Hostname: page-component-8448b6f56d-xtgtn Total loading time: 0 Render date: 2024-04-19T21:17:47.065Z Has data issue: false hasContentIssue false

Bring it on: Top five antimicrobial stewardship challenges in transplant infectious diseases and practical strategies to address them

Published online by Cambridge University Press:  27 April 2022

Miranda So
Affiliation:
Sinai Health-University Health Network Antimicrobial Stewardship Program, University Health Network, Toronto, Ontario, Canada Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
Helen Tsai
Affiliation:
Division of Infectious Diseases, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, United States
Neeraja Swaminathan
Affiliation:
Division of Infectious Diseases, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, United States
Rachel Bartash*
Affiliation:
Division of Infectious Diseases, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, United States
*
Author for correspondence: Rachel Bartash, MD, Infectious Diseases, Montefiore Medical Center, Albert Einstein College of Medicine, 3411 Wayne Avenue, Suite 4H, Bronx, NY10467. E-mail: rbartash@montefiore.org

Abstract

Antimicrobial therapies are essential tools for transplant recipients who are at high risk for infectious complications. However, judicious use of antimicrobials is critical to preventing the development of antimicrobial resistance. Treatment of multidrug-resistant organisms is challenging and potentially leads to therapies with higher toxicities, intravenous access, and intensive drug monitoring for interactions. Antimicrobial stewardship programs are crucial in the prevention of antimicrobial resistance, though balancing these strategies with the need for early and frequent antibiotic therapy in these immunocompromised patients can be challenging. In this review, we summarize 5 frequently encountered transplant infectious disease stewardship challenges, and we suggest strategies to improve practices for each clinical syndrome. These 5 challenging areas are: asymptomatic bacteriuria in kidney transplant recipients, febrile neutropenia in hematopoietic stem cell transplantation, antifungal prophylaxis in liver and lung transplantation, treatment of left-ventricular assist device infections, and Clostridioides difficile infection in solid-organ and hematopoietic stem-cell transplant recipients. Common themes contributing to these challenges include limited data specific to transplant patients, shortcomings in diagnostic testing, and uncertainties in pharmacotherapy.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction in any medium, provided the original article is properly cited.
Copyright
© The Author(s), 2022. Published by Cambridge University Press on behalf of The Society for Healthcare Epidemiology of America

Antimicrobial agents are life-saving medications for immunocompromised hosts who rely heavily on these medications. However, antimicrobial resistance (AMR) is a significant threat to these patients and, as such, judicious use of antimicrobials is critical. Antimicrobial stewardship (AMS) programs are essential in creating treatment guidelines as well as promoting and monitoring appropriate antimicrobial treatment in these complex patients. We discuss 5 important, frequently encountered transplant infectious disease stewardship challenges, with suggested strategies to address and improve antimicrobial practices for each clinical syndrome. Although each syndrome has its unique set of challenges, we discuss overarching themes of scarcity of data on stewardship interventions specific to transplant patients, limitations in diagnostic testing, and pharmacotherapy concerns.

1. Asymptomatic bacteriuria in renal transplant recipients

Asymptomatic bacteriuria (ASB), a common condition in renal transplant recipients, is often treated with antibiotics Reference Coussement, Maggiore and Manuel1 based on the theoretical risk of ascending infection leading to pyelonephritis and acute graft loss. However, data against routine treatment of ASB are fairly robust. The Infectious Diseases Society of America (IDSA) and the American Society for Transplantation (AST) updated their clinical care guidelines in 2019, recommending against treating ASB in renal transplant recipients >2 months after transplant. Reference Nicolle, Gupta and Bradley2,Reference Goldman and Julian3 These guidelines reflected results from several limited retrospective studies Reference Kotagiri, Chembolli, Ryan, Hughes and Toussaint4Reference Lee, Bang and Dadhania6 and a single randomized controlled trial Reference Origüen, López-Medrano and Fernández-Ruiz7 that found no significant differences in outcomes for patients who received antibiotics for ASB compared to those who did not. Following publication of these guidelines, additional randomized controlled trials and 1 meta-analysis have provided additional evidence that treating ASB does not offer benefit Reference Coussement, Kamar and Matignon8Reference Coussement, Kamar and Abramowicz10 but leads to excessive antibiotic use and increased risk of infection with multidrug-resistant organisms. Reference Coussement, Kamar and Matignon8 Despite strong evidence, ASB continues to be a stewardship challenge in renal transplant recipients.

Data on ASB outcomes in renal transplant recipients within 2 months of transplant or with anatomic genitourinary abnormalities, indwelling catheters, or ureteral stents, remains limited and guidelines do not make strong recommendations for these populations. Reference Nicolle, Gupta and Bradley2,Reference Goldman and Julian3 Many providers may favor treating ASB in these circumstances Reference Coussement, Maggiore and Manuel1 because of concerns that foreign material, intense immunosuppression, or genitourinary tract abnormalities could potentiate the risk of ASB progressing to graft pyelonephritis, although the benefit is unclear. Reference Coussement, Kamar and Abramowicz10

Stewardship efforts are further hindered by the diagnostic complexities of distinguishing ASB from urinary tract infections. In renal transplant recipients with nonspecific signs of infection but no clear urinary symptoms, positive urine cultures are difficult to interpret because denervation from surgery may limit the presence of urinary symptoms. Reference Goldman and Julian3,Reference Coussement, Kamar and Abramowicz10 Pyuria and positive urine cultures in the setting of chronic indwelling catheters or stents often reflect colonization or contamination rather than a UTI. Similarly, acute kidney injury and pyuria can be seen in both graft rejection and UTIs. Diagnostic ambiguities likely contribute to ASB overtreatment despite lack of clinical significance.

The use of other markers of infection (eg, white bold cell count, C- reactive protein level, and erythrocyte sedimentation rate) may be useful in differentiating asymptomatic bacteriuria from urinary tract infection in the setting of nonspecific clinical symptoms, but additional studies are needed. Improved biomarkers and diagnostic testing to discern the relevancy of positive urine cultures and to identify which renal transplant recipients would benefit from antibiotics can hopefully augment stewardship efforts in the future, but these are currently unavailable.

Moreover, whether ASB guidelines are reflected in clinical practice is unclear. In a survey study of European transplant centers, >70% reported routine screening for bacteriuria and treatment was common. Reference Coussement, Maggiore and Manuel1 One solution to limiting the treatment of ASB is avoiding routine surveillance cultures in the absence of symptoms or laboratory abnormalities because providers may be inclined to treat known positive results. Updating institutional treatment guidelines to include avoiding treatment and providing prescriber feedback are AMS tools that can help decrease the treatment of ASB.

