Next Article in Journal
Lytic Bacteriophage Is a Promising Adjunct to Common Antibiotics across Cystic Fibrosis Clinical Strains and Culture Models of Pseudomonas aeruginosa Infection
Next Article in Special Issue
Provision of Microbiology, Infection Services and Antimicrobial Stewardship in Intensive Care: A Survey across the Critical Care Networks in England and Wales
Previous Article in Journal
Comprehensive Study of Components and Antimicrobial Properties of Essential Oil Extracted from Carum carvi L. Seeds
Previous Article in Special Issue
Antimicrobial Prescribing before and after the Implementation of a Carbapenem-Focused Antimicrobial Stewardship Program in a Greek Tertiary Hospital during the COVID-19 Pandemic
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

Challenges and Opportunities in Antimicrobial Stewardship among Hematopoietic Stem Cell Transplant and Oncology Patients

1
Division of Allergy and Infectious Disease, Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
2
Division of Blood Disorders, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
3
Englewood Health, Englewood, NJ 07631, USA
4
Rutgers-Ernest Mario School of Pharmacy, Piscataway, NJ 08854, USA
*
Author to whom correspondence should be addressed.
Antibiotics 2023, 12(3), 592; https://doi.org/10.3390/antibiotics12030592
Submission received: 28 February 2023 / Revised: 11 March 2023 / Accepted: 13 March 2023 / Published: 16 March 2023

Abstract

:
Antimicrobial stewardship programs play a critical role in optimizing the use of antimicrobials against pathogens in the era of growing multi-drug resistance. However, implementation of antimicrobial stewardship programs among the hematopoietic stem cell transplant and oncology populations has posed challenges due to multiple risk factors in the host populations and the infections that affect them. The consideration of underlying immunosuppression and a higher risk for poor outcomes have shaped therapeutic decisions for these patients. In this multidisciplinary perspective piece, we provide a summary of the current landscape of antimicrobial stewardship, unique challenges, and opportunities for unmet needs in these patient populations.

1. Introduction

A report from The Lancet estimated that 4.95 million deaths globally were associated with antimicrobial resistance (AMR) in 2019 [1]. The formation of antimicrobial stewardship programs (ASPs) has been a crucial step in reducing the growing problem of antimicrobial resistance and poor patient outcomes through the optimization of antimicrobial use. The implementation of ASPs has gained more global recognition with the development of guidelines and has become a requirement for accredited hospitals by the Joint Commission in the United States [2,3]. It has been noted that the implementation of ASPs in transplant populations, including in solid organ transplant (SOT) recipients, poses challenges, such as increased susceptibility to infections and prolonged courses of antimicrobials [4,5]. However, no consensus guidelines currently exist for ASP implementation in hematopoietic stem cell transplant (HSCT) patients and patients living with malignancies [6,7].
In this perspective article, we provide a summary of the current landscape of ASP practices, unique challenges, and opportunities for unmet needs in the HSCT and oncology populations.

2. Unique Challenges to Antimicrobial Stewardship in Oncology Patients

There are multiple facets involved in the care of patients living with malignancies, which require unique approaches by antimicrobial stewardship. Here we highlight some host and treatment factors that affect antimicrobial use in the immunocompromised host living with malignancy.

2.1. Understanding Underlying Host Immune Status and Infectious Risks

There is substantial variability in immune status among oncology patients. The avoidance of immune surveillance, escape from immune destruction, and tumor-promoting inflammation, are some main attributes of cancer [8]. Observed classes of inflammatory and immune response are determined by the characteristics of the originating tissue, proinflammatory mediators released by the tumor or stromal milieu, nature of tissue damage, or the organism associated with carcinogenesis [8]. The increased susceptibility of patients with cancer to infections depends on the status of the malignancy, the dominant type of immune deficiency, and the duration and intensity of therapy-mediated immunosuppression.
Cancers may be related to a chronic infection, or inflammation related to an infection, such as with Epstein–Barr virus, Helicobacter pylori, Hepatitis B and C viruses, Human Immunodeficiency Virus (HIV), Human Papillomavirus (HPV), and Human T-Lymphotropic Virus (HTLV). Other malignancies may lead to altered intestinal composition, tissue injury, or physical interactions with hematopoietic cells, leading to an increased vulnerability to infectious complications [9]. The disruption of hematopoiesis with resultant neutropenia in patients with acute leukemias predisposes patients to serious bacterial, fungal, and viral infections [9,10]. A lack of splenic and humoral immunity in patients with B-cell disorders can also cause functional hypogammaglobulinemia [11].
The receipt of chemotherapeutic agents (e.g., cytotoxic, lymphocyte-depleting agents) can cause myelosuppression, impacting infection risk depending on the degree and duration of neutropenia and lymphopenia. Lower bone marrow reserve due to older age, the presence of multiple comorbidities, and poor performance status can propagate the risk of chemotherapy-induced neutropenia [12]. Many patients living with malignancies experience a disruption of mucosal barriers due to radiation and chemotherapy, predisposing them to mucositis, sinusitis, and bacterial translocation, in turn leading to bloodstream infections and neutropenic enterocolitis. Modern therapies, including small/signaling molecule inhibitors, monoclonal and bispecific antibodies, T-cell redirecting cellular therapies, and immune checkpoint inhibitors, can cause unique immune dysfunction, increasing the risk for opportunistic infections such as viral reactivation [13,14,15]. Reviews and case reports pertaining to infectious complications with these newer agents are increasingly being published, revealing that associated complications have yet to be reported regarding these therapies [16,17,18].

2.2. Additional Healthcare Considerations and Hematopoietic Stem Cell Transplantation

Besides inherent immune dysfunction resulting from an underlying malignancy and its treatment, patients living with malignancies are at risk for healthcare-associated infections. Patients within this population may also have indwelling devices, such as Ommaya reservoirs, central venous catheters, and urinary catheters, placing them at risk for device-related infections and complications [19]. Frequent exposure to the healthcare system can also predispose patients to acquisition and colonization with multi-drug resistant organisms (MDRO) [20,21,22].
Recipients of HSCT are predisposed to opportunistic infections due to the nature of conditioning regimens and their effects on the immune system, the disruption of hematopoiesis, the donor type and transplant graft source, the presence of mucosal injury, and the need for hospitalization. As highlighted in Figure 1 which is adapted from Tomblyn et al., the most frequently encountered infections in the pre-engraftment phase include bacterial infections related to gastrointestinal translocation, healthcare-associated infections, and Candida spp. infections, followed by infections from cytomegalovirus (CMV), Aspergillus spp., and Pneumocystis in the early post-engraftment period [23]. Figure 1 also provides examples of prevention strategies used to decrease the incidence of these infections which will be discussed later throughout this piece.
Different transplant graft sources (e.g., autologous stem cells collected from bone marrow or peripheral blood stem cells (PBSC); allogeneic stem cells collected from bone marrow, PBSC, or umbilical cord blood) impact the risk of the development of infections. In the month following an allogeneic bone marrow transplant (BMT), the incidence of infections is 47.9%, compared to 32.8% in allogeneic PBSC transplant, which is possibly related to quicker engraftment in the latter population [25,26,27]. Comparatively, umbilical cord blood transplantations carry the highest risk of infections during the first 100 days after transplant, possibly due to delayed engraftment [27]. The impact of donor type (e.g., matched–related, matched–unrelated) and the use of prophylaxis against graft-versus-host disease (GVHD) upon infectious risk remains unclear [28]. Due to the high probability of GVHD with allogeneic PBSCtransplant, there is an increased incidence of late fungal and viral infections when compared with allogeneic BMT [26]. Up to 20–80% of HSCT patients are affected by acute GVHD which may require heightened immunosuppression, placing patients at further risk for opportunistic and fungal infections [11,29,30].

