiC2, phiCD119, and phiCD27) and two to the Siphoviridae ...Missing: Clostridioides | Show results with:Clostridioides
  21. [21]
    Prophage Carriage and Genetic Diversity within Environmental ...
    Dec 19, 2023 · At least 11% of the genome of the C. difficile reference strain CD630 is composed of mobile genetic elements (MGEs), including prophages [13].Missing: plasticity rates
  22. [22]
    Diversity, Dynamics and Therapeutic Application of Clostridioides ...
    There are thirty-five C. difficile phage genomes publicly available to date and all have dsDNA genomes. Twenty-six of the phages were classified as myoviruses, ...4. Phage Mechanics Of... · 4.3. Impact Of Phage... · 6.2. Clostridium Shuttle...
  23. [23]
    Effect of phage infection on toxin production by Clostridium difficile
    One of the accessory toxin genes, tcdE, was detected in three phages, phiC2, phiC6 and phiC8; however, the non-repeating regions of tcdA and tcdB encoding the ...
  24. [24]
    Emerging applications of phage therapy and fecal virome ...
    May 9, 2023 · Clostridioides difficile, which causes life-threatening diarrheal disease, is considered an urgent threat to healthcare setting worldwide.
  25. [25]
    Bacteriophages Contribute to Shaping Clostridioides (Clostridium ...
    Although uncommon, as many as 5–6 different prophages were identified in a single C. ... Identification of large cryptic plasmids in Clostridioides (Clostridium) ...
  26. [26]
    Clostridioides difficile – phage relationship the RNA way
    Dec 15, 2021 · difficile strain carries usually between 1 and 3 and up to 6 prophages in addition to phage-related genomic regions (see Ref. [19] for review).
  27. [27]
    Clostridioides difficile as a Potential Pathogen of Importance to One ...
    ... many as 5 have been found in certain cases (Fortier, 2018). Bacteriophages found in C. difficile have been linked to increasing AMR, as reported in 2013 ...
  28. [28]
    Horizontal gene transfer converts non-toxigenic Clostridium difficile ...
    Oct 17, 2013 · A toxigenic C. difficile strain is capable of converting a non-toxigenic strain to a toxin producer by horizontal gene transfer.
  29. [29]
    Bacteriophages Contribute to Shaping Clostridioides (Clostridium ...
    Aug 31, 2018 · Although uncommon, as many as 5–6 different prophages were identified in a single C. difficile genome (Amy et al., 2018; Ramírez-Vargas et al., ...
  30. [30]
    Bacteriophage Combinations Significantly Reduce Clostridium ...
    The efficacy of phage lysis varied considerably. Three phages, phiCDHM3, phiCDHS1, and phiCDHM5, were able to lyse the most C. difficile isolates and ribotypes: ...
  31. [31]
    Future of Engineered Phage Therapy for Clostridium difficile Infections
    Jul 23, 2023 · This article provides a concise summary of existing CRISPR-Cas systems that can be utilized to create a lytic phage as a potential treatment for CDIs.<|control11|><|separator|>
  32. [32]
    [EPUB] Non-antibiotic therapies for multidrug-resistant gastrointestinal ...
    May 6, 2025 · For instance, preclinical trials have demonstrated that bacteriophages targeting C. difficile reduce bacterial colonization and toxin production ...
  33. [33]
    Distribution of Clostridioides difficile ribotypes and sequence types ...
    Nov 13, 2024 · C. difficile strains are distributed into ribotypes by PCR-based ribotyping, whereas genome-based sequence types (STs) divide strains into five ...
  34. [34]
    History and Evolution of the Hypervirulent Clostridioides difficile ...
    Oct 15, 2025 · Clostridioides difficile was first identified in 1935 and subsequently emerged over the next several decades as the predominant bacterial cause ...
  35. [35]
    Phylogenomic analysis of Clostridioides difficile ribotype 106 strains ...