2. Febrile neutropenia in stem-cell transplant recipients

Hematopoietic stem cell transplant (HSCT) recipients are vulnerable to infectious complications, especially during the pre-engraftment period in which significant neutropenia and mucosal damage increase the risk of bacteremia. Reference Dhakal, Giri and Levin11Reference Lind, Mooney and Carone13 HSCT recipients often receive several weeks of broad-spectrum antimicrobials to mitigate this risk. However, this extensive antimicrobial exposure, combined with prior antimicrobial therapy and conditioning chemotherapy, can contribute to gut dysbiosis and poor outcomes. Reference Shallis, Terry and Lim14,Reference Taplitz, Kennedy and Flowers15 AMR has been recognized as a leading cause of death globally, with carbapenem-resistant Enterobacterales (CRE) and vancomycin-resistant enterococci (VRE) emerging as major threats to HSCT recipients. Reference Murray, Ikuta and Sharara16Reference Kamboj, Cohen and Huang19 Although AMS has been advocated for patients with hematological malignancies and HSCT, clinicians have to balance maintaining adequate antimicrobial coverage against minimizing unnecessary antimicrobial exposure at the individual and population level. Reference Averbuch, Orasch and Cordonnier20Reference Gyssens, Kern and Livermore23 Strategies to guide antimicrobial use in the HSCT population are urgently needed.

One area of stewardship interest is antimicrobial use for febrile neutropenia in HSCT recipients. Routinely, broad-spectrum coverage is maintained until absolute neutrophil count recovers to >500 cells/µL and the patient is afebrile, irrespective of the presence of a documented infection or fever of unknown origin. Reference Freifeld, Bow and Sepkowitz24 Antimicrobial prescribing practice in HSCT patients with febrile neutropenia varies widely; it informs and is informed by regional epidemiology. Reference Verlinden, Mikulska and Knelange25

Recent data suggest that a shorter duration of antibiotic therapy is safe and effective for febrile neutropenia when coupled with close monitoring. The “How Long” study compared the conventional approach to high-risk febrile neutropenia with early discontinuation of broad-spectrum antimicrobials based on resolution of fever after 72 hours and clinical recovery. Reference Aguilar-Guisado, Espigado and Martín-Peña26 Patients in the experimental arm received a significantly shorter duration of antimicrobials with numerically fewer adverse events, indicating that the symptom-driven approach avoided unnecessary antimicrobial exposure. Reference Aguilar-Guisado, Espigado and Martín-Peña26 Although this study included HSCT patients, allogeneic recipients accounted for only 9% of the study population. Reference Aguilar-Guisado, Espigado and Martín-Peña26 Nevertheless, these findings show that in fever of unknown origin, shortening duration of empirical antimicrobial therapy with close monitoring can be safe and feasible. Two recent studies with a similar intervention and a larger representation of HSCT patients reported congruous findings, but well-designed, prospective studies are imperative to supporting practice changes in the era of AMR. Reference Le Clech, Talarmin and Couturier27Reference Stern, Carrara, Bitterman, Yahav, Leibovici and Paul29

In patients with documented infections, the decision to tailor therapy targeting the pathogen versus continuing with broad-spectrum antimicrobials has not been fully elucidated. One guideline recommended a patient-specific approach, which may lead to wide variability in antimicrobial prescribing. Reference Baden, Bensinger and Angarone30 A multinational, prospective, longitudinal study of patients with high-risk febrile neutropenia, including acute leukemia and HSCT (autologous and allogeneic) recipients, evaluated the association between bacteremia and mortality at 7 days and 30 days. Reference Weisser, Theilacker and Tschudin Sutter31 P. aeruginosa bacteremia was associated with the highest 7-day and 30-day mortality at 16.7% and 26.7%, respectively, compared to coagulase-negative staphylococci (2%) or streptococci (<1%), whereas enterococci were associated with an unexpected increase in mortality. Reference Kern, Roth and Bertz32 Candidemia and gram-negative bacteremia were independently associated with intensive care unit admission. Reference Kern, Roth and Bertz32 In a related study, predictors of bacteremia due to multidrug-resistant P. aeruginosa included prior exposure to piperacillin-tazobactam, antipseudomonal carbapenem, fluoroquinolone prophylaxis, underlying hematological disease, and presence of a urinary catheter. Reference Gudiol, Albasanz-Puig and Laporte-Amargós33 These data suggest that a pathogen-specific approach combined with judicious use of broad-spectrum antimicrobials may be optimal. Although prophylactic fluoroquinolones have been routinely recommended for patients with acute leukemia and HSCT to reduce the rates of bacteremia, a more thoughtful, risk-stratified approach should be considered given its implications for institutional epidemiology. Reference Taplitz, Kennedy and Flowers15,Reference Egan, Robinson and Martinez34Reference Mikulska, Averbuch and Tissot36

Implementing an institution-specific guideline for management of neutropenic fever in hematology-oncology patients that accounts for local susceptibility patterns is a recommended AMS intervention. Reference Barlam, Cosgrove and Abbo37,Reference Barreto, Aitken and Krantz38 To ensure sustained practice change, evaluating the quality of antimicrobial prescribing through auditing and provision of feedback to prescribers will help AMS programs identify areas for improvement and provide ongoing support for guideline-adherent practices in hematology patients and HSCT recipients. Reference Douglas, Hall and James39 In the management of high-risk febrile neutropenia, data supporting judicious prescribing with close monitoring of patients with fever of unknown origin are encouraging. A multipronged approach with risk stratification, implementation of local guidelines, and evaluation of quality of antimicrobial prescribing offers a potential solution that may overcome the challenges of AMS in this vulnerable patient population.

3. Antifungal prophylaxis in liver and lung transplant recipients

Antifungal prophylaxis has been advocated for SOT recipients because diagnostic limitations for invasive fungal infections can translate into treatment delays that confer significant morbidity and mortality. Reference Hosseini-Moghaddam, Ouédraogo and Naylor40 Stewardship challenges involving antifungal prophylaxis in liver transplant recipients include pharmacokinetic considerations and the availability of local epidemiological patterns of fungal infections. In lung transplant recipients, inhaled amphotericin or systemic azole therapy are used because invasive aspergillosis is a significant concern. However, stewardship efforts are hindered by the absence of strong evidence, and a better understanding of risk is needed.