2.3. Drug–Drug Interactions Affecting Antimicrobial Use

Significant drug–drug interactions (DDI) can affect the use of antimicrobials with concomitant chemotherapeutic and adjunctive treatments. For example, triazole agents, such as voriconazole, posaconazole, and fluconazole, are noted to cause QTc prolongation. Pharmacodynamically, triazole agents, fluoroquinolones, and macrolides can enhance the QTc-prolonging effects of antiemetics used to manage chemotherapy-induced nausea and vomiting, such as 5-HT3 receptor antagonists (e.g., ondansetron, granisetron) and dopamine receptor antagonists (e.g., metoclopramide) [31].
Antimicrobials that inhibit or induce the CYP3A4 enzyme can lead to increased or decreased levels of chemotherapeutic agents. For example, triazole agents are CYP3A4 inhibitors, which can affect concentrations of chemotherapeutic agents, such as taxane, vinca alkaloids, busulfan, etoposide, ifosfamide, cyclophosphamide, and tyrosine kinase inhibitors [32,33,34]. The CYP3A4-inhibiting antimicrobials can also complicate the management of serum concentrations of drugs with a narrow therapeutic index (e.g., tacrolimus, cyclosporine, and sirolimus): a notable example is voriconazole, which increases the AUC of sirolimus by 1000%, requiring at least a 90% dose reduction of sirolimus [34]. On the other hand, CYP3A4 inducers, such as rifampin, can decrease the serum concentrations of CYP3A4 substrates; therefore, the interacting medications may need to be dose-adjusted or switched to an alternative [34].
HSCT patients are particularly susceptible to significant drug–drug interactions, due to the frequent need for antimicrobial prophylaxis and treatment, polypharmacy, and the use of immunosuppressants [33]. For example, busulfan, a commonly used alkylating agent in stem cell transplant, can interact with triazoles and metronidazole. Voriconazole, a strong CYP3A4 inhibitor, can reduce busulfan clearance and increase its serum level, necessitating the close monitoring of patients regarding the occurrence of busulfan toxicity. Metronidazole also inhibits CYP3A4 in addition to competing for glutathione, which can increase busulfan levels; thus, metronidazole should be avoided for 72 h before and after busulfan administration [34]. Other antimicrobials that are CYP3A4 inhibitors and can cause DDI with immunosuppressants include macrolides and antiviral agents containing ritonavir (e.g., nirmatrelvir/ritonavir).
It is important to note these interactions as they may impact the therapeutic efficacy of antimicrobials, chemotherapeutic agents, and transplant-related medications. These examples also highlight a need for studies examining the clinical impact of DDI, alternative dosing strategies for mitigation of adverse effects, and development of antimicrobials with fewer interactions.

3. Unmet Needs and Opportunities for Antimicrobial Stewardship

Due to an increased susceptibility to infection, strategies such as antimicrobial prophylaxis and early empiric therapy have been studied with respect to reducing infection-related morbidity and mortality. However, the long-term consequences of these strategies can be serious. In this section, we review unmet needs, opportunities for further research, and strategies to improve antimicrobial stewardship in these immunocompromised populations.

3.1. Fever and Neutropenia

Approximately 80% of patients with hematologic malignancies and up to 50% of patients with solid tumor malignancies develop fever with underlying neutropenia, triggering the use of empiric antimicrobials to reduce morbidity [35]. Of those with febrile neutropenia, up to 45–50% of cases are due to unexplained fevers with no underlying microbiological cause [36]. The swift resolution of neutropenia facilitates the discontinuation of antibacterial agents for most patients with solid tumor malignancies, but antibacterial discontinuation remains an area of variability among patients with hematologic malignancies, due, in part, to the longer durations of neutropenia observed in these patients [37].
The extended use of empiric antimicrobials can lead to the selection of resistant organisms. For example, the prolonged use of vancomycin can result in increased rates of vancomycin-resistant enterococci infection [11]. Multiple studies evaluating bloodstream infections in patients with malignancies have revealed an increased incidence of extended spectrum beta-lactamase (ESBL) and carbapenem-resistant Enterobacterales (CRE), as well as increased rates of Pseudomonas aeruginosa and Acinetobacter baumanii infections [38,39]. Shorter durations of therapy have been studied in order to decrease bacterial selective pressure, and the implications of this intervention on morbidity are important points to consider. The ANTIBIOSTOP and the How Long studies have demonstrated that empiric antibacterials can be discontinued in hemodynamically stable neutropenic patients with a resolution of fever and lack of active infection; however, it should be noted that some high-risk patients such as those with GVHD were excluded from these studies [40,41]. This approach regarding the early discontinuation of empiric antibacterials has been recommended by The American Society of Transplantation and Cellular Therapy (ASTCT) and the European Conference on Infections in Leukemia (ECIL) [42,43]. Local centers may consider the development of collaborative guidelines that can account for the ongoing monitoring of patients with febrile neutropenia to streamline empiric antibacterial use.
The targeted use of empiric antimicrobials, such as the use of vancomycin, has been addressed in sources such as the National Comprehensive Cancer Network® (NCCN®) guidelines, in efforts to decrease antibacterial resistance [11]. A targeted review of epidemiology and resistance patterns among hospital units and/or among patients with underlying malignancy diagnosis, paired with rapid diagnostic tests (RDT), can help institutions evaluate their empiric antimicrobial choices, when to consider discontinuing antimicrobials, and when to escalate therapy in order to optimize therapeutic efficacy.

3.2. Antibacterial Prophylaxis

Antibacterial prophylaxis has been shown to decrease the incidence of Gram-negative infections in patients living with malignancy [11,44]. NCCN guidelines recommend the consideration of fluoroquinolone prophylaxis for neutropenic patients at an intermediate to a highrisk of infection, such as for patients with anticipated neutropenia longer than 10 days, or patients undergoing therapy for acute leukemia [11]. However, fluoroquinolone use is associated with increased rates of Clostridioides difficile (C. difficile) and may lead to the selection of resistant bacterial pathogens, notably decreasing their effectiveness in patients who are colonized with fluoroquinolone-resistant organisms [45,46]. Additionally, if a patient is receiving fluoroquinolone prophylaxis and develops fevers, empiric antibacterial treatment should not include fluoroquinolone and should entail the use of anti-pseudomonal β-lactams, such as piperacillin/tazobactam, cefepime, or meropenem [46,47,48].
A local data review of resistance patterns for MDRO can help centers create guidelines for the optimal use of fluoroquinolone prophylaxis. For patients with fluoroquinolone allergies, alternative agents, such as oral third-generation cephalosporins and trimethoprim/sulfamethoxazole, can be used [11]. Trimethoprim/sulfamethoxazole (TMP/SMX) prophylaxis has been shown to have similar benefits in terms of mortality when compared to fluoroquinolone prophylaxis; however TMP/SMX has been associated with more adverse effects [49]. In addition, TMP/SMX can compound the risk of leukopenia in patients recovering from chemotherapy. As discussed, it is worthwhile to consider the benefits and risks of antibacterial prophylaxis regimens that have potential adverse events such as antimicrobial resistance.

3.3. Antifungal Use and Prophylaxis

Patients undergoing cytotoxic chemotherapy and HSCT are at an increased risk for invasive fungal infections (IFI), with mortality reaching up to 30% [50,51,52,53]. There is considerable variability in the presentation of IFI, with concurrent thrombocytopenia often limiting the potential benefit of diagnostic bronchoscopy, leaving providers with imaging and indirect serum tests for diagnosis, such as Aspergillus galactomannan or beta-D-glucan testing [54,55]. The consensus group of the European Organization for Research and Treatment of Cancer/Invasive Fungal Infections Cooperative Group (EORTC), as well as the National Institute of Allergy and Infectious Diseases Mycoses Study Group (MSG), revised definitions to provide guidance about high-risk populations, imaging findings, and laboratory testing methods, needed to diagnose proven, probable, and possible IFI [54]. These guidelines have helped to standardize the diagnosis of IFI in patients with hematologic malignancies and note the subtleties in various indirect diagnostic methods [54].
Two main strategies have been utilized to treat IFI: a preemptive approach involving the use of diagnostic testing to guide treatment versus empiric antifungal therapy, where antifungal therapy is initiated while awaiting work-up. A recent Cochrane review concluded that pre-emptive antifungal treatment may reduce the duration of antifungal use without affecting IFI-related mortality in patients with febrile neutropenia when compared to empiric antifungal therapy, highlighting a need to study at-risk target populations and approaches to antifungal treatment [56]. The EORTC/MSG guidelines note that the T2Candida panel®, approved by the United States Food and Drug Administration (FDA) for the detection of common Candida species from whole-blood specimens, has a high negative predictive value, which can also aid ASPs in decreasing antifungal use [54].
Antifungal prophylaxis during periods of high-risk neutropenia and following allogeneic HSCT has now become an integral part of national guidelines and clinical care practices in interest of decreasing IFI incidence [11,57]. The NCCN and ECIL recommend posaconazole for antifungal prophylaxis in patients undergoing treatment for acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) [11,57]. Both organizations also note the increased risk of IFI in allogeneic HSCT patients, providing similar grading recommendations for the use of posaconazole, voriconazole, and amphotericin formulations (including NCCN grade 2B recommendation for isavuconazole), with the understanding that more research is needed to further support these gradings. For patients with significant DDI with azole agents, some centers have adopted the use of echinocandins, although there is a loss of anti-mold coverage when using this strategy [11,57]. Further studies are needed evaluating the use of antifungal prophylaxis strategies and its effect on IFI incidence as the primary endpoint.
It is estimated that up to 57% of antifungals prescribed are not optimal in many cancer patients due to issues such as inappropriate agent selection and dosing frequency [58]. Although antifungal stewardship is encompassed within antimicrobial stewardship, few consensus guidelines exist to support antifungal stewardship programs [58]. The Mycoses Study Group Education and Research Consortium highlights this need and offers recommendations specific to antifungal use based on the core elements of ASP as described by the Centers for Disease Control and Prevention (CDC) [59,60]. Other suggested strategies for improvement in antifungal stewardship have been published, including local education and guideline development, improvements in diagnostics for identification and susceptibility testing, optimizing antifungal dosing, and multidisciplinary team involvement [61,62].