    Dec 17, 2020 · ... closest-related isolates (GV597 and GV753) were divergent by 113 SNPs. ... difficile strains that claded with RT106 in the phylogenetic tree ...
  36. [36]
  37. [37]
    Emerging Clostridioides difficile strains belonging to PCR ribotype ...
    Aug 12, 2025 · End 2023, the UK Health Security Agency sent an alert about a new hypervirulent Clostridioides difficile PCR ribotype, ribotype 955 (RT955), ...
  38. [38]
    Effective Colonization by Nontoxigenic Clostridioides difficile REA ...
    Mar 28, 2023 · Colonization with nontoxigenic Clostridioides difficile strain M3 (NTCD-M3) has been demonstrated in susceptible hamsters and humans when administered after ...
  39. [39]
    The burden of CDI in the United States: a multifactorial challenge - NIH
    Mar 7, 2023 · Results of a recent meta-analysis reveals that C. difficile accounts for 20% of all antibiotic-associated diarrhea cases among hospitalized ...
  40. [40]
    [PDF] Clostridioides difficile Infection Summary Agent Transmission
    C. difficile accounts for 20–30% of antibiotic-associated diarrhea. Laboratory Diagnosis. Most diagnostic testing for C ...
  41. [41]
    Pseudomembranous colitis - Symptoms & causes - Mayo Clinic
    Sep 3, 2025 · The most common cause of pseudomembranous colitis is serious infection with a bacterium called Clostridioides difficile, also called C. diff.Overview · Symptoms · When to see a doctor · Causes
  42. [42]
    Pseudomembranous colitis attributable to Clostridioides difficile
    Clostridioides difficile is the primary causative agent of pseudomembranous colitis, a condition characterized by multiple episodes of watery diarrhea with ...Educational Case · Question/discussion Points... · Teaching Points
  43. [43]
    Clostridioides difficile infection - StatPearls - NCBI Bookshelf - NIH
    Apr 10, 2024 · This obligate anaerobic bacillus is recognized for its ability to produce toxins and cause diarrhea, which is often associated with antibiotic ...
  44. [44]
    An Update on Clostridioides difficile Binary Toxin - PMC - NIH
    Apr 27, 2022 · 20% of C. difficile strains produce a binary toxin (CDT) encoded by the tcdA and tcdB genes, which is thought to enhance TcdA and TcdB toxicity.
  45. [45]
    Non-human Clostridioides difficile Reservoirs and Sources - PubMed
    Soil seem to be the habitat of divergent unusual lineages of C. difficile. But the most important aspect of animals and environment is their role in C.Missing: primary | Show results with:primary
  46. [46]
    Clostridioides (Clostridium) difficile background Clostridioides ...
    Oct 5, 2022 · diff, is a gram positive, anaerobic, spore-forming, toxigenic bacterium that was first recognized as a cause of disease in 1978. It can be ...
  47. [47]
    the new epidemic of Clostridium difficile-associated enteric disease
    Nov 21, 2006 · Early work implicated Staphylococcus aureus, but in 1978 Clostridium difficile became the established pathogen in the vast majority of cases.
  48. [48]
    [PDF] Clostridioides difficile Infection: Preventing Transmission - CDC
    Cohen SH, Gerding DN, Johnson S, et al. Clinical Guidelines for Clostridium difficile Infection in. Adults: 2010 Update by the Society for Healthcare ...
  49. [49]
    The Role of Clostridioides difficile Within the One Health Framework
    Feb 16, 2025 · difficile strains known to cause disease in humans suggests that neonatal pigs may serve as a potential reservoir for human C. difficile ...
  50. [50]
    Genomic Delineation of Zoonotic Origins of Clostridium difficile
    Indeed, given the significant differences between C. difficile and some other pathogenic clostridia, it has been proposed that it be renamed Clostridioides ...
  51. [51]
    Phenotypic and genotypic antimicrobial resistance profiles of clinical ...