Targeted rather than universal antifungal prophylaxis based on a risk stratification approach outlined by the AST is preferred for liver transplant recipients, but prospective studies to guide treatment duration are lacking. Reference Aslam and Rotstein41 Withholding antifungal prophylaxis in low-risk liver transplant recipients has been shown to be safe, to reduce unnecessary exposure, and to avoid potential drug–drug interactions with immunosuppressants. Reference Aslam and Rotstein41,Reference Lavezzo, Stratta and Ballaris42 In a 2008 study, 28% and 72% of North American transplant centers surveyed used universal and targeted prophylaxis, respectively. Reference Singh, Wagener, Cacciarelli and Levitsky43 More recent data on prescribing trends are lacking, but regular institutional review of prescribing patterns in low-risk recipients is a realistic AMS tool to ensure that evidence-based practice is followed and that antifungal overuse is limited.

The various antifungal agents available for targeted prophylaxis in high-risk liver transplant recipients have notable limitations. Liposomal amphotericin B is effective, Reference Fortún, Martín-Davila and Moreno44 but it offers unnecessarily broad coverage, it is costly, and it is limited to intravenous administration. Fluconazole, the preferred agent based on expert opinion, Reference Aslam and Rotstein41 is faced with rising resistance, increasing rates of non-albicans Candida spp infections, Reference Fortún, Muriel and Martín-Dávila45 and known interactions with calcineurin inhibitors. A single-center study demonstrated that fixed fluconazole dosing was effective, and no invasive fungal infections with reduced fluconazole-susceptible strains occurred. Reference Jorgenson, Descourouez and Marka46 However, applicability across institutions with different local epidemiologies and among critically ill patients with renal dysfunction (in whom fluconazole pharmacokinetics are variable) are concerns. Reference Muilwijk, de Lange and Schouten47 Echinocandins are associated with fewer toxicities and drug–drug interactions, but there are significant pharmacokinetic–pharmacodynamic limitations. Echinocandins achieve limited therapeutic concentrations intra-abdominally because of their molecular characteristics, Reference Gatti, Rinaldi and Ferraro48 predisposing patients to the emergence of echinocandin resistance. Reference Shields, Nguyen, Press and Clancy49 An 8% acquired resistance rate Reference Prigent, Aït-Ammar and Levesque50 and breakthrough invasive fungal infections Reference Gatti, Rinaldi and Ferraro48 while on echinocandin therapy have been noted. To help prevent the emergence of fluconazole and echinocandin resistance, AMS programs can assist in optimizing dosing for patients who are critically ill, who require renal replacement therapy, or who have infections at sites of known poor drug penetration. Reference Muilwijk, de Lange and Schouten47,Reference Gatti, Rinaldi and Ferraro48 The risk of invasive aspergillosis in high-risk liver recipients is another crucial consideration. Reference Lavezzo, Romagnoli, Balagna and De Rosa51 The decision to administer antimold coverage involves weighing the local incidence of and a recipient’s risk for invasive aspergillosis Reference Lum, Lee, Vu, Strasser and Davis52 against potential toxicities, drug–drug interactions, and emergence of azole-resistant Aspergillus. This challenging situation underscores the importance of updated local epidemiological data and of longitudinal monitoring of fungal susceptibilities, outcomes, and adverse events to form AMS strategies that can be feasibly adopted in clinical practice. Reference Johnson, Lewis and Dodds Ashley53,Reference Khanina, Urbancic and Haeusler54

For lung transplant recipients, the preferred choice between universal or pre-emptive antifungal prophylaxis against Aspergillus (the latter involving routine surveillance with broncho-alveolar lavage culture and galactomannan) is undefined, but either approach is recommended over no prophylaxis. Reference Husain and Camargo55,Reference De Mol, Bos and Beeckmans56 Although 90% of US transplant centers had previously reported routine universal antifungal prophylaxis for lung transplant recipients, a review of administrative claims data showed that only 41.5% of patients received antifungal prophylaxis. Reference Pennington, Baqir, Erwin, Razonable, Murad and Kennedy57 The reasons for this incongruence is unknown, but further analysis could potentially provide insights to stewardship areas of interest. An understanding of the current practices would help direct AMS efforts toward high-yield measures. Data on universal and pre-emptive therapies are mixed because studies are limited by small sample size, variable study design, and heterogeneous immunosuppression and antifungal agents included. 58 Prospective studies comparing universal and pre-emptive prophylaxis are needed not only to evaluate efficacy but also to characterize potential stewardship benefits of pre-emptive prophylaxis.

If universal prophylactic therapy for lung transplant recipients is employed, the recommended duration of prophylaxis is 4–6 months. Nevertheless, 22.2% of transplant centers in one survey continued universal prophylaxis for >12 months. Reference Pennington, Yost, Escalante, Razonable and Kennedy59 The use of long-term or lifelong azole prophylaxis has not been shown to alter the incidence of invasive fungal infection in lung transplant recipients, even in the setting of therapeutic azole levels, and it is associated with medication toxicities, healthcare costs, and potential resistance. Reference Chong, Kennedy, Hathcock, Kremers and Razonable60 Another area of stewardship concern is whether routine prophylaxis is driving the emergence of delayed aspergillosis and invasive fungal infections in lung transplant recipients after prophylaxis is discontinued, beyond the traditional risk period. Reference Vazquez, Vazquez-Guillamet, Suarez, Mooney, Montoya and Dhillon61,Reference Bae, Lee and Jo62 One center reported the median time of onset for invasive aspergillosis in lung transplant recipients to be 363 days. Reference Vazquez, Vazquez-Guillamet, Suarez, Mooney, Montoya and Dhillon61 The incidence and consequences of invasive aspergillosis occurring beyond the first year have not been clearly established, even though immunosuppression may be less intensive and the risk of anastomotic fungal infection or ulcerative tracheobronchitis may be lower.

Perhaps a more tailored approach to antifungal prophylaxis in lung transplant recipients is necessary. Evidence-based risk stratification models to identify recipients who would benefit from a short course versus a standard course or from lifelong antifungal prophylaxis, relative to local incidence of invasive fungal infection, would be valuable for AMS programs. We also suggest that AMS programs monitor closely for a potential risk in delayed invasive aspergillosis and that they analyze any occurrences because such cases will have significant local and population-level stewardship implications.