3.4. Antiviral Use and Prophylaxis

Similar to patients undergoing SOT, HSCT patients with cytomegalovirus (CMV) infection have poor outcomes [63,64]. In the past, CMV viremia occurring post-HSCT was largely addressed through a pre-emptive approach, with viral load monitoring and corresponding treatment initiation with the antiviral agents valganciclovir or ganciclovir; however, treatment with these agents is limited by myelosuppression. In 2017, letermovir was approved by the FDA for CMV prophylaxis through day 100 post-HSCT in CMV-seropositive patients, later becoming a Grade A-1 recommendation by the ASTCT [24,65]. Cesaro et al. surveyed treatment approaches to CMV infection in a 2020 survey among European BMT centers, highlighting that up to 62% of centers are adopting a prophylaxis-based approach with letermovir [66]. Different strategies and the impact of letermovir prophylaxis on CMV reactivation are being studied [63,67,68].
Ganciclovir-resistant CMV is an emerging concern in the post-HSCT population, with treatment options including foscarnet, cidofovir, and maribavir. Notably, these antivirals have adverse effects: foscarnet and cidofovir can cause nephrotoxicity, while maribavir can interact with CYP3A4 substrates resulting in increased tacrolimus concentrations [69,70]. Publications by Jorgenson et al. describe a pharmacy-directed approach to CMV antiviral stewardship in SOT programs that has been associated with a decrease in the number of patients requiring treatment and a reduced ganciclovir resistance rate with a monitoring protocol for CMV viremia in patients receiving prophylaxis and treatment [71,72]. This type of antiviral stewardship approach may be an area of further exploration toward preventing CMV resistance in the post-HSCT population.
Other viral infections unique to oncology patients include acyclovir-resistant herpesvirus, BK polyoma virus, and adenovirus. Acyclovir (ACV)-resistant herpesvirus (HSV) has a prevalence of 3.5–10% among HSCT patients and can cause significant morbidity with prolonged viral shedding [73,74]. There are limited treatment options for ACV-resistant HSV, including foscarnet, cidofovir, and the investigative drug, pritelivir. Interventions, such as accessible diagnostic methods for resistance testing and improved dosing strategies for antiviral agents, may assist ASP in preventing antiviral resistance. Notably, no approved antiviral treatment options exist for BK polyoma virus or adenovirus with studies revealing a cautious benefit of cidofovir at the cost of nephrotoxicity; this example highlights another area requiring further research in terms of the identification of less harmful and more effective therapeutic approaches [67].

3.5. Adverse Effects of Antimicrobials

In addition to the adverse effects of antimicrobials, including systemic toxicity and DDI, antimicrobials can increase MDRO incidence through multiple mechanisms. As described previously, exposure to the healthcare system and antimicrobials can lead to the selection of and colonization with resistant organisms (e.g., ESBL organisms, CRE, VRE) [20,21]. Treatment centers should review resistance patterns and prevalence of MDRO at their institutions with consideration of ASP interventions to optimize the use of antimicrobials in an effort to prevent antimicrobial resistance [37,75,76].
Long-term antimicrobial use has also been linked to the disruption of the intestinal microbiome and an increased incidence of C. difficile infections in all populations [77]. C. difficile occurs with an increased incidence among patients with hematologic malignancies (up to 6.5 times higher) and HSCT (up to 9 times higher) when compared to the general inpatient population [78,79]. It is important to note that in addition to microbiota injury from antimicrobial exposure, HSCT recipients are also exposed to conditioning regimens and mucositis that can also disrupt the gut microbiota [80,81,82,83].
The disruption of the gut microbiome can result in serious, long-term consequences for patients living with malignancies. Galloway-Peña et al. observed that patients with a lower stool Shannon diversity index during initiation of chemotherapy for AML with resulting neutropenia experienced an increased infection incidence [84]. Lack of microbiome diversity during the peri-transplant period has been associated with poor overall survival post-HSCT and increased GVHD incidence [82,85,86]. Antimicrobial use after stem cell transplantation can compound this microbiota disruption and damage [87,88].

3.6. Expansion of Diagnostic and Susceptibility Testing Methods

The development of newer RDT platforms has made it possible for the earlier diagnosis of infections and for the determination of appropriate antimicrobial treatments. When compared to conventional microbiologic methods for pathogen identification, which may include subculture isolation, rapid molecular methods and matrix-assisted laser desorption/ionization–time-of-flight (MALDI-TOF) mass spectrometry offer quicker turn-around times in microbe identification [89,90,91]. Multiplex polymerase chain reaction (PCR) syndromic panels, such as panels developed for respiratory and gastrointestinal illnesses, allow for rapid diagnosis through the testing of multiple pathogens from a single sample.
Genotypic and phenotypic testing platforms performed on direct blood culture specimens (rather than on colonies isolated from subculture) have facilitated the rapid de-escalation of antibacterial agents and the early detection of MDRO for the potential escalation of therapy [92,93]. Platforms such as Accelerate Pheno® can offer rapid pathogen identification results within 90 min, as well as report antimicrobial susceptibility test results within 7 h [93]. The T2Candida® panel, which utilizes direct whole-blood specimens, can aid ASPs, both in the discontinuation of empiric antifungal use for candidemia due to its high negative predictive value for candidemia, as well as support the continuation of antifungals with a positive result due to increased sensitivity compared to blood cultures [54]. Local centers may consider the benefits of running RDT on-site and rapid turn-around time for results to optimize antimicrobial use, including the early identification of MDRO for use of extended-spectrum antimicrobials, as well as assistance with antibiotic de-escalation.
Another diagnostic challenge is the diagnosis of IFI. Currently, the sensitivity of serum Aspergillus galactomannan is low in patients receiving mold-active antifungals, which may limit its use in diagnosing IFI in patients receiving antifungal prophylaxis [54,55]. Further studies regarding the use of indirect testing methods for the diagnosis of IFI, as well ongoing evaluations of diagnostic definitions, may assist clinicians in diagnosing IFI and might assist ASPs in optimizing antifungal use.

3.7. Lack of Dedicated ASP Guidelines and the Need for Multidisciplinary Implementation

In 2016, the Infectious Diseases Society of America (IDSA) and the Society for Healthcare Epidemiology of America (SHEA) created a joint guideline regarding general ASP implementation in hospitals. Within these guidelines, specific recommendations regarding HSCT and oncology patients are made, suggesting the development of facility-specific guidelines for fever and neutropenia, as well as efforts towards antifungal stewardship, including the incorporation of non-invasive fungal markers to optimize antifungal use [60]. Currently, no consensus society guidelines exist regarding the implementation of ASPs specifically targeting immunocompromised patients with malignancies.
Emerging data continue to note the importance of different stewardship-related interventions in HSCT and oncology patients; however, the consistency of stewardship implementation has varied across the centers who manage these patients [6,7]. An Australian survey of antimicrobial prescription practices highlights differences in the adherence to guidelines for febrile neutropenia and antimicrobial use among BMT and other oncology services, emphasizing the need for the development of local guidelines [94]. In a survey done by Seo et al., approximately 76% of respondents from facilities who performed HSCT in the United States were involved in local guideline developments that were unique to the oncology population, such as those targeting febrile neutropenia; however, only 34% of the centers tracked outcomes related to antimicrobial use for these patients [6].
Figure 2 outlines a proposed multidisciplinary team approach towards ASP implementation to optimize educational opportunities for providers and to optimize patient outcomes. Elements in this model include stakeholder input from the patient and/or caregiver, oncology provider, infectious disease provider, pharmacists, and nurses, with ongoing input from the infection prevention department and microbiology laboratories. It is important that these teams work collaboratively within the lens of reporting, quality, and safety in order to create a comprehensive approach regarding antimicrobial use.