    Jul 10, 2025 · Purpose. Antimicrobial resistance (AMR) is important in the pathogenesis and spread of Clostridioides difficile infection (CDI).
  52. [52]
  53. [53]
    Clostridioides difficile Infection: Diagnosis and Treatment Challenges
    Jan 27, 2024 · The evolution of antibiotic resistance in C. difficile involves the acquisition of new resistance mechanisms, which can be shared among various ...
  54. [54]
    Correlating Antibiotic-Induced Dysbiosis to Clostridioides difficile ...
    C. difficile infection (CDI) occurs due to C. difficile overgrowth upon disruption of the host gut microbiome, causing the secretion of toxins (toxin A, or ...Abstract · Dysbiosis Enhances... · Methods
  55. [55]
    Bile acids as germinants for Clostridioides difficile spores, evidence ...
    This review summarizes current literature on the effects of bile acids and their metabolites on spore germination in the gut
  56. [56]
    Clostridioides difficile Spore Formation and Germination
    This review summarizes our current understanding of the mecha- nisms controlling C. difficile spore formation and germination and describes strategies for ...
  57. [57]
    Clostridium difficile Toxins: Mechanism of Action and Role in Disease
    By glucosylating small GTPases, TcdA and TcdB cause actin condensation and cell rounding, which is followed by death of the cell. TcdA elicits effects primarily ...
  58. [58]
    The role of toxins in Clostridium difficile infection - Oxford Academic
    Oct 18, 2017 · The pathogenesis of C. difficile is primarily mediated by the actions of two large clostridial glucosylating toxins, toxin A (TcdA) and toxin B (TcdB).INTRODUCTION · ROLE OF TcdA, TcdB AND... · CELLULAR EFFECTS OF...
  59. [59]
    Toxic megacolon associated Clostridium difficile colitis - PMC - NIH
    It produces toxins A and B, causing severe mucosal destruction and pseudomembrane formation[1-5].
  60. [60]
    Gut microbiome and Clostridioides difficile infection - NIH
    The gut microbiota performs a fundamental role in resisting the colonization of C. difficile and hence, perturbation of this microbial structure creates an ...
  61. [61]
    Clostridium difficile flagella predominantly activate TLR5-linked NF ...
    We demonstrate that the interaction of flagellin and TLR5 predominantly activates the NF-κB and, in a lesser degree, the MAPK pathways, via TLR5.
  62. [62]
    The interplay between host immunity and Clostridioides difficile ...
    In agreement with a role for MyD88 during CDI, the MyD88-dependent Toll-like receptors (TLRs) TLR2, TLR4, and TLR5 were reported to mediate immune responses ...
  63. [63]
    Toll-Like Receptor 5 Stimulation Protects Mice from Acute ...
    We demonstrate that restoring intestinal innate immune tone by TLR stimulation in antibiotic-treated mice ameliorates intestinal inflammation and prevents death ...
  64. [64]
    IL-8 release and neutrophil activation by Clostridium difficile toxin ...
    difficile toxins, and the conditioned media were harvested for cytokine and functional assays. Monocytes exposed to C. difficile toxin A (10(-10) M) or toxin B ...Missing: innate inflammasome
  65. [65]
    Clostridium difficile Toxin–Induced Inflammation and Intestinal Injury ...
    Our data clearly show that C difficile toxins trigger the inflammasome-dependent release of IL-1β, which contributes to toxin-induced inflammation and ...
  66. [66]
    Understanding host immune responses in Clostridioides difficile ...
    May 11, 2024 · Toxins released by C. difficile activate the pyrin inflammasome, initiating the processing and release of IL-1β, thereby augmenting the ...<|separator|>
  67. [67]
    Host Immune Responses to Clostridioides difficile: Toxins and Beyond
    Dec 20, 2021 · Clostridium difficile flagella predominantly activate TLR5-linked NF-κB pathway in epithelial cells. Anaerobe 38, 116–124. doi: 10.1016/j ...