4. Left ventricular assist device infections

Infections, a leading complication of left ventricular assist devices (LVADs), are estimated to occur in nearly 40% of recipients. Reference Zinoviev, Lippincott, Keller and Gilotra63 Despite the high incidence, management guidelines for LVAD infections are based on observational data and expert opinion due to the absence of randomized controlled trials. Reference Kusne, Mooney and Danziger-Isakov64 The lack of strong evidence, along with diagnostic complexities and uncertain effects of LVADs on antimicrobial pharmacokinetics, contribute to the challenges facing AMS in LVAD-specific and related infections. Compounding these issues are the nature of the infections, which are potentially incurable without source control through transplantation, and the growing proportion of LVADs implanted for destination therapy. Reference Molina, Shah, Kiernan and Cornwell65 In 2019, 73.1% of LVADs implanted were for destination therapy. Reference Molina, Shah, Kiernan and Cornwell65 Because LVADS are increasingly used for destination therapy, these infectious complications will be a growing challenge for AMS.

Device driveline infections, which account for 12%–35% of all LVAD-specific or related infections Reference Zinoviev, Lippincott, Keller and Gilotra63 , occur most frequently but diagnosing and distinguishing superficial from deep infection is problematic. Clinical and physical exam features of driveline or endovascular infections can be subtle, nonspecific, or absent. Reference Blanco-Guzman, Wang, Vader, Olsen and Dubberke66,Reference Koval and Stosor67 Imaging to assist in diagnosis is not standardized and has limitations: Computed tomography has variable performance and is affected by device artifact, ultrasound detects only superficial fluid collections, and access to FDG-PET may be a barrier, Reference Koval and Stosor67 although gallium single-photon emission computed tomography (SPECT) appears to be a promising imaging modality. Reference Puius, Parkar and Tlamsa68

Even if the extent of infection is successfully diagnosed, uncertainties remain regarding duration of therapy and the role of chronic antimicrobial suppression (CAS). The evidence for the current treatment duration recommendations for superficial and deep driveline infections is limited Reference Kusne, Mooney and Danziger-Isakov64,Reference Koval and Stosor67 and, in clinical practice, widely variable. Reference Koval and Stosor67 Whether superficial driveline infections progress to deeper infections or if CAS for driveline infections significantly reduces recurrence remains to be determined. Conflicting data are likely driven by the various LVAD-specific infections included in each study. Several studies estimate a 30% failure rate of CAS, Reference Radcliffe, Doilicho, Niu and Grant69Reference Hamad, Blanco-Guzman and Olsen71 with recurrence even in superficial driveline infections. Reference Jennings, Chopra, Chambers and Morgan70 One study has suggested that CAS resulted in no significant difference in the proportion of patients with relapse. Reference Hamad, Blanco-Guzman and Olsen71 For an infection associated with foreign material that may not be reasonably removed and that can occur in LVAD recipients who have altered immune responses, Reference Koval and Stosor67 the implications of these retrospective data on clinical practice are unclear.

In addition to ongoing questions regarding duration of therapy for LVAD infections, 2 studies have suggested altered intravenous vancomycin pharmacokinetics from LVADs, which may further complicate AMS efforts in these patients. Those with LVADs had a significantly higher incidence of supra-therapeutic trough levels, potentially due to an overestimated volume of distribution and rate of elimination. Reference Hall, Athans, Wanek, Wang, Estep and Williams72,Reference Jennings, Makowski, Chambers and Lanfear73 Further characterization of this finding and its implications are important because S. aureus and coagulase-negative Staphylococcus are the predominant etiological agents of LVAD infections.

The challenges facing AMS in LVAD infections are driven by the need for improved diagnostics and well-designed studies on treatment. In the absence of heart transplantation, determining the therapy end point is complex and requires careful consideration of a patient’s clinical, microbiological, radiographic, and surgical factors. The role for CAS remains ambiguous due to the limited evidence on efficacy and adverse effects. As more individuals receive LVADs for destination therapy, studies describing long-term outcomes of CAS categorized by each type of LVAD infection and pathogen involved are needed to assist in optimizing antimicrobial use.

5. Clostridioides difficile infection

Clostridioides difficile infection (CDI) is a major cause of morbidity and mortality that disproportionally affects HSCT and SOT recipients. Compared to the general inpatient population, HSCT and SOT patients have a higher incidence of CDI, are more likely to have severe infection, and are at greater risk of recurrence. Reference Revolinski and Munoz-Price74 For these reasons, reducing CDI rates is a priority of AMS programs. Interventions aimed at restricting antimicrobial exposure and providing provider education and feedback have been highly successful. Reference Mullane and Dubberke75,Reference Pouch and Friedman-Moraco76 Effective CDI antimicrobial stewardship practices are an interdisciplinary effort engaging diagnostic stewardship and infection prevention and control. Reference Revolinski and Munoz-Price74,Reference Pouch and Friedman-Moraco76 Active adaptation of these practices to the dynamic and unique factors of each transplant center is crucial for sustained progress in reducing CDI burden in this population. Stewardship areas of uncertainty include appropriate patient selection for testing, implications of asymptomatic screening, and the role of anti-CDI therapeutic prophylaxis.

The use of multistep testing algorithms has improved the analytic diagnostic stage, but CDI is a clinical diagnosis that depends on preanalytic decisions. Reference Pouch and Friedman-Moraco76 Differentiating asymptomatic colonization from CDI is a long-standing diagnostic conundrum that is particularly problematic in the transplant population. Transplant patients are at risk of overdiagnosis because of increased risk of toxigenic C. difficile carriage Reference Boly, Reske and Kwon77 and multiple confounding factors that cause diarrhea, including antibiotic use, immunosuppressive medications, mucositis, and graft-versus-host disease. Reference Revolinski and Munoz-Price78 Comprehensive review for other etiologies of diarrhea relative to a patient’s clinical symptoms such as degree of diarrhea, should be a priority prior to testing. Reference Alonso, Maron and Kamboj79

Screening transplant patients at admission for C. difficile colonization facilitates early implementation of infection prevention measures and may result in decreased horizontal transmission. Reference Revolinski and Munoz-Price78,Reference Barker, Krasity, Musuuza and Safdar80 In one study of patients admitted to an inpatient hematological unit, colonization with C. difficile conferred an 11.6 times higher odds of progression to CDI compared to those without colonization. Reference Cannon, Musuuza and Barker81 Identification of these asymptomatic carriers is an opportunity for targeted risk-reduction measures, such as antimicrobial review, to reduce the risk not only for symptomatic infection but for vertical transmission as well. However, the role of prophylactic, pharmacological measures to prevent the progression of colonization to infection is unknown. It is unclear how to optimally use asymptomatic screening for AMS efforts without unintentionally causing inappropriate treatment from misinterpretation of tests. Reference Barker, Krasity, Musuuza and Safdar80,Reference McCort, Oehler and Enriquez82