3.8. Summary of Opportunities for ASP Interventions Specific to Oncology and HSCT Patients

In 2019, the CDC outlined core elements of hospital antibiotic stewardship programs, which included leadership commitment, accountability, pharmacy expertise, action, tracking, reporting, and education. In Table 1, we summarize some ASP interventions previously discussed in this article and described in other studies within this framework as areas of focus, improvement, and future research regarding oncology and HSCT patients [6,95].

4. Conclusions

Patients living with solid tumors and hematologic malignancies are at an increased risk of infection due to underlying malignancies, immune dysfunction due to therapeutic agents, and other unique aspects of care. Many advances have occurred regarding expanded treatment options for solid tumors and hematologic malignancies; however, emerging data suggest that some of these newer therapies are associated with an increased risk for opportunistic infections. Studies have supported antimicrobial strategies to decrease the incidence and morbidity associated with opportunistic infections in these immunocompromised populations, such as antimicrobial prophylaxis; yet, the implementation of these strategies remains inconsistent.
More research is needed pertaining to the optimal use and development of novel antimicrobial agents for infections that are unique to the HSCT and oncologic populations, including antibacterial treatment for MDRO infections, antifungals for prophylaxis and treatment of resistant IFI, and the antiviral treatment of resistant CMV and HSV. Downstream consequences of different antimicrobial agents, including the development of resistant pathogens, disruption of the host microbiome, and long-term morbidity and mortality, are poorly described. Ongoing advances in RDT aimed at early pathogen detection and resistance genotype and phenotype determination will help ASPs and clinicians optimize antimicrobial use.
Given the complex components required to optimally care for patients undergoing HSCT and patients who are being treated for underlying malignancy, a multidisciplinary approach is imperative in order to successfully implement ASP and its interventions. It is important for institutions that care for these high-risk populations to implement collaborative ASP strategies to optimize antimicrobial use and to deliver safe outcomes in patients living with malignancies.