  68. [68]
    The interplay between host immunity and Clostridioides difficile ...
    Jul 1, 2025 · Clostridium difficile toxin-induced inflammation and intestinal injury are mediated by the inflammasome. Gastroenterology 139:542–552. Go to ...
  69. [69]
    Progranulin protects against Clostridioides difficile infection by ...
    Sep 30, 2024 · Our findings thus indicate a critical role for PGRN-promoted CD4 + T cell IL-22 production in shaping gut immunity and reestablishing the intestinal barrier ...
  70. [70]
    Human C. difficile toxin–specific memory B cell repertoires encode ...
    Jul 14, 2020 · We report that TcdB-specific memory B cell–encoded antibodies showed somatic hypermutation but displayed limited isotype class switch.
  71. [71]
    Effect of Aging on Host Response to Clostridium difficile Infections
    Jan 25, 2016 · Aging has been identified in the literature as a risk factor for CDI as well as adverse outcome from CDI.
  72. [72]
    Age-associated defects in the adaptive immune response to ...
    Age-associated defects in the adaptive immune response to Clostridium difficile infection impair the development of a protective immune response in the elderly.
  73. [73]
    Clostridium difficile infection in older adults - PMC - NIH
    Older adults' increased risk of recurrent C. difficile infections probably reflects immune senescence and age-related changes in the gut microbiome, as ...
  74. [74]
    Impaired Th17 immunity in recurrent C. difficile infection is ... - medRxiv
    Jun 10, 2020 · These data suggest that effective T cell immunity to C. difficile requires the development of Th17 cells.Missing: Tregs | Show results with:Tregs
  75. [75]
    Monoclonal antibody-mediated neutralization of Clostridioides ...
    Monoclonal antibodies (mAbs) that neutralize Toxin A and Toxin B, actoxumab and bezlotoxumab, respectively, significantly reduce disease severity.
  76. [76]
    Bezlotoxumab prevents extraintestinal organ damage induced by ...
    Aug 31, 2022 · Administration of bezlotoxumab during C. difficile infection prevents systemic disease and thymic atrophy, without blocking gut damage.
  77. [77]
    Safety and Immunogenicity of an Adjuvanted Clostridioides difficile ...
    Oct 24, 2024 · The GSK C. difficile vaccine candidate was immunogenic, especially when given with AS01, and was well tolerated with an acceptable safety profile.
  78. [78]
    A multivalent mRNA-LNP vaccine protects against Clostridioides ...
    Oct 3, 2024 · A recent Pfizer phase 3 clinical trial showed promise in reducing the severity and duration of CDI but failed to prevent initial infection, ...
  79. [79]
    Clostridium difficile as a potent trigger of colorectal carcinogenesis
    May 24, 2025 · Clostridium difficile, traditionally recognized as a cause of antibiotic-associated colitis, has emerged as a potential oncogenic factor in colorectal cancer ( ...
  80. [80]
  81. [81]
    Human Colon Cancer–Derived Clostridioides difficile Strains Drive ...
    Aug 5, 2022 · The patient-derived Clostridioides difficile bacterial strain was found to drive colon tumorigenesis through its toxin TcdB, which induced ...
  82. [82]
    Prevention - CDC Stacks
    ... associated diarrhea, resulting in increased incidence of community-associated infection ... Patients commonly develop antibiotic-related diarrhea with C difficile ...Missing: percentage | Show results with:percentage
  83. [83]
    [PDF] Clostridioides difficile Infection - CDC Stacks
    Oct 2, 2019 · This article provides updated recommendations regarding the prevention, diagnosis, and treatment of incident and recurrent C difficile infection ...Missing: percentage | Show results with:percentage
  84. [84]
    Effectiveness of alcohol-based hand rubs for removal of ... - PubMed
    Alcohol is not effective against Clostridium difficile spores. We examined the retention of C. difficile spores on the hands of volunteers after ABHR use ...