Oral vancomycin prophylaxis is an attractive option to prevent CDI. Retrospective reviews suggest that primary prophylaxis in allogenic HSCT recipients is associated with significantly lower rates of CDI Reference Ganetsky, Han and Hughes83,Reference Altemeier and Konrardy84 and that secondary prophylaxis is effective in reducing CDI recurrence in kidney transplant patients. Reference Splinter, Kerstenetzky and Jorgenson85 Although these studies found no instances of vancomycin-resistant Enterococcus (VRE) colonization or bacteremia, Reference Ganetsky, Han and Hughes83,Reference Altemeier and Konrardy84 several other studies have found that the use of oral vancomycin increased the risk of VRE overgrowth and infection Reference Lee, Plechot, Gohil and Le86,Reference Zacharioudakis, Zervou, Dubrovskaya and Phillips87 and can alter gut microbiome, which is linked to poor outcomes. Reference Alonso, Maron and Kamboj79 Despite the suggested benefit, a number of questions on the potential role for prophylaxis remain, including optimal duration, cost implications, effects on the intestinal microbiome, potential to drive the emergence of vancomycin-resistant C. difficile strains, and the benefit of secondary prophylaxis if fidaxomicin was used as initial therapy. Additional research can help characterize factors that may portend increased risk of CDI in HSCT and SOT recipients to build a risk stratification approach to prophylaxis.

In conclusion, the challenges facing AMS in transplant infectious diseases illustrate the difficulties in integrating the available evidence and diagnostic uncertainties with host-specific and local epidemiological factors to implement measures catered to both individuals and larger populations. The challenges, which are summarized in Table 1, have shared features, but specific solutions vary and should be personalized to institutional epidemiological patterns. In these uniquely vulnerable hosts, there is no “one size fits all” approach to AMS. For AMS challenges supported by strong evidence, we suggest that the implementation of practices in context of local epidemiology and dynamic evaluation over time to develop sustained, targeted measures. For challenges driven by knowledge gaps, recognizing the limitations of current evidence and engaging interdisciplinary teams to help risk-stratify patients are important to inform clinical practice. Employing thoughtful strategies is crucial for this population, which is disproportionally affected by infections and at risk for adverse effects of antimicrobial misuse and overuse.

Table 1. Summary of the Top 5 Antimicrobial Stewardship Challenges and Potential Mitigating Strategies

Note. HSCT, hematopoietic stem cell transplant.

Acknowledgments

Financial support

No financial support was provided relevant to this article.

Conflicts of interest

All authors report no conflicts of interest relevant to this article.