Author Contributions

Conceptualization, A.M., N.N., K.S.K. and P.J.B.; writing—original draft preparation, A.M., M.R.S., J.J.P. and P.J.B.; writing—review and editing, A.M., M.R.S., J.J.P., N.N., K.S.K. and P.J.B.; supervision, N.N., K.S.K. and P.J.B.; visualization, A.M. and P.J.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Murray, C.J.; Ikuta, K.S.; Sharara, F.; Swetschinski, L.; Aguilar, G.R.; Gray, A.; Han, C.; Bisignano, C.; Rao, P.; Wool, E.; et al. Global Burden of Bacterial Antimicrobial Resistance in 2019: A Systematic Analysis. Lancet 2022, 399, 629–655. [Google Scholar] [CrossRef] [PubMed]
  2. European Centre for Disease Prevention and Control. Proposals for EU Guidelines on the Prudent Use of Antimicrobials in Humans; ECDC: Stockholm, Sweden, 2017; Available online: http://ecdc.europa.eu/en/publications/_layouts/forms/Publication_DispForm.aspx?List=4f55ad51-4aed-4d32-b960-af70113dbb90&ID=1643 (accessed on 4 December 2022).
  3. WHO. Antimicrobial Stewardship Programmes in Health-Care Facilities in Low- and Middle-Income Countries: A Practical Toolkit; World Health Organization: Geneva, Switzerland, 2019. [Google Scholar]
  4. So, M.; Hand, J.; Forrest, G.; Pouch, S.M.; Te, H.; Ardura, M.I.; Bartash, R.M.; Dadhania, D.M.; Edelman, J.; Ince, D.; et al. White Paper on Antimicrobial Stewardship in Solid Organ Transplant Recipients. Am. J. Transplant. 2022, 22, 96–112. [Google Scholar] [CrossRef] [PubMed]
  5. Forrest, G.; Husain, S. Special Issue: The Urgent Need for Antimicrobial Stewardship in Transplantation. Transpl. Infect. Dis. 2022, 24, e13642. [Google Scholar] [CrossRef]
  6. Seo, S.K.; Lo, K.; Abbo, L.M. Current State of Antimicrobial Stewardship at Solid Organ and Hematopoietic Cell Transplant Centers in the United States. Infect. Control. Hosp. Epidemiol 2016, 37, 1195–1200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Barreto, J.N.; Aitken, S.L.; Krantz, E.M.; Nagel, J.L.; Dadwal, S.S.; Seo, S.K.; Liu, C. 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. [Google Scholar] [CrossRef] [PubMed]
  8. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Goldszmid, R.S.; Dzutsev, A.; Trinchieri, G. Host Immune Response to Infection and Cancer: Unexpected Commonalities. Cell Host Microbe 2014, 15, 295–305. [Google Scholar] [CrossRef] [Green Version]
  10. Atkins, S.; He, F. Chemotherapy and Beyond. Infect. Dis. Clin. N. Am. 2019, 33, 289–309. [Google Scholar] [CrossRef]
  11. National Comprehensive Cancer Network. Referenced with Permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Prevention and Treatment of Cancer-Related Infections; Version 3.2022; National Comprehensive Cancer Network, Inc.: Plymouth Meeting, PA, USA, 2022. [Google Scholar]
  12. Lyman, G.H.; Lyman, C.H.; Agboola, O. Anc Study Group Risk Models for Predicting Chemotherapy-Induced Neutropenia. Oncologist 2005, 10, 427–437. [Google Scholar] [CrossRef] [Green Version]
  13. Tillman, B.F.; Pauff, J.M.; Satyanarayana, G.; Talbott, M.; Warner, J.L. Systematic Review of Infectious Events with the Bruton Tyrosine Kinase Inhibitor Ibrutinib in the Treatment of Hematologic Malignancies. Eur. J. Haematol. 2018, 100, 325–334. [Google Scholar] [CrossRef]
  14. Safdar, A.; Armstrong, D. Infections in Patients With Hematologic Neoplasms and Hematopoietic Stem Cell Transplantation: Neutropenia, Humoral, and Splenic Defects. Clin. Infect. Dis. 2011, 53, 798–806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Davis, J.S.; Ferreira, D.; Paige, E.; Gedye, C.; Boyle, M. Infectious Complications of Biological and Small Molecule Targeted Immunomodulatory Therapies. Clin. Microbiol. Rev. 2020, 33, e00035-19. [Google Scholar] [CrossRef] [PubMed]
  16. Varughese, T.; Taur, Y.; Cohen, N.; Palomba, M.L.; Seo, S.K.; Hohl, T.M.; Redelman-Sidi, G. Serious Infections in Patients Receiving Ibrutinib for Treatment of Lymphoid Cancer. Clin. Infect. Dis. 2018, 67, 687–692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Ball, S.; Das, A.; Vutthikraivit, W.; Edwards, P.J.; Hardwicke, F.; Short, N.J.; Borthakur, G.; Maiti, A. Risk of Infection Associated With Ibrutinib in Patients With B-Cell Malignancies: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Clin. Lymphoma Myeloma Leuk. 2020, 20, 87–97.e5. [Google Scholar] [CrossRef] [PubMed]
  18. Redelman-Sidi, G.; Michielin, O.; Cervera, C.; Ribi, C.; Aguado, J.M.; Fernández-Ruiz, M.; Manuel, O. ESCMID Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the Safety of Targeted and Biological Therapies: An Infectious Diseases Perspective (Immune Checkpoint Inhibitors, Cell Adhesion Inhibitors, Sphingosine-1-Phosphate Receptor Modulators and Proteasome Inhibitors). Clin. Microbiol. Infect. 2018, 24, S95–S107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Baier, C.; Linke, L.; Eder, M.; Schwab, F.; Chaberny, I.F.; Vonberg, R.-P.; Ebadi, E. Incidence, Risk Factors and Healthcare Costs of Central Line-Associated Nosocomial Bloodstream Infections in Hematologic and Oncologic Patients. PLoS ONE 2020, 15, e0227772. [Google Scholar] [CrossRef]
  20. Satlin, M.J.; Walsh, T.J. Multidrug-Resistant Enterobacteriaceae, Pseudomonas Aeruginosa and Vancomycin-Resistant Enterococcus: Three Major Threats to Hematopoietic Stem Cell Transplant Recipients. Transpl. Infect. Dis. 2017, 19, e12762. [Google Scholar] [CrossRef]
  21. Satlin, M.J.; Cohen, N.; Ma, K.C.; Gedrimaite, Z.; Soave, R.; Askin, G.; Chen, L.; Kreiswirth, B.N.; Walsh, T.J.; Seo, S.K. Bacteremia Due to Carbapenem-Resistant Enterobacteriaceae in Neutropenic Patients with Hematologic Malignancies. J. Infect. 2016, 73, 336–345. [Google Scholar] [CrossRef] [Green Version]
  22. Martirosov, D.M.; Lodise, T.P. Emerging Trends in Epidemiology and Management of Infections Caused by Carbapenem-Resistant Enterobacteriaceae. Diagn. Microbiol. Infect. Dis. 2016, 85, 266–275. [Google Scholar] [CrossRef]
  23. Tomblyn, M.; Chiller, T.; Einsele, H.; Gress, R.; Sepkowitz, K.; Storek, J.; Wingard, J.R.; Young, J.-A.H.; Boeckh, M.A. Guidelines for Preventing Infectious Complications among Hematopoietic Cell Transplantation Recipients: A Global Perspective. Biol. Blood Marrow Transplant. 2009, 15, 1143–1238. [Google Scholar] [CrossRef] [Green Version]
  24. Hakki, M.; Aitken, S.L.; Danziger-Isakov, L.; Michaels, M.G.; Carpenter, P.A.; Chemaly, R.F.; Papanicolaou, G.A.; Boeckh, M.; Marty, F.M. American Society for Transplantation and Cellular Therapy Series: #3—Prevention of Cytomegalovirus Infection and Disease After Hematopoietic Cell Transplantation. Transplant. Cell. Ther. 2021, 27, 707–719. [Google Scholar] [CrossRef]
  25. Young, J.-A.H.; Logan, B.R.; Wu, J.; Wingard, J.R.; Weisdorf, D.J.; Mudrick, C.; Knust, K.; Horowitz, M.M.; Confer, D.L.; Dubberke, E.R.; et al. Infections after Transplantation of Bone Marrow or Peripheral Blood Stem Cells from Unrelated Donors. Biol. Blood Marrow Transplant. 2016, 22, 359–370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Anderson, D.; DeFor, T.; Burns, L.; McGlave, P.; Miller, J.; Wagner, J.; Weisdorf, D. A Comparison of Related Donor Peripheral Blood and Bone Marrow Transplants: Importance of Late-Onset Chronic Graft-versus-Host Disease and Infections. Biol. Blood Marrow Transplant. 2003, 9, 52–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Parody, R.; Martino, R.; Rovira, M.; Vazquez, L.; Vázquez, M.J.; de la Cámara, R.; Blazquez, C.; Fernández-Avilés, F.; Carreras, E.; Salavert, M.; et al. Severe Infections after Unrelated Donor Allogeneic Hematopoietic Stem Cell Transplantation in Adults: Comparison of Cord Blood Transplantation with Peripheral Blood and Bone Marrow Transplantation. Biol. Blood Marrow Transplant. 2006, 12, 734–748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Akhmedov, M. Infectious Complications in Allogeneic Hematopoietic Cell Transplant Recipients: Review of Transplant-related Risk Factors and Current State of Prophylaxis. Clin. Transplant. 2021, 35, e14172. [Google Scholar] [CrossRef]