  85. [85]
    [PDF] Clostridioides difficile infection, 2019 - CDC
    Emerging Infections Program, Healthcare-Associated. Infections – Community Interface Surveillance Report, Clostridioides difficile infection (CDI), 2019.
  86. [86]
    Environmental and Health Care Personnel Transmission of C ...
    Apr 4, 2025 · On average, we recovered C difficile from 1.89% (95% CI, 1.59%-2.20%) of HCP hands per occupied room per day. We found lower rates of toxigenic ...Missing: percentage | Show results with:percentage
  87. [87]
    Characterization of Adult and Pediatric Healthcare-Associated and ...
    May 8, 2025 · Pediatric HA CDI rates decreased by 29.6%, from 4.52 to 3.18 cases/10,000 patient-days (p = 0.003), and CA CDI rates decreased by 58.3%, from ...
  88. [88]
    Study Suggests C. Difficile May Be a Foodborne Illness
    Apr 17, 2023 · Patients who are vulnerable to Clostridioides difficile (C. difficile) infection (CDI) should not consume or handle shellfish or pork, according to a recent ...
  89. [89]
    Prevalence of Clostridium difficile in retail pork | Request PDF
    Aug 6, 2025 · Clostridium difficile was isolated from 1.8% (7/393) of retail pork samples obtained from 4 Canadian provinces.<|control11|><|separator|>
  90. [90]
    Detection of Clostridium difficile in Feces of Asymptomatic Patients ...
    Approximately 5 to 15% of newly hospital admitted patients carry C. difficile in their feces (4, 5, 14–17). Carriage rates of residents in long-term-care ...Missing: Clostridioides | Show results with:Clostridioides<|control11|><|separator|>
  91. [91]
    Screening for Asymptomatic Clostridioides difficile Carriage Among ...
    Sep 13, 2023 · Asymptomatic detection of C. difficile is reported in ~ 10-20% of admitted patients. Risk factors for carriage include recent hospitalization, ...Missing: percentage | Show results with:percentage
  92. [92]
    Clostridium (Clostridioides) difficile in animals - J. Scott Weese, 2020
    Jan 6, 2020 · It is a potentially important zoonotic pathogen, but there is limited direct evidence of animal–human transmission. Although C. difficile has ...
  93. [93]
    Identification and Characterization of Escherichia coli, Salmonella ...
    May 27, 2019 · C. difficile was isolated from three (3.9%) reptiles. Two of these isolates were toxigenic and classified into ribotypes/MLST 081/ST9 and 106/ ...
  94. [94]
    Low Clostridioides difficile positivity rate in wild animal shelter in ...
    Positivity rate in wild animal shelter was low (4/22 animals). · C. difficile was isolated only from birds. · Common C. difficile ribotypes can be present in ...
  95. [95]
    The housefly Musca domestica as a mechanical vector of ... - PubMed
    This study describes the potential for M. domestica to contribute to environmental persistence and spread of C. difficile in hospitals, highlighting flies ...
  96. [96]
    The burden of CDI in the United States: a multifactorial challenge
    Mar 7, 2023 · difficile infections (rCDI); up to 35% of index CDI will recur and of these up to 60% will further recur with multiple recurrences observed. The ...
  97. [97]
    C. difficile infection - Symptoms and causes - Mayo Clinic
    Sep 1, 2023 · A bacterium that causes an infection of the colon, the longest part of the large intestine. Symptoms can range from diarrhea to life-threatening damage to the ...Missing: authoritative sources
  98. [98]
    Clostridioides difficile infection (CDI) - Vaccines Europe
    Nearly 124,000 healthcare-associated C. difficile infections (CDIs) occur annually in acute care hospitals in the EU/EEA, and 3,700 deaths.
  99. [99]
    Pilot study of Clostridioides difficile infection (CDI) in hospitals, Italy ...
    Jan 9, 2025 · In the Italian hospitals, the mean overall CDI and HA-CDI rates were higher than those reported in a previous study in the EU/EEA countries and ...