References

Coussement, J, Maggiore, U, Manuel, O, et al. Diagnosis and management of asymptomatic bacteriuria in kidney transplant recipients: a survey of current practice in Europe. Nephrol Dial Transpl 2018;33:16611668.CrossRefGoogle ScholarPubMed
Nicolle, LE, Gupta, K, Bradley, SF, et al. Clinical practice guideline for the management of asymptomatic bacteriuria: 2019 update by the Infectious Diseases Society of America. Clin Infect Dis 2019;68(10):e83e110.CrossRefGoogle ScholarPubMed
Goldman, JD, Julian, K. Urinary tract infections in solid organ transplant recipients: guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin Transpl 2019;33:e13507.CrossRefGoogle ScholarPubMed
Kotagiri, P, Chembolli, D, Ryan, J, Hughes, PD, Toussaint, ND. Urinary tract infections in the first year post–kidney transplantation: potential benefits of treating asymptomatic bacteriuria. Transpl Proc 2017;49:20702075.CrossRefGoogle ScholarPubMed
Green, H, Rahamimov, R, Goldberg, E, et al. Consequences of treated versus untreated asymptomatic bacteriuria in the first year following kidney transplantation: retrospective observational study. Eur J Clin Microbiol Infect Dis 2013;32:127131.10.1007/s10096-012-1727-2CrossRefGoogle ScholarPubMed
Lee, JR, Bang, H, Dadhania, D, et al. Independent risk factors for urinary tract infection and for subsequent bacteremia or acute cellular rejection: a single-center report of 1,166 kidney allograft recipients. Transplantation 2013;96:732738.CrossRefGoogle ScholarPubMed
Origüen, J, López-Medrano, F, Fernández-Ruiz, M, et al. Should asymptomatic bacteriuria be systematically treated in kidney transplant recipients? Results from a randomized controlled trial. Am J Transpl 2016;16:29432953.CrossRefGoogle ScholarPubMed
Coussement, J, Kamar, N, Matignon, M, et al. Antibiotics versus no therapy in kidney transplant recipients with asymptomatic bacteriuria (BiRT): a pragmatic, multicentre, randomized, controlled trial. Clin Microbiol Infect 2021;27:398405.10.1016/j.cmi.2020.09.005CrossRefGoogle ScholarPubMed
Sabé, N, Oriol, I, Melilli, E, et al. Antibiotic treatment versus no treatment for asymptomatic bacteriuria in kidney transplant recipients: a multicenter randomized trial. Open Forum Infect Dis 2019;6(6):ofz243.CrossRefGoogle ScholarPubMed
Coussement, J, Kamar, N, Abramowicz, D. New evidence shows it is time to stop unnecessary use of antibiotics in kidney transplant recipients with asymptomatic bacteriuria. Nephrol Dial Transpl 2021;36:754756.CrossRefGoogle ScholarPubMed
Dhakal, B, Giri, S, Levin, A, et al. Factors associated with unplanned 30-day readmissions after hematopoietic cell transplantation among US hospitals. JAMA Netw Open 2019;2:e196476.10.1001/jamanetworkopen.2019.6476CrossRefGoogle ScholarPubMed
Kumar, G, Ahmad, S, Taneja, A, et al. Severe sepsis in hematopoietic stem cell transplant recipients. Crit Care Med 2015;43:411421.CrossRefGoogle ScholarPubMed
Lind, ML, Mooney, SJ, Carone, M, et al. Development and validation of a machine learning model to estimate bacterial sepsis among immunocompromised recipients of stem cell transplant. JAMA Netw Open 2021;4:e214514.CrossRefGoogle ScholarPubMed
Shallis, RM, Terry, CM, Lim, SH. Changes in intestinal microbiota and their effects on allogeneic stem cell transplantation. Am J Hematol 2018;93:122128.CrossRefGoogle ScholarPubMed
Taplitz, RA, Kennedy, EB, Flowers, CR. Antimicrobial prophylaxis for adult patients with cancer-related immunosuppression: ASCO and IDSA clinical practice guideline update summary. J Oncol Pract 2018;14:692695.CrossRefGoogle ScholarPubMed
Murray, CJL, Ikuta, KS, Sharara, F, et al. Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. Lancet 2022.CrossRefGoogle Scholar
Giannella, M, Bartoletti, M, Conti, M, Righi, E. Carbapenemase-producing Enterobacteriaceae in transplant patients. J Antimicrob Chemother 2021;76 suppl 1:i27i39.CrossRefGoogle Scholar
Ford, CD, Gazdik, MA, Lopansri, BK, et al. Vancomycin-resistant Enterococcus colonization and bacteremia and hematopoietic stem cell transplantation outcomes. Biol Blood Marrow Transpl 2017;23:340346.CrossRefGoogle ScholarPubMed
Kamboj, M, Cohen, N, Huang, YT, et al. Impact of empiric treatment for vancomycin-resistant Enterococcus in colonized patients early after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transpl 2019;25:594598.CrossRefGoogle ScholarPubMed
Averbuch, D, Orasch, C, Cordonnier, C, et al. European guidelines for empirical antibacterial therapy for febrile neutropenic patients in the era of growing resistance: summary of the 2011 4th European Conference on Infections in Leukemia. Haematologica 2013;98:18261835.CrossRefGoogle ScholarPubMed
Averbuch, D, Tridello, G, Hoek, J, et al. Antimicrobial resistance in gram-negative rods causing bacteremia in hematopoietic stem cell transplant recipients: Intercontinental Prospective Study of the Infectious Diseases Working Party of the European Bone Marrow Transplantation Group. Clin Infect Dis 2017;65:18191828.CrossRefGoogle ScholarPubMed
Gudiol, C, Albasanz-Puig, A, Cuervo, G, Carratala, J. Understanding and managing sepsis in patients with cancer in the era of antimicrobial resistance. Front Med (Lausanne) 2021;8:636547.CrossRefGoogle ScholarPubMed
Gyssens, IC, Kern, WV, Livermore, DM. The role of antibiotic stewardship in limiting antibacterial resistance among hematology patients. Haematologica 2013;98:18211825.CrossRefGoogle ScholarPubMed
Freifeld, AG, Bow, EJ, Sepkowitz, KA, et al. Clinical practice guideline for the use of antimicrobial agents in neutropenic patients with cancer: 2010 update by the Infectious Diseases Society of America. Clin Infect Dis 2011;52:e56e93.CrossRefGoogle ScholarPubMed
Verlinden, A, Mikulska, M, Knelange, NS, et al. Current antimicrobial practice in febrile neutropenia across Europe and Asia: the EBMT Infectious Disease Working Party survey. Bone Marrow Transpl 2020;55:15881594.CrossRefGoogle ScholarPubMed
Aguilar-Guisado, M, Espigado, I, Martín-Peña, A, et al. Optimisation of empirical antimicrobial therapy in patients with haematological malignancies and febrile neutropenia (How Long study): an open-label, randomised, controlled phase 4 trial. Lancet Haematol 2017;4:e573e583.CrossRefGoogle ScholarPubMed
Le Clech, L, Talarmin, JP, Couturier, MA, et al. Early discontinuation of empirical antibacterial therapy in febrile neutropenia: the ANTIBIOSTOP study. Infect Dis (Lond) 2018;50:539549.CrossRefGoogle ScholarPubMed
Rearigh, L, Stohs, E, Freifeld, A, Zimmer, A. De-escalation of empiric broad-spectrum antibiotics in hematopoietic stem cell transplant recipients with febrile neutropenia. Ann Hematol 2020;99:19171924.CrossRefGoogle ScholarPubMed
Stern, A, Carrara, E, Bitterman, R, Yahav, D, Leibovici, L, Paul, M. Early discontinuation of antibiotics for febrile neutropenia versus continuation until neutropenia resolution in people with cancer. Cochrane Database Syst Rev 2019;1:CD012184.Google ScholarPubMed