  29. Lussana, F.; Cattaneo, M.; Rambaldi, A.; Squizzato, A. Ruxolitinib-associated Infections: A Systematic Review and Meta-analysis. Am. J. Hematol. 2018, 93, 339–347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. National Comprehensive Cancer Network. Referenced with Permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Hematopoietic Cell Transplantation (HCT); Version 2.2022; National Comprehensive Cancer Network, Inc.: Plymouth Meeting, PA, USA, 2022. [Google Scholar]
  31. Goldstein, E.J.C.; Owens, R.C., Jr.; Nolin, T.D. Antimicrobial-Associated QT Interval Prolongation: Pointes of Interest. Clin. Infect. Dis. 2006, 43, 1603–1611. [Google Scholar] [CrossRef] [Green Version]
  32. Teo, Y.L.; Ho, H.K.; Chan, A. Metabolism-related Pharmacokinetic Drug−drug Interactions with Tyrosine Kinase Inhibitors: Current Understanding, Challenges and Recommendations. Br. J. Clin. Pharmacol. 2015, 79, 241–253. [Google Scholar] [CrossRef] [Green Version]
  33. Leather, H.L. Drug Interactions in the Hematopoietic Stem Cell Transplant (HSCT) Recipient: What Every Transplanter Needs to Know. Bone Marrow Transplant. 2004, 33, 137–152. [Google Scholar] [CrossRef] [Green Version]
  34. Glotzbecker, B.; Duncan, C.; Alyea, E.; Campbell, B.; Soiffer, R. Important Drug Interactions in Hematopoietic Stem Cell Transplantation: What Every Physician Should Know. Biol. Blood Marrow Transplant. 2012, 18, 989–1006. [Google Scholar] [CrossRef] [Green Version]
  35. Klastersky, J. Management of Fever in Neutropenic Patients with Different Risks of Complications. Clin. Infect. Dis. 2004, 39, S32–S37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Nesher, L.; Rolston, K.V.I. The Current Spectrum of Infection in Cancer Patients with Chemotherapy Related Neutropenia. Infection 2014, 42, 5–13. [Google Scholar] [CrossRef]
  37. Pillinger, K.E.; Bouchard, J.; Withers, S.T.; Mediwala, K.; McGee, E.U.; Gibson, G.M.; Bland, C.M.; Bookstaver, P.B. Inpatient Antibiotic Stewardship Interventions in the Adult Oncology and Hematopoietic Stem Cell Transplant Population: A Review of the Literature. Ann. Pharmacother. 2020, 54, 594–610. [Google Scholar] [CrossRef]
  38. Amanati, A.; Sajedianfard, S.; Khajeh, S.; Ghasempour, S.; Mehrangiz, S.; Nematolahi, S.; Shahhosein, Z. Bloodstream Infections in Adult Patients with Malignancy, Epidemiology, Microbiology, and Risk Factors Associated with Mortality and Multi-Drug Resistance. BMC Infect. Dis. 2021, 21, 636. [Google Scholar] [CrossRef] [PubMed]
  39. Carvalho, A.S.; Lagana, D.; Catford, J.; Shaw, D.; Bak, N. Bloodstream Infections in Neutropenic Patients with Haematological Malignancies. Infect. Dis. Health 2020, 25, 22–29. [Google Scholar] [CrossRef] [PubMed]
  40. Le Clech, L.; Talarmin, J.-P.; Couturier, M.-A.; Ianotto, J.-C.; Nicol, C.; Le Calloch, R.; Dos Santos, S.; Hutin, P.; Tandé, D.; Cogulet, V.; et al. Early Discontinuation of Empirical Antibacterial Therapy in Febrile Neutropenia: The ANTIBIOSTOP Study. Infect. Dis. 2018, 50, 539–549. [Google Scholar] [CrossRef]
  41. Aguilar-Guisado, M.; Espigado, I.; Martín-Peña, A.; Gudiol, C.; Royo-Cebrecos, C.; Falantes, J.; Vázquez-López, L.; Montero, M.I.; Rosso-Fernández, C.; de la Luz Martino, M.; 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, e573–e583. [Google Scholar] [CrossRef]
  42. Averbuch, D.; Orasch, C.; Cordonnier, C.; Livermore, D.M.; Mikulska, M.; Viscoli, C.; Gyssens, I.C.; Kern, W.V.; Klyasova, G.; Marchetti, O.; 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, 1826–1835. [Google Scholar] [CrossRef] [Green Version]
  43. Satlin, M.J.; Weissman, S.J.; Carpenter, P.A.; Seo, S.K.; Shelburne, S.A. American Society of Transplantation and Cellular Therapy Series, 1: Enterobacterales Infection Prevention and Management after Hematopoietic Cell Transplantation. Transplant. Cell. Ther. 2021, 27, 108–114. [Google Scholar] [CrossRef]
  44. Bucaneve, G.; Micozzi, A.; Menichetti, F.; Martino, P.; Dionisi, M.S.; Martinelli, G.; Allione, B.; D’Antonio, D.; Buelli, M.; Nosari, A.M.; et al. Levofloxacin to Prevent Bacterial Infection in Patients with Cancer and Neutropenia. N. Engl. J. Med. 2005, 353, 977–987. [Google Scholar] [CrossRef] [Green Version]
  45. Bartlett, J.G.; Perl, T.M. The New Clostridium Difficile—What Does It Mean? N. Engl. J. Med. 2005, 353, 2503–2505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Satlin, M.J.; Chavda, K.D.; Baker, T.M.; Chen, L.; Shashkina, E.; Soave, R.; Small, C.B.; Jacobs, S.E.; Shore, T.B.; van Besien, K.; et al. Colonization With Levofloxacin-Resistant Extended-Spectrum β-Lactamase-Producing Enterobacteriaceae and Risk of Bacteremia in Hematopoietic Stem Cell Transplant Recipients. Clin. Infect. Dis. 2018, 67, 1720–1728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Zimmer, A.J.; Freifeld, A.G. Optimal Management of Neutropenic Fever in Patients With Cancer. J. Oncol. Pract. 2019, 15, 19–24. [Google Scholar] [CrossRef] [PubMed]
  48. Flowers, C.R.; Seidenfeld, J.; Bow, E.J.; Karten, C.; Gleason, C.; Hawley, D.K.; Kuderer, N.M.; Langston, A.A.; Marr, K.A.; Rolston, K.V.I.; et al. Antimicrobial Prophylaxis and Outpatient Management of Fever and Neutropenia in Adults Treated for Malignancy: American Society of Clinical Oncology Clinical Practice Guideline. J. Clin. Oncol. 2013, 31, 794–810. [Google Scholar] [CrossRef] [Green Version]
  49. Gafter-Gvili, A.; Fraser, A.; Paul, M.; Vidal, L.; Lawrie, T.A.; van de Wetering, M.D.; Kremer, L.C.; Leibovici, L. Antibiotic Prophylaxis for Bacterial Infections in Afebrile Neutropenic Patients Following Chemotherapy. Cochrane Database Syst. Rev. 2012, 2018, CD004386. [Google Scholar] [CrossRef] [PubMed]
  50. Pagano, L.; Caira, M.; Candoni, A.; Offidani, M.; Martino, B.; Specchia, G.; Pastore, D.; Stanzani, M.; Cattaneo, C.; Fanci, R.; et al. Invasive Aspergillosis in Patients with Acute Myeloid Leukemia: A SEIFEM-2008 Registry Study. Haematologica 2010, 95, 644–650. [Google Scholar] [CrossRef] [Green Version]
  51. Hsu, L.Y.; Lee, D.G.; Yeh, S.P.; Bhurani, D.; Khanh, B.Q.; Low, C.Y.; Norasetthada, L.; Chan, T.; Kwong, Y.L.; Vaid, A.K.; et al. Epidemiology of Invasive Fungal Diseases among Patients with Haematological Disorders in the Asia-Pacific: A Prospective Observational Study. Clin. Microbiol. Infect. 2015, 21, 594.e7–594.e11. [Google Scholar] [CrossRef] [Green Version]
  52. Kontoyiannis, D.P.; Marr, K.A.; Park, B.J.; Alexander, B.D.; Anaissie, E.J.; Walsh, T.J.; Ito, J.; Andes, D.R.; Baddley, J.W.; Brown, J.M.; et al. Prospective Surveillance for Invasive Fungal Infections in Hematopoietic Stem Cell Transplant Recipients, 2001–2006: Overview of the Transplant-Associated Infection Surveillance Network (TRANSNET) Database. Clin. Infect. Dis. 2010, 50, 1091–1100. [Google Scholar] [CrossRef]
  53. Neofytos, D.; Horn, D.; Anaissie, E.; Steinbach, W.; Olyaei, A.; Fishman, J.; Pfaller, M.; Chang, C.; Webster, K.; Marr, K. Epidemiology and Outcome of Invasive Fungal Infection in Adult Hematopoietic Stem Cell Transplant Recipients: Analysis of Multicenter Prospective Antifungal Therapy (PATH) Alliance Registry. Clin. Infect. Dis. 2009, 48, 265–273. [Google Scholar] [CrossRef]
  54. Donnelly, J.P.; Chen, S.C.; Kauffman, C.A.; Steinbach, W.J.; Baddley, J.W.; Verweij, P.E.; Clancy, C.J.; Wingard, J.R.; Lockhart, S.R.; Groll, A.H.; et al. Revision and Update of the Consensus Definitions of Invasive Fungal Disease From the European Organization for Research and Treatment of Cancer and the Mycoses Study Group Education and Research Consortium. Clin. Infect. Dis. 2020, 71, 1367–1376. [Google Scholar] [CrossRef] [Green Version]
  55. Patterson, T.F.; Thompson, G.R., III; Denning, D.W.; Fishman, J.A.; Hadley, S.; Herbrecht, R.; Kontoyiannis, D.P.; Marr, K.A.; Morrison, V.A.; Nguyen, M.H.; et al. Practice Guidelines for the Diagnosis and Management of Aspergillosis: 2016 Update by the Infectious Diseases Society of America. Clin. Infect. Dis. 2016, 63, e1–e60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Uneno, Y.; Imura, H.; Makuuchi, Y.; Tochitani, K.; Watanabe, N. Pre-Emptive Antifungal Therapy versus Empirical Antifungal Therapy for Febrile Neutropenia in People with Cancer. Cochrane Database Syst. Rev. 2022, 2022, 5. [Google Scholar] [CrossRef]