  100. [100]
    COVID-19 & Antimicrobial Resistance - CDC
    Feb 5, 2025 · Antimicrobial-resistant infections and Clostridioides difficile ... In nursing homes, antibiotic use spiked alongside surges of COVID-19 cases but ...
  101. [101]
    C. difficile cases increase by one-third over three years, government ...
    May 12, 2025 · In a research and analysis paper, published on 1 May 2025, the UKHSA revealed that CDI cases increased by 33% between the 2020/2021 and 2023/ ...
  102. [102]
    UK health warning after cases of deadly bacteria C. diff soar to 12 ...
    Jul 11, 2025 · There were 19,239 cases of C. diff reported to the UK Health Security Agency (UKHSA) from February 2024 to January 2025. This is the highest ...
  103. [103]
    Dominance of toxigenic Clostridioides difficile strains and the ...
    Aug 11, 2025 · The emergence of RT955, closely related to a UK epidemic strain, suggests a possible shared origin and epidemiological link. Keywords: ...3. Results · Table 1 · 3.2. Microbiological...
  104. [104]
  105. [105]
    Community-acquired Clostridium difficile infection: an increasing ...
    Recent reports have corroborated the fear that new strains are emerging with non-027 and non-078 “hypervirulent” strains causing severe infection in both the ...
  106. [106]
    Clostridioides (formerly Clostridium) difficile–Induced Colitis
    Clostridioides difficile (C. difficile)–induced colitis is an inflammation of the large intestine (colon) that results in diarrhea.
  107. [107]
    Clostridioides difficile Infection: A Clinical Review of Pathogenesis ...
    Dec 27, 2023 · Clostridioides difficile is a gram-positive bacillus spore-forming obligate anaerobe. It is commonly found in soil, water, and human and animal ...
  108. [108]
    C. diff: Facts for Clinicians - CDC
    Mar 5, 2024 · Any surface, device or material that becomes contaminated with feces could serve as a reservoir for the C. diff spores. Examples include:.Missing: survival | Show results with:survival
  109. [109]
    Clinical Practice Guidelines for Clostridium difficile Infection ... - IDSA
    Feb 15, 2018 · This guideline updates recommendations regarding epidemiology, diagnosis, treatment, infection prevention, and environmental management.Missing: renamed | Show results with:renamed
  110. [110]
    Clostridioides difficile Infection: Update on Management - AAFP
    Feb 1, 2020 · The presence of peripheral leukocytosis, lactic acidosis, or hypoalbuminemia indicates more severe C. difficile infection. Adjuvant tests ...
  111. [111]
    A Case of Fulminant Clostridioides Difficile Infection - LWW
    Although fulminant CDI occurs infrequently, it is associated with mortality rates of 30%–40%. ... FMT remains an effective treatment for FCDI, with studies ...
  112. [112]
    Extraintestinal Clostridioides difficile infection - ScienceDirect.com
    Extra-intestinal manifestations of Clostridioides (Clostridium) difficile infection (CDI) (including bacteremia and tissue infection) are extremely rare.
  113. [113]
    Predictors of First Recurrence of Clostridium difficile Infection - NIH
    Symptomatic recurrence of Clostridium difficile infection (CDI) occurs in approximately 20% of patients and is challenging to treat. Identifying those at high ...Results · Recurrence Rates · Relapse Versus Reinfection
  114. [114]
    Prevalence and duration of asymptomatic Clostridium difficile ...
    Previous studies suggested that 7 to 15% of healthy adults are colonized with toxigenic Clostridium difficile.
  115. [115]
    Clinical Testing and Diagnosis for C. diff Infection - CDC
    Mar 6, 2024 · FDA-approved PCR assays are same-day tests that are highly sensitive and specific for the presence of a toxin-producing C. diff organism.
  116. [116]
  117. [117]
    Laboratory Tests for the Diagnosis of Clostridium difficile - PMC - NIH
    Although CCCNA is considered as a reference method, and is a sensitive method for the detection of toxin B, clinical sensitivity is <90% ( Table 1 ). This is ...
  118. [118]
    Clostridioides difficile infection in adults: Clinical manifestations and ...
    Aug 26, 2025 · Clostridioides difficile is a spore-forming, toxin-producing, and gram-positive anaerobic bacterium that causes antibiotic-associated colitis.
  119. [119]
    Clinical Practice Guidelines for the Management of Clostridioides ...
    Jun 14, 2021 · This clinical practice guideline is a focused update on management of Clostridioides difficile infection (CDI) in adults specifically addressing the use of ...Recommendations (Abridged) · Methodology · Full Recommendation on the...
  120. [120]
    Idorsia's breakthrough synthetic glycan vaccine platform validated ...
    Jun 30, 2025 · In addition to the C. difficile vaccine that is in Phase 1 clinical development, Idorsia is also developing synthetic glycan vaccines for ...
  121. [121]
    Phage therapy for Clostridioides difficile infection - Frontiers
    In this review, we summarize the importance of phage therapy against C. difficile, and describe a novel next-generation phage therapy developed using ...Missing: trials | Show results with:trials
  122. [122]
    Clinical Guidance for C. diff Infection Prevention in Acute Care ...
    Mar 8, 2024 · Isolate and initiate contact precautions for suspected or confirmed Clostridioides (formerly known as Clostridium) difficile infection (CDI).
  123. [123]
    Preventing C. diff - CDC
    Dec 18, 2024 · C. diff is a germ that causes diarrhea and colitis (inflammation of the colon). It can be life-threatening. View All ...Key Points · Stop The Spread · Kill C. Diff Germs At HomeMissing: symptoms | Show results with:symptoms<|control11|><|separator|>
  124. [124]
    Current practices to reduce the risk of transmission of Clostridioides ...
    Oct 9, 2025 · In healthcare settings, interventions like contact precautions (CP), hand hygiene with soap and water, sporicidal environmental cleaning, and ...<|control11|><|separator|>
  125. [125]
    Strategies to prevent Clostridioides difficile infections in acute-care ...
    Implementing an antibiotic stewardship program: guidelines by the Infectious Diseases Society of America and the Society for Healthcare Epidemiology of ...
  126. [126]
    The Effect of Saccharomyces boulardii Primary Prevention on Risk ...
    Saccharomyces boulardii administered to hospitalized patients prescribed antibiotics frequently linked with HO-CDI was associated with a reduced risk of HO-CDI.
  127. [127]
    The American College of Gastroenterology recommends against ...
    Oct 19, 2021 · The American College of Gastroenterology recommends against use of probiotics for primary or secondary prevention of C. difficile. Prof. Daniel ...
  128. [128]
    Bezlotoxumab for Prevention of Recurrent Clostridium difficile Infection
    Jan 26, 2017 · The monoclonal antibody AZD5148 confers broad protection against TcdB-diverse Clostridioides difficile strains in mice, PLOS Pathogens, 21 ...
  129. [129]
    CLOVER (CLOstridium difficile Vaccine Efficacy tRial) Study: A ...
    The phase 3 CLOVER study assessed the PF-06425090 vaccine for primary Clostridioides difficile infection (CDI) prevention in adults. Although the primary e.
  130. [130]
    Clostridioides difficile Infection (CDI) Surveillance | HAIs - CDC
    Jun 24, 2025 · The Clostridioides difficile infection (CDI) Surveillance Program collects data for describing incidence and trends of these infections.Explore The Data · About The Data · Eip Sites Conducting...Missing: distribution | Show results with:distribution
  131. [131]
    Clostridioides difficile infection: Prevention and control - UpToDate
    Aug 26, 2025 · - Surveillance · - Prevention strategies · Early detection and isolation · Contact precautions · Hand hygiene · Environmental cleaning and ...<|control11|><|separator|>