Baden, LR, Bensinger, W, Angarone, M, et al. NCCN Clinical practice guidelines in oncology. Prevention and treatment of cancer-related infections version 1.2021. National Comprehensive Cancer Network website. https://www.gov.br/ans/pt-br/arquivos/acesso-a-informacao/participacao-da-sociedade/comites-e-comissoes/cosaude-comite-permanente-de-regulacao-da-atencao-a-saude/atas-e-reunioes/03/cosaude-3reuniao-nccn-cancer-related-infections.pdf. Published 2021. Accessed April 14, 2022.Google Scholar
Weisser, M, Theilacker, C, Tschudin Sutter, S, et al. Secular trends of bloodstream infections during neutropenia in 15 181 haematopoietic stem cell transplants: 13-year results from a European multicentre surveillance study (ONKO-KISS). Clin Microbiol Infect 2017;23:854859.CrossRefGoogle Scholar
Kern, WV, Roth, JA, Bertz, H, et al. Contribution of specific pathogens to bloodstream infection mortality in neutropenic patients with hematologic malignancies: results from a multicentric surveillance cohort study. Transpl Infect Dis 2019;21:e13186.CrossRefGoogle ScholarPubMed
Gudiol, C, Albasanz-Puig, A, Laporte-Amargós, J, et al. Clinical predictive model of multidrug resistance in neutropenic cancer patients with bloodstream infection due to Pseudomonas aeruginosa . Antimicrob Agents Chemother 2020;64:e0249419.CrossRefGoogle ScholarPubMed
Egan, G, Robinson, PD, Martinez, JPD, et al. Efficacy of antibiotic prophylaxis in patients with cancer and hematopoietic stem cell transplantation recipients: a systematic review of randomized trials. Cancer Med 2019;8:45364546.CrossRefGoogle ScholarPubMed
Signorelli, J, Zimmer, A, Liewer, S, Shostrom, VK, Freifeld, A. Incidence of febrile neutropenia in autologous hematopoietic stem cell transplant (HSCT) recipients on levofloxacin prophylaxis. Transpl Infect Dis 2020;22:e13225.CrossRefGoogle ScholarPubMed
Mikulska, M, Averbuch, D, Tissot, F, et al. Fluoroquinolone prophylaxis in haematological cancer patients with neutropenia: ECIL critical appraisal of previous guidelines. J Infect 2018;76:2037.CrossRefGoogle ScholarPubMed
Barlam, TF, Cosgrove, SE, Abbo, LM, et al. Implementing an antibiotic stewardship program: guidelines by the Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America. Clin Infect Dis 2016;62(10):e51e77.CrossRefGoogle Scholar
Barreto, JN, Aitken, SL, Krantz, EM, et al. Variation in clinical practice and attitudes on antibacterial management of fever and neutropenia in patients with hematologic malignancy: a survey of cancer centers across the United States. Open Forum Infect Dis 2022;9:ofac005.CrossRefGoogle ScholarPubMed
Douglas, AP, Hall, L, James, RS, et al. Quality of inpatient antimicrobial use in hematology and oncology patients. Infect Control Hosp Epidemiol 2021;42:12351244.CrossRefGoogle Scholar
Hosseini-Moghaddam, SM, Ouédraogo, A, Naylor, KL, et al. Incidence and outcomes of invasive fungal infection among solid-organ transplant recipients: a population-based cohort study. Transpl Infect Dis 2020;22:e13250.CrossRefGoogle ScholarPubMed
Aslam, S, Rotstein, C. Candida infections in solid-organ transplantation: guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin Transpl 2019;33:e13623.CrossRefGoogle ScholarPubMed
Lavezzo, B, Stratta, C, Ballaris, MA, et al. Invasive Candida infections in low-risk liver transplant patients given no antifungal prophylaxis in the postoperative period. Transpl Proc 2014;46:23122313.CrossRefGoogle Scholar
Singh, N, Wagener, MM, Cacciarelli, TV, Levitsky, J. Antifungal management practices in liver transplant recipients. Am J Transpl 2008;8:426431.CrossRefGoogle ScholarPubMed
Fortún, J, Martín-Davila, P, Moreno, S, et al. Prevention of invasive fungal infections in liver transplant recipients: the role of prophylaxis with lipid formulations of amphotericin B in high-risk patients. J Antimicrob Chemother 2003;52:813819.CrossRefGoogle ScholarPubMed
Fortún, J, Muriel, A, Martín-Dávila, P, et al. Caspofungin versus fluconazole as prophylaxis of invasive fungal infection in high-risk liver transplantation recipients: a propensity score analysis. Liver Transpl 2016;22:427435.CrossRefGoogle ScholarPubMed
Jorgenson, MR, Descourouez, JL, Marka, NA, et al. a targeted fungal prophylaxis protocol with static dosed fluconazole significantly reduces invasive fungal infection after liver transplantation. Transpl Infect Dis 2019;21:e13156.CrossRefGoogle ScholarPubMed
Muilwijk, EW, de Lange, DW, Schouten, JA, et al. Suboptimal dosing of fluconazole in critically ill patients: time to rethink dosing. Antimicrob Agents Chemother 2020;64:e0098420.CrossRefGoogle ScholarPubMed
Gatti, M, Rinaldi, M, Ferraro, G, et al. Breakthrough invasive fungal infections in liver transplant recipients exposed to prophylaxis with echinocandins vs other antifungal agents: a systematic review and meta-analysis. Mycoses 2021;64:13171327.CrossRefGoogle ScholarPubMed
Shields, RK, Nguyen, MH, Press, EG, Clancy, CJ. Abdominal candidiasis is a hidden reservoir of echinocandin resistance. Antimicrob Agents Chemother 2014;58:76017605.CrossRefGoogle ScholarPubMed
Prigent, G, Aït-Ammar, N, Levesque, E, et al. Echinocandin resistance in Candida species isolates from liver transplant recipients. Antimicrob Agents Chemother 2017;61:e0122916.CrossRefGoogle ScholarPubMed
Lavezzo, B, Romagnoli, R, Balagna, R, De Rosa, FG. The issue of the antifungal drug choice in prophylaxis of invasive fungal infection after liver transplant. Transpl Infect Dis 2020;22:e13220.CrossRefGoogle ScholarPubMed
Lum, L, Lee, A, Vu, M, Strasser, S, Davis, R. Epidemiology and risk factors for invasive fungal disease in liver transplant recipients in a tertiary transplant center. Transpl Infect Dis 2020;22:e13361.CrossRefGoogle Scholar
Johnson, MD, Lewis, RE, Dodds Ashley, ES, et al. Core recommendations for antifungal stewardship: a statement of the mycoses study group education and research consortium. J Infect Dis 2020;222 suppl 3:S175S198.CrossRefGoogle Scholar
Khanina, A, Urbancic, KF, Haeusler, GM, et al. Establishing essential metrics for antifungal stewardship in hospitals: the results of an international Delphi survey. J Antimicrob Chemother 2020;76:253262.CrossRefGoogle Scholar
Husain, S, Camargo, JF. Invasive aspergillosis in solid-organ transplant recipients: guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin Transplant 2019;33:e13544.CrossRefGoogle ScholarPubMed
De Mol, W, Bos, S, Beeckmans, H, et al. Antifungal prophylaxis after lung transplantation: where are we now? Transplantation 2021;105:25382545.CrossRefGoogle ScholarPubMed
Pennington, KM, Baqir, M, Erwin, PJ, Razonable, RR, Murad, MH, Kennedy, CC. Antifungal prophylaxis in lung transplant recipients: a systematic review and meta-analysis. Transpl Infect Dis 2020;22:e13333.CrossRefGoogle ScholarPubMed
Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention. Updated norovirus outbreak management and disease prevention guidelines. MMWR Recomm Rep 2011;60(Rr-3):118.Google Scholar
Pennington, KM, Yost, KJ, Escalante, P, Razonable, RR, Kennedy, CC. Antifungal prophylaxis in lung transplant: a survey of US transplant centers. Clin Transpl 2019;33:e13630.CrossRefGoogle Scholar
Chong, PP, Kennedy, CC, Hathcock, MA, Kremers, WK, Razonable, RR. Epidemiology of invasive fungal infections in lung transplant recipients on long-term azole antifungal prophylaxis. Clin Transpl 2015;29:311318.CrossRefGoogle ScholarPubMed
Vazquez, R, Vazquez-Guillamet, MC, Suarez, J, Mooney, J, Montoya, JG, Dhillon, GS. Invasive mold infections in lung and heart-lung transplant recipients: Stanford University experience. Transpl Infect Dis 2015;17:259266.CrossRefGoogle ScholarPubMed
Bae, M, Lee, S-O, Jo, K-W, et al. Infections in lung transplant recipients during and after prophylaxis. Infect Chemother 2020;52:600610.CrossRefGoogle ScholarPubMed
Zinoviev, R, Lippincott, CK, Keller, SC, Gilotra, NA. In full flow: left ventricular assist device infections in the modern era. Open Forum Infect Dis 2020;7(5):ofaa124.CrossRefGoogle ScholarPubMed
Kusne, S, Mooney, M, Danziger-Isakov, L, et al. An ISHLT consensus document for prevention and management strategies for mechanical circulatory support infection. J Heart Lung Transpl 2017;36:11371153.CrossRefGoogle ScholarPubMed
Molina, EJ, Shah, P, Kiernan, MS, Cornwell, WK, et al. The Society of Thoracic Surgeons Intermacs 2020 annual report. Ann Thorac Surg 2021;111:778792.CrossRefGoogle Scholar
Blanco-Guzman, MO, Wang, X, Vader, JM, Olsen, MA, Dubberke, ER. Epidemiology of left ventricular assist device infections: findings from a large nonregistry cohort. Clin Infect Dis 2021;72:190197.CrossRefGoogle ScholarPubMed
Koval, CE, Stosor, V. Ventricular assist device-related infections and solid organ transplantation—guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin Transpl 2019;33:e13552.CrossRefGoogle ScholarPubMed
Puius, YA, Parkar, F, Tlamsa, AP, et al. Gallium-67 single-photon emission computed tomography affects management of infections of left ventricular assist devices. Asaio J 2021;67:746751.Google ScholarPubMed
Radcliffe, C, Doilicho, N, Niu, YS, Grant, M. Efficacy and safety of chronic antimicrobial suppression therapy for left ventricular assist device driveline infections: a single-center descriptive experience. Transpl Infect Dis 2020;22:e13379.CrossRefGoogle ScholarPubMed
Jennings, DL, Chopra, A, Chambers, R, Morgan, JA. Clinical outcomes associated with chronic antimicrobial suppression therapy in patients with continuous-flow left ventricular assist devices. Artif Organs 2014;38:875879.CrossRefGoogle ScholarPubMed
Hamad, Y, Blanco-Guzman, MO, Olsen, MA, et al. The role of chronic suppressive antibiotics therapy in superficial drive line infection relapse of left ventricular assist devices: a retrospective cohort from a tertiary care center. Transpl Infect Dis 2021;23:e13686.CrossRefGoogle ScholarPubMed
Hall, SF, Athans, V, Wanek, MR, Wang, L, Estep, JD, Williams, B. Evaluation of a hospital-wide vancomycin-dosing nomogram in patients with continuous-flow left ventricular assist devices. Int J Artif Organs 2021;44:411417.CrossRefGoogle ScholarPubMed
Jennings, DL, Makowski, CT, Chambers, RM, Lanfear, DE. Dosing of vancomycin in patients with continuous-flow left ventricular assist devices: a clinical pharmacokinetic analysis. Int J Artif Organs 2014;37:270274.CrossRefGoogle ScholarPubMed
Revolinski, SL, Munoz-Price, LS. Clostridium difficile in immunocompromised hosts: a review of epidemiology, risk factors, treatment, and prevention. Clin Infect Dis 2019;68:21442153.CrossRefGoogle ScholarPubMed
Mullane, KM, Dubberke, ER. Management of Clostridioides (formerly Clostridium) difficile infection (CDI) in solid-organ transplant recipients: guidelines from the American Society of Transplantation Community of Practice. Clin Transpl 2019;33:e13564.CrossRefGoogle ScholarPubMed
Pouch, SM, Friedman-Moraco, RJ. Prevention and treatment of Clostridium difficile–associated diarrhea in solid-organ transplant recipients. Infect Dis Clin N Am 2018;32:733748.CrossRefGoogle ScholarPubMed
Boly, FJ, Reske, KA, Kwon, JH. The role of diagnostic stewardship in Clostridioides difficile testing: challenges and opportunities. Curr Infect Dis Rep 2020;22(3):7.CrossRefGoogle ScholarPubMed
Revolinski, SL, Munoz-Price, LS. Clostridioides difficile in transplant patients: early diagnosis, treatment, and prevention. Curr Opin Infect Dis 2019;32:307313.CrossRefGoogle ScholarPubMed
Alonso, CD, Maron, G, Kamboj, M, et al. American Society for Transplantation and Cellular Therapy Series: #5–Management of Clostridioides difficile infection in hematopoietic cell transplant recipients. Transpl Cell Ther 2022. doi: 10.1016/j.jtct.2022.02.013.CrossRefGoogle ScholarPubMed
Barker, AK, Krasity, B, Musuuza, J, Safdar, N. Screening for asymptomatic Clostridium difficile among bone marrow transplant patients: a mixed-methods study of intervention effectiveness and feasibility. Infect Control Hosp Epidemiol 2018;39:177185.CrossRefGoogle ScholarPubMed
Cannon, CM, Musuuza, JS, Barker, AK, et al. Risk of Clostridium difficile infection in hematology-oncology patients colonized with toxigenic C. difficile . Infect Control Hosp Epidemiol 2017;38:718720.CrossRefGoogle ScholarPubMed
McCort, MN, Oehler, C, Enriquez, M, et al. Universal molecular Clostridioides difficile screening and overtreatment in solid organ transplant recipients. Transpl Infect Dis 2020;22:e13375.CrossRefGoogle ScholarPubMed
Ganetsky, A, Han, JH, Hughes, ME, et al. Oral vancomycin prophylaxis is highly effective in preventing Clostridium difficile infection in allogeneic hematopoietic cell transplant recipients. Clin Infect Dis 2019;68:20032009.CrossRefGoogle ScholarPubMed
Altemeier, OJ, Konrardy, KT. Oral vancomycin for Clostridioides difficile prophylaxis in allogenic hematopoietic cell transplant. Transpl Infect Dis 2022;24:e13790.CrossRefGoogle ScholarPubMed
Splinter, LE, Kerstenetzky, L, Jorgenson, MR, et al. Vancomycin prophylaxis for prevention of Clostridium difficile infection recurrence in renal transplant patients. Ann Pharmacother 2018;52:113119.CrossRefGoogle ScholarPubMed
Lee, HS, Plechot, K, Gohil, S, Le, J. Clostridium difficile: diagnosis and the consequence of over diagnosis. Infect Dis Ther 2021;10:687697.CrossRefGoogle ScholarPubMed
Zacharioudakis, IM, Zervou, FN, Dubrovskaya, Y, Phillips, MS. Oral vancomycin prophylaxis against recurrent Clostridioides difficile infection: efficacy and side effects in two hospitals. Infect Control Hosp Epidemiol 2020;41:908913.CrossRefGoogle ScholarPubMed
Figure 0

Table 1. Summary of the Top 5 Antimicrobial Stewardship Challenges and Potential Mitigating Strategies