  57. Maertens, J.A.; Girmenia, C.; Brüggemann, R.J.; Duarte, R.F.; Kibbler, C.C.; Ljungman, P.; Racil, Z.; Ribaud, P.; Slavin, M.A.; Cornely, O.A.; et al. European Guidelines for Primary Antifungal Prophylaxis in Adult Haematology Patients: Summary of the Updated Recommendations from the European Conference on Infections in Leukaemia. J. Antimicrob. Chemother. 2018, 73, 3221–3230. [Google Scholar] [CrossRef] [PubMed]
  58. Khanina, A.; Tio, S.Y.; Ananda-Rajah, M.R.; Kidd, S.E.; Williams, E.; Chee, L.; Urbancic, K.; Thursky, K.A.; Australasian Antifungal Guidelines Steering Committee; Slavin, M.A.; et al. Consensus Guidelines for Antifungal Stewardship, Surveillance and Infection Prevention, 2021. Intern. Med. J. 2021, 51, 18–36. [Google Scholar] [CrossRef] [PubMed]
  59. Johnson, M.D.; Lewis, R.E.; Dodds Ashley, E.S.; Ostrosky-Zeichner, L.; Zaoutis, T.; Thompson, G.R.; Andes, D.R.; Walsh, T.J.; Pappas, P.G.; Cornely, O.A.; et al. Core Recommendations for Antifungal Stewardship: A Statement of the Mycoses Study Group Education and Research Consortium. J. Infect. Dis. 2020, 222, S175–S198. [Google Scholar] [CrossRef]
  60. Barlam, T.F.; Cosgrove, S.E.; Abbo, L.M.; MacDougall, C.; Schuetz, A.N.; Septimus, E.J.; Srinivasan, A.; Dellit, T.H.; Falck-Ytter, Y.T.; Fishman, N.O.; 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, e51–e77. [Google Scholar] [CrossRef] [Green Version]
  61. Science, M.; Timberlake, K. Antifungal Stewardship: A Budding Branch of Antimicrobial Stewardship. Pediatr. Blood Cancer 2020, 67, e28145. [Google Scholar] [CrossRef]
  62. Michallet, M.; Sobh, M.; Deray, G.; Gangneux, J.-P.; Pigneux, A.; Larrey, D.; Ribaud, P.; Mira, J.-P.; Nivoix, Y.; Yakoub-Agha, I.; et al. Antifungal Stewardship in Hematology: Reflection of a Multidisciplinary Group of Experts. Clin. Lymphoma Myeloma Leuk. 2021, 21, 35–45. [Google Scholar] [CrossRef]
  63. Einsele, H.; Ljungman, P.; Boeckh, M. How I Treat CMV Reactivation after Allogeneic Hematopoietic Stem Cell Transplantation. Blood 2020, 135, 1619–1629. [Google Scholar] [CrossRef]
  64. Green, M.L.; Leisenring, W.; Xie, H.; Mast, T.C.; Cui, Y.; Sandmaier, B.M.; Sorror, M.L.; Goyal, S.; Özkök, S.; Yi, J.; et al. Cytomegalovirus Viral Load and Mortality after Haemopoietic Stem Cell Transplantation in the Era of Pre-Emptive Therapy: A Retrospective Cohort Study. Lancet Haematol. 2016, 3, e119–e127. [Google Scholar] [CrossRef] [Green Version]
  65. Merck & Co., Inc. Letermovir [Package Insert]. 2022. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/209939Orig1s000,209940Orig1s000lbl.pdf (accessed on 22 January 2023).
  66. Cesaro, S.; Ljungman, P.; Tridello, G.; Mikulska, M.; Wendel, L.; Styczynski, J.; Averbuch, D.; de la Camara, R. New Trends in the Management of Cytomegalovirus Infection after Allogeneic Hematopoietic Cell Transplantation: A Survey of the Infectious Diseases Working Pary of EBMT. Bone Marrow Transplant. 2022, 58, 203–208. [Google Scholar] [CrossRef] [PubMed]
  67. Dadwal, S.S.; Papanicolaou, G.; Boeckh, M. How I Prevent Viral Reactivation in High-Risk Patients. Blood 2022. [Google Scholar] [CrossRef] [PubMed]
  68. Sperotto, A.; Candoni, A.; Gottardi, M.; Facchin, G.; Stella, R.; De Marchi, R.; Michelutti, A.; Cavallin, M.; Rosignoli, C.; Patriarca, F.; et al. Cytomegalovirus Prophylaxis versus Pre-Emptive Strategy: Different CD4+ and CD8+ T Cell Reconstitution after Allogeneic Hematopoietic Stem Cell Transplantation. Transplant. Cell. Ther. 2021, 27, 518.e1–518.e4. [Google Scholar] [CrossRef] [PubMed]
  69. Takeda Pharmaceuticals, Inc. Maribavir [Package Insert]. 2021. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/215596lbl.pdf (accessed on 22 January 2023).
  70. Gilead Sciences, Inc. Cidofovir [Package Insert]. 2000. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/1999/020638s003lbl.pdf (accessed on 22 January 2023).
  71. Jorgenson, M.R.; Descourouez, J.L.; Kleiboeker, H.; Goldrosen, K.; Schulz, L.; Rice, J.P.; Odorico, J.S.; Mandelbrot, D.A.; Smith, J.A.; Saddler, C.M. Cytomegalovirus Antiviral Stewardship in Solid Organ Transplant Recipients: A New Gold Standard. Transpl. Infect. Dis. 2022, 24, e13864. [Google Scholar] [CrossRef]
  72. Jorgenson, M.R.; Descourouez, J.L.; Schulz, L.T.; Goldrosen, K.A.; Rice, J.P.; Redfield, R.R.; Saddler, C.M.; Smith, J.A.; Mandelbrot, D.A. The Development and Implementation of Stewardship Initiatives to Optimize the Prevention and Treatment of Cytomegalovirus Infection in Solid-Organ Transplant Recipients. Infect. Control Hosp. Epidemiol. 2020, 41, 1068–1074. [Google Scholar] [CrossRef]
  73. Anton-Vazquez, V.; Mehra, V.; Mbisa, J.L.; Bradshaw, D.; Basu, T.N.; Daly, M.-L.; Mufti, G.J.; Pagliuca, A.; Potter, V.; Zuckerman, M. Challenges of Aciclovir-Resistant HSV Infection in Allogeneic Bone Marrow Transplant Recipients. J. Clin. Virol. 2020, 128, 104421. [Google Scholar] [CrossRef]
  74. Piret, J.; Boivin, G. Resistance of Herpes Simplex Viruses to Nucleoside Analogues: Mechanisms, Prevalence, and Management. Antimicrob. Agents Chemother. 2011, 55, 459–472. [Google Scholar] [CrossRef] [Green Version]
  75. Bartash, R.; McCort, M.E.; Cowman, K.; Orner, E.; Szymczak, W.; Nori, P.; Nori, P. 192. A Hematology/Oncology Unit-Specific Antibiogram Emphasizes the Need for Intensified Local Stewardship. Open Forum Infect. Dis. 2020, 7, S101–S102. [Google Scholar] [CrossRef]
  76. Smith, Z.R.; Tajchman, S.K.; Dee, B.M.; Bruno, J.J.; Qiao, W.; Tverdek, F.P. Development of a Combination Antibiogram for Pseudomonas Aeruginosa Bacteremia in an Oncology Population. J. Oncol. Pharm. Pract. 2016, 22, 409–415. [Google Scholar] [CrossRef]
  77. Abt, M.C.; McKenney, P.T.; Pamer, E.G. Clostridium Difficile Colitis: Pathogenesis and Host Defence. Nat. Rev. Microbiol. 2016, 14, 609–620. [Google Scholar] [CrossRef]
  78. Schuster, M.G.; Cleveland, A.A.; Dubberke, E.R.; Kauffman, C.A.; Avery, R.K.; Husain, S.; Paterson, D.L.; Silveira, F.P.; Chiller, T.M.; Benedict, K.; et al. Infections in Hematopoietic Cell Transplant Recipients: Results From the Organ Transplant Infection Project, a Multicenter, Prospective, Cohort Study. Open Forum Infect. Dis. 2017, 4, ofx050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Misch, E.A.; Safdar, N. Clostridioides Difficile Infection in the Stem Cell Transplant and Hematologic Malignancy Population. Infect. Dis. Clin. N. Am. 2019, 33, 447–466. [Google Scholar] [CrossRef]
  80. Shouval, R.; Waters, N.R.; Gomes, A.L.C.; Zuanelli Brambilla, C.; Fei, T.; Devlin, S.M.; Nguyen, C.L.; Markey, K.A.; Dai, A.; Slingerland, J.B.; et al. Conditioning Regimens Are Associated with Distinct Patterns of Microbiota Injury in Allogeneic Hematopoietic Cell Transplantation. Clin. Cancer Res. 2023, 29, 165–173. [Google Scholar] [CrossRef] [PubMed]
  81. Holler, E.; Butzhammer, P.; Schmid, K.; Hundsrucker, C.; Koestler, J.; Peter, K.; Zhu, W.; Sporrer, D.; Hehlgans, T.; Kreutz, M.; et al. Metagenomic Analysis of the Stool Microbiome in Patients Receiving Allogeneic Stem Cell Transplantation: Loss of Diversity Is Associated with Use of Systemic Antibiotics and More Pronounced in Gastrointestinal Graft-versus-Host Disease. Biol. Blood Marrow Transplant. 2014, 20, 640–645. [Google Scholar] [CrossRef] [Green Version]
  82. Khan, N.; Lindner, S.; Gomes, A.L.C.; Devlin, S.M.; Shah, G.L.; Sung, A.D.; Sauter, C.S.; Landau, H.J.; Dahi, P.B.; Perales, M.-A.; et al. Fecal Microbiota Diversity Disruption and Clinical Outcomes after Auto-HCT: A Multicenter Observational Study. Blood 2021, 137, 1527–1537. [Google Scholar] [CrossRef]
  83. Alonso, C.D.; Maron, G.; Kamboj, M.; Carpenter, P.A.; Gurunathan, A.; Mullane, K.M.; Dubberke, E.R. American Society for Transplantation and Cellular Therapy Series: #5—Management of Clostridioides Difficile Infection in Hematopoietic Cell Transplant Recipients. Transplant. Cell. Ther. 2022, 28, 225–232. [Google Scholar] [CrossRef]
  84. Galloway-Peña, J.R.; Shi, Y.; Peterson, C.B.; Sahasrabhojane, P.; Gopalakrishnan, V.; Brumlow, C.E.; Daver, N.G.; Alfayez, M.; Boddu, P.C.; Khan, M.A.W.; et al. Gut Microbiome Signatures Are Predictive of Infectious Risk Following Induction Therapy for Acute Myeloid Leukemia. Clin. Infect. Dis. 2020, 71, 63–71. [Google Scholar] [CrossRef]
  85. Weber, D.; Jenq, R.R.; Peled, J.U.; Taur, Y.; Hiergeist, A.; Koestler, J.; Dettmer, K.; Weber, M.; Wolff, D.; Hahn, J.; et al. Microbiota Disruption Induced by Early Use of Broad-Spectrum Antibiotics Is an Independent Risk Factor of Outcome after Allogeneic Stem Cell Transplantation. Biol. Blood Marrow Transplant. 2017, 23, 845–852. [Google Scholar] [CrossRef] [Green Version]
  86. Peled, J.U.; Gomes, A.L.C.; Devlin, S.M.; Littmann, E.R.; Taur, Y.; Sung, A.D.; Weber, D.; Hashimoto, D.; Slingerland, A.E.; Slingerland, J.B.; et al. Microbiota as Predictor of Mortality in Allogeneic Hematopoietic-Cell Transplantation. N. Engl. J. Med. 2020, 382, 822–834. [Google Scholar] [CrossRef]
  87. Shono, Y.; Docampo, M.D.; Peled, J.U.; Perobelli, S.M.; Velardi, E.; Tsai, J.J.; Slingerland, A.E.; Smith, O.M.; Young, L.F.; Gupta, J.; et al. Increased GVHD-Related Mortality with Broad-Spectrum Antibiotic Use after Allogeneic Hematopoietic Stem Cell Transplantation in Human Patients and Mice. Sci. Transl. Med. 2016, 8, 339ra71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Tanaka, J.S.; Young, R.R.; Heston, S.M.; Jenkins, K.; Spees, L.P.; Sung, A.D.; Corbet, K.; Thompson, J.C.; Bohannon, L.; Martin, P.L.; et al. Anaerobic Antibiotics and the Risk of Graft-versus-Host Disease after Allogeneic Hematopoietic Stem Cell Transplantation. Biol. Blood Marrow Transplant. 2020, 26, 2053–2060. [Google Scholar] [CrossRef] [PubMed]
  89. Deak, E.; Charlton, C.L.; Bobenchik, A.M.; Miller, S.A.; Pollett, S.; McHardy, I.H.; Wu, M.T.; Garner, O.B. Comparison of the Vitek MS and Bruker Microflex LT MALDI-TOF MS Platforms for Routine Identification of Commonly Isolated Bacteria and Yeast in the Clinical Microbiology Laboratory. Diagn. Microbiol. Infect. Dis. 2015, 81, 27–33. [Google Scholar] [CrossRef] [PubMed]
  90. Charnot-Katsikas, A.; Tesic, V.; Love, N.; Hill, B.; Bethel, C.; Boonlayangoor, S.; Beavis, K.G. Use of the Accelerate Pheno System for Identification and Antimicrobial Susceptibility Testing of Pathogens in Positive Blood Cultures and Impact on Time to Results and Workflow. J. Clin. Microbiol. 2018, 56, e01166-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Giacobbe, D.R.; Giani, T.; Bassetti, M.; Marchese, A.; Viscoli, C.; Rossolini, G.M. Rapid Microbiological Tests for Bloodstream Infections Due to Multidrug Resistant Gram-Negative Bacteria: Therapeutic Implications. Clin. Microbiol. Infect. 2020, 26, 713–722. [Google Scholar] [CrossRef]
  92. Banerjee, R.; Teng, C.B.; Cunningham, S.A.; Ihde, S.M.; Steckelberg, J.M.; Moriarty, J.P.; Shah, N.D.; Mandrekar, J.N.; Patel, R. Randomized Trial of Rapid Multiplex Polymerase Chain Reaction–Based Blood Culture Identification and Susceptibility Testing. Clin. Infect. Dis. 2015, 61, 1071–1080. [Google Scholar] [CrossRef] [Green Version]
  93. Banerjee, R.; Komarow, L.; Virk, A.; Rajapakse, N.; Schuetz, A.N.; Dylla, B.; Earley, M.; Lok, J.; Kohner, P.; Ihde, S.; et al. Randomized Trial Evaluating Clinical Impact of RAPid Identification and Susceptibility Testing for Gram-Negative Bacteremia: RAPIDS-GN. Clin. Infect. Dis. 2021, 73, e39–e46. [Google Scholar] [CrossRef]
  94. Douglas, A.P.; Hall, L.; James, R.S.; Worth, L.J.; Slavin, M.A.; Thursky, K.A. Quality of Inpatient Antimicrobial Use in Hematology and Oncology Patients. Infect. Control. Hosp. Epidemiol. 2021, 42, 1235–1244. [Google Scholar] [CrossRef]
  95. Aitken, S.L.; Nagel, J.L.; Abbo, L.; Alegria, W.; Barreto, J.N.; Dadwal, S.; Freifeld, A.G.; Jain, R.; Pergam, S.A.; Tverdek, F.P.; et al. Antimicrobial Stewardship in Cancer Patients: The Time Is Now. J. Natl. Compr. Canc. Netw. 2019, 17, 772–775. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Phases of opportunistic infections among allogeneic HSCT recipients and examples of prevention strategies—Adapted with permission from Ref. [23]. Copyright 2016, Elsevier. 1 Reference [11]. 2 Reference [24].
Figure 1. Phases of opportunistic infections among allogeneic HSCT recipients and examples of prevention strategies—Adapted with permission from Ref. [23]. Copyright 2016, Elsevier. 1 Reference [11]. 2 Reference [24].
Antibiotics 12 00592 g001
Figure 2. Multidisciplinary approach for ASP interventions in HSCT and oncology patients—Adapted with permission from Ref. [4]. Copyright 2022, Elsevier.
Figure 2. Multidisciplinary approach for ASP interventions in HSCT and oncology patients—Adapted with permission from Ref. [4]. Copyright 2022, Elsevier.
Antibiotics 12 00592 g002
Table 1. Examples of ASP Interventions Specific to Oncology Patients Aligned with CDC Core Elements.
Table 1. Examples of ASP Interventions Specific to Oncology Patients Aligned with CDC Core Elements.
CDC Core ElementSample Interventions Focused on HSCT/Oncology Patients
Hospital Leadership Commitment
Dedicate necessary human, financial, and information technology resources.
Accessible information systems (e.g., electronic medical record, surveillance data)
Dedicated staff for antimicrobial stewardship
Accountability
Appoint a leader or co-leaders, such as a physician and pharmacist, responsible for program management and outcomes.
Multidisciplinary approach among hematology/oncology, infectious disease, and pharmacy (“handshake stewardship”)
Pharmacy Expertise
Appoint a pharmacist, ideally as the co-leader of the stewardship program, to lead implementation efforts to improve antibiotic use.
Antibacterial, antifungal, and antiviral prophylaxis
Dose optimization (e.g., extended infusion of beta-lactams)
Duration of empiric antimicrobials for febrile neutropenia
IV to PO conversion
Action
Implement interventions, such as prospective auditing and feedback, or preauthorization, to improve antibiotic use.
Development of population specific guidelines
Febrile neutropenia
Antifungal prophylaxis and treatment
Cytomegalovirus prophylaxis
Use of microbiology methods to assist with prescribing
Tracking
Monitor antibiotic prescribing, impact of interventions, and other important outcomes such as C. difficile infection and resistance patterns.
Population- and/or unit-specific antibiograms
Prevalence of MDRO
Prospective audit and formulary restriction
Reporting
Regularly report information on antibiotic use and resistance to prescribers, pharmacists, nurses, and hospital leadership.
Tracking and shared reporting of outcomes specific to HSCT/oncology
C. difficile
Catheter-related infections
Prevalence of MDRO
Education
Educate prescribers, pharmacists, and nurses about adverse reactions to antibiotics, antibiotic resistance, and optimal prescribing.
Population-specific antibiograms
Microbiome diversity
Abbreviations: ASP = antimicrobial stewardship program, CDC = Centers for Disease Control and Prevention, C. difficile = Clostridioides difficile, HSCT = hematopoietic stem cell transplant, IV = intravenous, MDRO = multi-drug resistant organism, PO = by mouth.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Majumdar, A.; Shah, M.R.; Park, J.J.; Narayanan, N.; Kaye, K.S.; Bhatt, P.J. Challenges and Opportunities in Antimicrobial Stewardship among Hematopoietic Stem Cell Transplant and Oncology Patients. Antibiotics 2023, 12, 592. https://doi.org/10.3390/antibiotics12030592

AMA Style

Majumdar A, Shah MR, Park JJ, Narayanan N, Kaye KS, Bhatt PJ. Challenges and Opportunities in Antimicrobial Stewardship among Hematopoietic Stem Cell Transplant and Oncology Patients. Antibiotics. 2023; 12(3):592. https://doi.org/10.3390/antibiotics12030592

Chicago/Turabian Style

Majumdar, Anjali, Mansi R. Shah, Jiyeon J. Park, Navaneeth Narayanan, Keith S. Kaye, and Pinki J. Bhatt. 2023. "Challenges and Opportunities in Antimicrobial Stewardship among Hematopoietic Stem Cell Transplant and Oncology Patients" Antibiotics 12, no. 3: 592. https://doi.org/10.3390/antibiotics12030592

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop