Overview
The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for the Prevention and Treatment of Cancer-Related Infections are largely divided into 5 sections comprising discussions on the following: (1) risk factors for infection (major host factors that predispose patients to infectious diseases); (2) prevention of infectious complications (including the use of antimicrobial prophylaxis and pre-emptive therapy); (3) infection concerns and recommendations regarding immune and targeted treatments; (4) management of neutropenic fever; and (5) management of site-specific infections (eg, pneumonia, abdominal infections, catheter-associated infections). These guidelines provide a framework for prevention and treatment of infections that should be applied in conjunction with careful, individualized patient evaluation and with an understanding of both the host factors that predispose patients to specific infectious diseases and antimicrobial susceptibility patterns. Additionally, the guidelines are based primarily on studies with adult patients, and application of these recommendations to pediatric patients may differ. Consultation with an infectious disease expert is highly recommended.
CAR T-Cell Therapy
Several CAR T-cell products targeting CD19 (eg, axicabtagene ciloleucel, tisagenlecleucel, lisocabtagene maraleucel) or B-cell maturation antigen (eg, idecabtagene vicleucel, ciltacabtagene autoleucel) have been approved for hematologic malignancies. However, CAR T-cell products confer a risk of potentially life-threatening immunologic toxicities, like neutropenia, lymphopenia, and hypogammaglobulinemia, and thus may increase the risk of infection in patients.1 The highest risk for infection occurs within the first 30 days of administration of CAR T-cell therapy, and bacterial infections predominate during this period. Opportunistic infections can be expected, and prophylaxis, particularly for the prevention of Pneumocystis jirovecii pneumonia (PJP), herpes simplex virus (HSV), and varicella zoster virus (VZV), is warranted until immune reconstitution. Preinfusion risk factors for infection include certain underlying malignancies, prior lines of therapy with or without autologous/allogeneic hematopoietic cell transplantation (HCT), antecedent infection, and neutropenia. Risk factors postinfusion may include prolonged neutropenia, cytokine release syndrome (CRS)/immune effector cell-associated neurotoxicity syndrome and associated treatment (eg, high-dose steroids, IL-6 inhibitors), lymphopenia, and hypogammaglobulinemia. Beyond day +30, respiratory viral infections predominate. Fungal infection risk remains low but may vary based on prior therapies, degree of immunosuppression, and other relevant risk factors.
Relevant serologic screening for HIV, hepatitis B virus (HBV), and hepatitis C virus (HCV) is recommended. Moreover, cytomegalovirus (CMV) and additional screening may be considered based on epidemiologic risks. Antibacterial and antifungal prophylaxis can be considered for patients who are neutropenic. PJP and HSV/VZV prophylaxis are recommended. The specific risk profile of the patient (duration of neutropenia, prior allogeneic HCT, previous infections, and local antibiotic resistance profiles) should guide diagnostic workup and selection of antimicrobial agents. In patients with additional risk factors, such as prolonged neutropenia, prior allogeneic HCT, or augmented immunosuppressive therapy (IST) for CRS/immune effector cell-associated neurotoxicity syndrome, mold-active antifungal prophylaxis can be considered (see Figures 1 and 2).1–4 Patients should also be monitored for CRS, which may mimic sepsis. The incidence of CRS depends on the CAR T-cell product, disease characteristics, and CRS grading system used.1–4
Prevention of Infectious Diseases
Preventive measures against infections in patients with cancer include upfront prophylaxis or pre-emptive therapy using broad-spectrum antimicrobial agents directed against the most common infecting pathogens (including bacterial, viral, and fungal) in patients at high risk for infections. Vaccination and minimization of potential exposures to opportunistic pathogens that may be harmful to patients who are immunocompromised due to cancer are additional components of infectious disease prevention.
Antibacterial Prophylaxis
During Neutropenia
Patients with cancer and chemotherapy-induced neutropenia are at risk for severe bacterial infections. Fluoroquinolones are the most commonly used prophylactic antibacterial agents in adults with chemotherapy-induced neutropenia.
Two large, randomized, placebo-controlled studies showed the benefit of levofloxacin prophylaxis in neutropenia at different levels of risk for infectious complications.5,6 The main advantage of levofloxacin prophylaxis (in patients with intermediate and higher risk of infections) with chemotherapy-induced neutropenia was a reduction in clinically significant bacterial infections, including gram-negative rod bacteremia.5 In contrast, the main advantage of prophylaxis in lower risk neutropenia was a small, but statistically significant, reduction in fever and hospitalization for neutropenic fever.6 Neither study conducted a systematic long-term evaluation of antimicrobial resistance. The NCCN panel considers that reduction in the incidence of significant infections is a more clinically meaningful endpoint than reduction in the incidence of neutropenic fever. Using prevention of neutropenic fever as the primary endpoint in this study by Cullen et al6 1,000 hypothetical patients with low risk of infections would have to receive prophylaxis during each cycle of chemotherapy-induced neutropenia to benefit only 44 patients.
In a single-center randomized study in patients undergoing high-dose therapy followed by autologous HCT (n=157), patients were randomized to receive prophylaxis (with oral ciprofloxacin 500 mg twice daily and intravenous vancomycin 1,000 mg once daily) or no prophylaxis; all patients received antifungal prophylaxis with fluconazole.7 Empirical therapy (comprising amikacin, ceftazidime, and full-dose vancomycin) was initiated when neutropenic fever developed. The use of antibacterial prophylaxis significantly reduced the incidences of neutropenic fever (56% vs 91%; P<.001) and bacteremia (6% vs 35%; P=.005) compared with no prophylaxis, but at the expense of decreased response to first-line empirical therapy (66% vs 84%; P=.025). Among the patients who received prophylaxis and developed neutropenic fever, 34% required second-line therapy that included a carbapenem, suggesting that these patients developed infections resistant to the prophylactic regimen. Duration of hospitalization and overall survival rates were similar between study arms. These results led the study investigators to conclude that routine antibacterial prophylaxis was not recommended in patients undergoing high-dose therapy and autologous HCT.7 It should be noted, however, that the prophylactic regimen in this study included vancomycin (albeit at a lower dose) and is not supported by the NCCN or IDSA panels for use as either antimicrobial prophylaxis or standard initial empirical therapy for fever and neutropenia.8
In a systematic review and meta-analysis by Gafter-Gvili et al (based on 109 trials, n=13,579 patients) comparing antibacterial prophylaxis with placebo, no intervention, or prevention with another agent in patients with afebrile neutropenia, the use of antibacterial prophylaxis was found to significantly reduce the risk of all-cause mortality (risk ratio, 0.66; 95% CI, 0.55–0.79) as well as infection-related deaths (risk ratio, 0.61; 95% CI, 0.48–0.77) compared with placebo or no intervention.9 The use of prophylaxis also significantly reduced the incidence of fever and clinically or microbiologically documented infections. Although no significant differences in all-cause or infection-related mortality were seen between prophylactic quinolones or trimethoprim/sulfamethoxazole (TMP/SMX), the use of quinolones was associated with decreased drug resistance and fewer adverse events that subsequently reduced the incidence of drug discontinuation.9
A systematic review and meta-analysis evaluated the risks associated with colonization and infections by fluoroquinolone-resistant bacteria.10 Most of the studies (48 of 56 trials) included patients with hematologic malignancies or HCT recipients. Results of the analysis (based on 56 trials, n=7,878 patients; data on colonization by resistant bacteria based on 27 trials) showed that quinolone prophylaxis was associated with an increase in colonization with quinolone-resistant organisms compared with placebo or no intervention, although the increase was not statistically significant (relative risk [RR], 1.68; 95% CI, 0.71–4.00). However, no difference was observed in the incidence of infections caused by quinolone-resistant organisms (RR, 1.04; 95% CI, 0.73–1.50), regardless of whether these were resistant gram-negative or gram-positive bacteria.10 Moreover, in an analysis of trials comparing quinolones with TMP/SMX (11 trials), prophylaxis with quinolones was associated with fewer incidents of colonization and infections by resistant bacteria (those resistant to the prophylactic agents) compared with the use of TMP/SMX.10 This analysis suggests that prophylaxis with quinolones does not appear to increase the rate of infections by resistant organisms.
A 2017 prospective intercontinental study assessed gram-negative rod resistance to fluoroquinolones, noncarbapenem β-lactams (including ceftazidime, cefepime or β-lactam/β-lactamase inhibitors), and carbapenems (meropenem/imipenem/doripenem) in HCT recipients. Of 655 gram-negative rod bacteremia episodes, half were found to be fluoroquinolone and noncarbapenem resistant. The total resistance rates were higher in patients with allogeneic HCT than in those with autologous HCT. Risk factors for fluoroquinolone resistance in allogeneic HCT included prolonged neutropenia and breakthrough on fluoroquinolones. Mortality was found to be significantly more common in infections caused by resistant bacteria.11
A 2018 meta-analysis of studies published in the years 2006–2014 assessed the role of fluoroquinolone prophylaxis during neutropenia in consideration of increasing antibiotic resistance globally.12 The meta-analysis found no effect of the background rate of fluoroquinolone resistance on the efficacy of fluoroquinolone prophylaxis. However, in a few studies, fluoroquinolone prophylaxis resulted in an increased colonization or infection with fluoroquinolone- or multidrug-resistant strains. Fluoroquinolone prophylaxis had no effect on mortality, but it reduced the rate of bloodstream infections and episodes of fever.12
An important consideration for those with low risk with short durations of neutropenia is whether fluoroquinolone prophylaxis is of greater benefit than outpatient fluoroquinolone treatment of fever and neutropenia, should it occur. Both the NCCN Guidelines and IDSA8 panels recommend oral fluoroquinolone-based regimens as outpatient empirical therapy for neutropenic fever in adults who meet criteria for a low risk of complications. Fluoroquinolone prophylaxis may preclude its subsequent use as empirical therapy for neutropenic fever in the same patient. The modest difference in rates of hospitalization for suspected infection in levofloxacin compared with placebo recipients (15.7% vs 21.6%, respectively) may be offset by exclusion of outpatient oral empirical therapy in patients receiving fluoroquinolone prophylaxis. To limit antibacterial use, Cullen et al13 have suggested prophylaxis with levofloxacin on cycle 1 of myelosuppressive cancer chemotherapy and only in subsequent cycles if a febrile episode occurs.
The decision whether to use antibacterial prophylaxis and the selection of the specific agent requires a balance between expected benefit and risk (explained in greater detail in a review by Hoffman et al14). The concept of risk applies to immediate adverse effects of the drug (eg, rash, gastrointestinal intolerance), the potential for selection for resistant pathogens that can harm the individual receiving prophylaxis, and the risk of resistant organisms to a specific population of patients (eg, those being treated at a cancer center). Thus, the emergence of multidrug-resistant organisms, disruption of the microbiome, and antibiotic toxicities must be considered when choosing an antimicrobial prophylactic agent. The link between fluoroquinolone use and severe Clostridium difficile as well as methicillin-resistant S. aureus infections provides an additional cautionary note regarding excess use of fluoroquinolones.15–18
NCCN Recommendations for Antibacterial Prophylaxis
Antibacterial prophylaxis is not recommended for patients with a low risk of overall infection (see Figure 3). In patients with neutropenia who are at lower risk of infectious complications (a category that includes most patients with solid tumor malignancies), the main benefit of antibacterial prophylaxis is a reduction in fever rather than in documented infections. In patients with neutropenia expected to last <7 days who are not receiving immunosuppressive regimens (eg, systemic corticosteroids), the panel suggests no antibiotic prophylaxis.8 In patients deemed at intermediate or high risk, the NCCN Guidelines panel advises that fluoroquinolone prophylaxis (levofloxacin is preferred) be considered in patients with an expected duration of neutropenia (absolute neutrophil count [ANC] ≤500 neutrophils/mcL or ≤1,000 neutrophils/mcL and a predicted decline to ≤500/mcL over the next 48 hours) for >7 days. This is in agreement with the recommendations of the IDSA guidelines for the use of antimicrobial agents in patients with neutropenia with cancer.8 For patients with intolerance to fluoroquinolone, TMP/SMX or an oral third-generation cephalosporin may be considered.
Clinical practice guidelines have also been developed for systemic antibacterial prophylaxis in pediatric patients with cancer and those undergoing HCT.19 The panel for these guidelines recommends consideration of antibacterial prophylaxis in children with acute myeloid leukemia and relapsed acute lymphoblastic leukemia receiving intensive chemotherapy expected to result in severe neutropenia (ANC <500/mcL) for at least 7 days. The panel recommends against the routine use of antibacterial prophylaxis for children undergoing induction chemotherapy for ALL, autologous HCT and allogeneic HCT. A strong recommendation against its routine use was made for children whose therapy is not expected to result in prolonged severe neutropenia. If used, prophylaxis with levofloxacin is recommended as a preferred option during severe neutropenia.19
Prophylaxis for Pneumococcal Infection
Prophylaxis for pneumococcal infection is advised in allogeneic HCT recipients. Patients undergoing allogeneic HCT are at an increased risk for pneumococcal sepsis due to functional asplenia and impaired B-cell immunity. Pneumococcal sepsis is most common in the late transplant period, between 3 months to several years after HCT.20,21 IST for graft-versus-host disease (GVHD) delays reconstitution of B-cell immunity and significantly increases the risk of posttransplant pneumococcal sepsis.21,22
The NCCN Guidelines panel advises that penicillin prophylaxis be initiated at 3 months after HCT and be continued until at least 1 year after transplant in patients with GVHD. Patients should receive prophylaxis regardless of prior administration of pneumococcal vaccines.23 Prophylaxis should be continued in patients with chronic GVHD until IST has been discontinued. Posttransplant pneumococcal infection is generally community acquired, and the frequency of resistance to antibiotics reflects regional susceptibility patterns. In some regions as many as 35% of pneumococcal isolates have intermediate- or high-level resistance to penicillin,24 and cross-resistance to other classes of antibiotics is common. Breakthrough pneumococcal sepsis in HCT recipients receiving penicillin prophylaxis is well described.25 Thus, in areas with a significantly higher frequency of penicillin-resistant pneumococcal isolates, alternative agents should be considered based on local susceptibility patterns. Daily TMP/SMX used as prophylaxis for PJP is likely to be protective against pneumococcal disease but depends on local susceptibility patterns. In high-risk populations (eg, allogeneic HCT recipients with GVHD), prophylaxis with penicillin and TMP/SMX should be considered. Vaccination with pneumococcal vaccine is also strongly recommended (see “Vaccination,” page 633) 3 to 6 months post-HCT.
Antifungal Prophylaxis
Antifungal prophylaxis should not be used routinely in all patients with neutropenia. The rationale for antifungal prophylaxis is to prevent fungal infections in a targeted group of patients with high risk, especially those with longer durations of neutropenia or with GVHD after allogeneic HCT.8 Selection of an antifungal agent is determined by the disease or therapy and includes azoles, amphotericin B products, and echinocandins.
Azoles
Azoles are among the most commonly used medications for the prevention and treatment of fungal infections. Early-generation azoles such as ketoconazole are used less commonly now because of toxicity, drug interactions, and limited spectrum of activity. Some “first-generation” triazoles (ie, fluconazole and itraconazole) are used widely due to their low cost and minimal toxicity but are limited by increasing resistance among Candida species and lack of activity against most molds. Several “second-generation” triazoles (eg, voriconazole and posaconazole) have been subsequently developed. These drugs extend the spectrum of activity of triazoles to include potent activity against many molds (importantly, activity differs within the class) but can also have complicated drug interactions and distinct toxicities and remain extremely costly with extended use.
Prophylaxis with voriconazole was compared with fluconazole in a large, randomized, double-blind study that included serum galactomannan surveillance in allogeneic HCT recipients (n=600).26 Patients were randomized to receive study drugs for 100 days or for 180 days in the higher risk cohort of patients. No difference was noted in the primary endpoint (invasive fungal infection-free survival rate at 180 days) between the fluconazole and voriconazole prophylaxis arms (75% vs 78%, respectively), but a trend for reduced incidence of Aspergillus infections (17% vs 9%), reduced incidence of invasive fungal infections (11% vs 7%), and less frequent use of empirical antifungal treatment (30% vs 24%) was noted in the voriconazole arm, although the differences were not statistically significant. No differences in relapse-free and overall survival rates, nor incidence of severe adverse events were seen between treatment arms.26
In a multicenter randomized trial, prophylaxis with posaconazole in patients with neutropenia with acute myeloid leukemia or myelodysplastic syndromes receiving induction or reinduction chemotherapy significantly reduced the rate of invasive fungal infections during the treatment period (2% vs 8%; P<.001) and during the 100 days following randomization (5% vs 11%; P=.003). Posaconazole prophylaxis also reduced the incidence of invasive aspergillosis (1% vs 7%; P<.001) and was associated with a significant survival benefit (P=.04) compared with the fluconazole/itraconazole arm.27 Data from a prospective, randomized study showed that posaconazole was as effective as fluconazole in allogeneic HCT recipients with severe GVHD and reported reduced incidence of invasive aspergillosis and overall invasive fungal infections in patients receiving posaconazole compared with those receiving fluconazole.28 Posaconazole is equally effective compared with fluconazole as primary therapy for oropharyngeal candidiasis.29 Moreover, in a 2021 phase III randomized controlled trial, posaconazole was found to be noninferior to voriconazole as primary therapy for invasive aspergillosis, and participants had fewer treatment-related adverse events in the posaconazole group than in the voriconazole group.30
A 2020 systematic review and meta-analysis of 69 randomized clinical trials compared prophylaxis with various antifungal agents in patients with hematologic disease or who received HCT. Posaconazole prophylaxis significantly reduced invasive fungal infections (RR, 0.57; 95% CI, 0.42–0.79) and invasive aspergillosis (RR, 0.36; 95% CI, 0.15–0.85) compared with placebo. Additionally, voriconazole was associated with a significant reduction in invasive candidiasis (RR, 0.15; 95% CI, 0.09–0.26) compared with placebo. However, patients taking posaconazole had a higher incidence of withdrawal due to adverse effects.31
Isavuconazonium sulfate is a second-generation azole that was approved in March 2015 for the treatment of invasive aspergillosis and invasive mucormycosis. Isavuconazonium sulfate may be considered for antifungal prophylaxis when standard therapy is contraindicated, due to drug interactions or the risk of QT interval corrected for heart rate (QTc) prolongation.32
Toxicities and Drug-Drug Interactions of Azoles
Experience to date suggests that fluconazole and posaconazole are generally well tolerated and serious adverse events, primarily liver toxicity, are rare. Toxicities for voriconazole include neurologic and ophthalmic adverse events. In the context of renal dysfunction, accumulation of cyclodextrin vehicle may occur when the intravenous formulation is used. Data suggest that long-term use of voriconazole may be associated with severe photosensitivity and other adverse events including cutaneous malignancies, elevated serum fluoride levels, and periosteitis.33–37 Itraconazole may be associated with hepatic toxicity and gastrointestinal intolerance38 and is contraindicated in patients with a decreased cardiac ejection fraction or a history of congestive heart failure based on its negative inotropic properties. It can also increase cyclophosphamide metabolites, which in turn are associated with hyperbilirubinemia and nephrotoxicity during the early transplant period.39 Fluconazole, itraconazole, posaconazole, and voriconazole may cause QTc prolongation. Conversely, isavuconazonium sulfate has been associated with dose-dependent QTc shortening in healthy individuals.
Azole-associated drug–drug interactions are common clinical occurrences. Both the addition and withdrawal of azoles can result in either increased uptake of these other drugs or subtherapeutic exposure and potential transplant rejection or GVHD. Several studies demonstrate the interaction of azoles with hepatic enzymatic pathways. Administration of itraconazole with medications that are metabolized by the 3A4 isoenzyme can increase plasma concentrations causing QTc prolongation and ventricular tachyarrhythmias.40 These findings reinforce a note of caution about itraconazole (and by extension, fluconazole, voriconazole, isavuconazonium sulfate, and posaconazole), with regard to potential serious drug–drug interactions through inhibition of the cytochrome P450 3A4 isoenzyme. Additionally, fluconazole and voriconazole have demonstrated inhibition of CYP2C9 and CYP2C19 enzymes and high interpatient variability of genetic CYP2C19 polymorphisms that may also affect dosing.
The potential for QTc prolongation is a concern exacerbated by the combination of azoles and other drugs (eg, fluoroquinolones, macrolides, ondansetron) and with some chemotherapies (eg, nilotinib for chronic myeloid leukemia, panobinostat for myeloma). Itraconazole and posaconazole are also known inhibitors of gastric P-glycoprotein, which can increase systemic levels of drugs that are affected by this transport system. The list of drug–drug interactions is expansive and continues to grow. Although azoles may be necessary for antifungal therapy, they should only be incorporated into treatment after consultation with an infectious disease expert.
Therapeutic Drug Monitoring of Azoles
Therapeutic drug monitoring (TDM) for the pharmacokinetic evaluation of antifungal agents provides guidance for achieving adequate plasma drug concentration while reducing toxicity.
TDM should be considered for patients receiving triazoles; there is no current evidence to support the use of TDM for the evaluation of polyenes or echinocandins. Fluconazole and isavuconazonium sulfate are the 2 triazoles that do not require TDM. Fluconazole has linear pharmacokinetics that eliminate the need for TDM,41–45 though patients in renal failure should receive a modified dose. Studies intended to define a therapeutic range for isavuconazonium sulfate have not been performed; thus, TDM is not routinely recommended for isavuconazonium sulfate. TDM should be considered for posaconazole, voriconazole, and itraconazole. Variability of therapeutic drug levels may be affected by the route of drug administration, drug formulation, timing of monitoring, location of the infection, and intrinsic patient factors (ie, age, weight).
There are 3 formulations of posaconazole: oral suspension, delayed-release tablet, and intravenous solution. Pharmacokinetic studies with the oral suspension of posaconazole in healthy individuals showed that administration with or after a high-fat meal, or with any meal or nutritional supplement, greatly enhanced its absorption up to 400%.46 The plasma concentration of posaconazole can be reduced by proton pump inhibitors due to the increase in gastric pH when given orally.46 Subtherapeutic concentrations and breakthrough fungal infections have been reported.47,48 As reviewed by Brüggemann et al49 a substantial list of drug interactions with azole antifungal drugs can result in subtherapeutic effects or toxicity. The delayed-release (DR) tablet formulation of posaconazole was approved in 2013 and has improved absorption and more predictable bioavailability. Gastric pH does not affect plasma concentration of DR posaconazole,50 nor does it have the same interaction with proton pump inhibitors or metoclopramide.51 The intravenous formulation has also demonstrated similar pharmacokinetics and safety compared with the DR tablet.52 A target concentration of posaconazole for prophylactic TDM of >0.7 mcg/mL is supported by individual studies48,53,54 as well as 2 phase III studies;27,28 however, doses as low as 0.5 mcg/mL have also been reported as effective.48,54–58 TDM may not be necessary when using either the DR tablet or intravenous formulation in the prophylactic setting because data indicate that a dose of 300 mg/day results in at least 0.5 mcg/mL in >95% of patients. Treatment of an established infection is recommended to have a trough concentration >1 mcg/mL with potentially higher doses based on the pathogen minimal inhibitor concentrations and concerns for resistance.59,60
Studies of itraconazole demonstrate a significant rate of breakthrough infections when plasma drug concentrations are <0.5 mcg/mL;61,62 however, increased mortality was observed at plasma drug concentrations >5 mcg/mL.63,64 Targeting a lower itraconazole plasma concentration for prophylaxis and a higher dose if an active infection is being treated may be beneficial. Studies suggest that trough concentrations of itraconazole between 1 and 2 mcg/mL have shown the best therapeutic responses for invasive infections,65–68 while a trough concentration of >0.5 mcg/mL may be sufficient for prophylaxis. Currently, an upper limit of 17 mcg/mL measured by bioassay has been suggested,69 but studies for the upper limit have not been extensive. Itraconazole solution should be given either 1 hour before or 1 hour after meals based on the 43% increase in bioavailability in patients who fasted,70 while the capsule formulation should be given with a full meal. An alternative formulation, SUBA-itraconazole (for “super bioavailability”), addresses the absorption concerns of conventional itraconazole formulations with enhanced bioavailability that allows for lower dosing, while achieving similar serum concentrations. SUBA-itraconazole is FDA approved for the treatment of blastomycosis, histoplasmosis, and aspergillosis.71,72
Target voriconazole trough values between 0.5 and 4 mcg/mL have been proposed in clinical studies.73–80 Although 0.5 mcg/mL is a suggested target for prophylaxis, a higher range of 1 to 4 mcg/mL may be necessary for active disease and for patients with disease that has a poor prognosis. Higher concentrations may also benefit the patients who are immunocompromised by reducing breakthrough infection.81,82 Trough concentrations ≥4 mcg/mL have correlated with toxicity in various studies.73,77,80,83–87 Voriconazole bioavailability was lowered by about 22% when taken with food and by 34% when given with a high-fat meal.88 Therefore, voriconazole should be given either 1 hour before or 1 hour after meals.
Studies have shown a general consensus regarding a minimal level of plasma concentration necessary for the triazoles, though the lack of prospective studies has limited the adoption of formal monitoring standards. The Society of Infectious Diseases Pharmacists has published its guidelines for the use of TDM of antifungal agents based on available literature.89 Consideration of TDM is recommended by the NCCN panel in conjunction with involvement of an infectious disease expert.
Amphotericin B Formulations
Amphotericin B formulations are broad-spectrum antifungal agents that bind to ergosterol in the fungal cell membrane, leading to the formation of pores in the membrane and subsequent cell death. The original formulation, amphotericin B deoxycholate (AmB-D), was associated with dose-limiting toxicities including infusion-related reactions and nephrotoxicity. Three lipid-associated formulations, amphotericin B lipid complex, liposomal amphotericin B (l-AmB), and amphotericin B colloidal dispersion, have since been developed to have reduced toxicity. When available, amphotericin B lipid complex, and l-AmB are preferred over AmB-D.
Low-dose amphotericin B formulations have been studied in patients at high risk and have been shown to provide protection against invasive molds, although no survival benefit in randomized studies was seen when compared with fluconazole.38,90,91 Based on the toxicity of amphotericin B products and the availability of safer and equally effective alternative agents, amphotericin B products are considered a category 2B recommendation for prophylaxis. If an amphotericin B product is used, a lipid formulation is generally preferred because of less infusional and renal toxicity compared with conventional amphotericin B. Use of the lipid formulation is particularly important for patients at high risk for renal failure, such as patients with pre-existing renal disease, HCT recipients, and patients who are concurrently receiving other nephrotoxic agents.92,93
Aerosolized delivery of amphotericin B products has been considered for several years with the advantage of local delivery to the lungs while simultaneously avoiding systemic toxicity. A randomized, placebo-controlled trial found that aerosolized l-AmB was useful for preventing invasive pulmonary aspergillosis in patients with prolonged neutropenia.94 Limitations to the use of aerosolized amphotericin B for prophylaxis relate to the variability of this treatment due to different nebulizers and amphotericin B formulations, the lack of dosing optimization, and a dearth of direct comparative data with systemically administered mold-active azoles or echinocandins.95
Echinocandins
Echinocandins are a class of antifungal agents that disrupt the integrity of the fungal cell wall through noncompetitive inhibition of β-(1,3)-D-glucan synthase, an enzyme involved in the biosynthesis of β-(1,3)-D-glucan, which is a component specific to the cell wall of many fungi. Echinocandins have fungicidal activity against Candida species and are fungistatic toward Aspergillus species. Combination therapy with amphotericin B or triazoles has been proposed to improve activity against molds; however, clinical evidence for this remains quite limited. Advantages of this family of antifungals are the relatively low toxicity profiles and limited drug-drug interactions. Though echinocandins demonstrate activity against Candida species that are resistant to other antifungal agents,96 there is limited or no activity against dimorphic fungi. Four echinocandins are approved for use: caspofungin, micafungin, anidulafungin, and rezafungin. Caspofungin, micafungin, and anidulafungin are all approved for the treatment of esophageal candidiasis, while rezafungin is approved for the treatment of candidemia and invasive candidiasis in adults with limited or no alternative treatment options. Caspofungin and anidulafungin have additional indications for the treatment of candidemia and other infections caused by Candida species. Caspofungin is indicated for treatment of candidal pleural space infections, empirical treatment of fungal infections in neutropenia, and treatment of invasive aspergillosis in patients who are refractory to or intolerant of other antifungal agents. Micafungin has the additional indication for prophylaxis of candidal infections in patients receiving HCT.
Caspofungin was evaluated in a double-blind study including 128 patients with esophageal candidiasis.97 Patients received either caspofungin or AmB-D. Two doses of caspofungin were evaluated (50 mg or 70 mg intravenously once daily) with a greater response in the patients given the higher dose (96% vs 85%). Both groups treated with caspofungin had a better response than patients receiving amphotericin B (78%). At the 2-week follow-up, a greater percentage of patients remained negative for candidiasis with the caspofungin treatment (89% in the 70 mg group, 74% in the 50 mg group, and 63% in the amphotericin B group). Furthermore, drug-related adverse events were lower with caspofungin (7%, 4%, and 24%, respectively). Several studies have evaluated the role of caspofungin in the treatment of invasive aspergillosis in patients refractory to or intolerant of other antifungals, supporting its recommendation in this capacity.98,99
Micafungin is an echinocandin approved for prophylaxis against Candida infections in patients undergoing HCT. In a randomized, double-blind trial of autologous and allogeneic HCT recipients, the success rate with micafungin was superior to fluconazole (80% vs 73.5%; absolute difference +6.5%; 95% CI, 0.9%–12%; P=.03) based on prespecified criteria for treatment success (absence of suspected, proven, or probable invasive fungal infections during treatment period and absence of proven or probable infection during the 4-week period after treatment).100 The duration of this study encompassed the neutropenic period, but not the period after neutrophil recovery when GVHD would be expected to occur. The frequency of breakthrough candidemia was similar in both arms, but there was a trend to fewer episodes of invasive aspergillosis in allogeneic HCT recipients receiving micafungin. Survival and drug-related toxicity were similar between treatment arms.100 Micafungin has shown activity in the treatment of aspergillosis in patients refractory to or intolerant of other antifungal agents.101–103
Anidulafungin has been shown to be an effective antifungal agent against Candida infection in several studies. A randomized double-blind study in 601 patients with esophageal candidiasis demonstrated noninferiority of intravenous anidulafungin to oral fluconazole (97.2% vs 98.8%, respectively) and lower adverse effects (9.3% vs 12.0%) and recurring infections at the 2-week follow-up (64.4% vs 89.5%).104 In a smaller study of 19 patients with triazole-refractory mucosal candidiasis, anidulafungin treatment resolved infection in 18 of the patients.105 A larger phase III trial similarly showed superiority of anidulafungin compared with fluconazole in the treatment of candidemia and invasive candidiasis (75.6% vs 60.2%).106 The response at 2-week follow-up was 64.6% in the anidulafungin group versus 49.2% in the fluconazole group.
Rezafungin was evaluated in a phase III multicenter, double-blind randomized trial.107 In this trial, 199 patients with candidemia or invasive candidiasis were randomly assigned to either the rezafungin group (50%) or to the caspofungin group (50%). Rezafungin was found to be noninferior to caspofungin in the treatment of candidemia and invasive candidiasis (59% vs 61%, respectively) as well as for 30-day all-cause mortality (24% vs 21%, respectively).
NCCN Recommendations for Antifungal Prophylaxis
CYP3A4 inhibition by azoles can lead to toxicity when administered with several classes of drugs used in cancer therapy, including proteasome inhibitors, tyrosine kinase inhibitors, and vinca alkaloids.108 Thus, mold-active azoles should be stopped several days before the potential interacting drug is given. These azoles should also not be started until the other agent has been discontinued and sufficient time has elapsed for the drug to be eliminated. Due to variations in drug pharmacokinetics, firm recommendations regarding a minimum time from drug discontinuation to azole administration cannot be made, though some institutions consider waiting at least 10 days following administration of these classes of drugs. Use of echinocandin prophylaxis may be considered in the place of azoles. Consultation with pharmacology and infectious disease experts is recommended.
The NCCN Guidelines panel recommends posaconazole (category 1) for antifungal prophylaxis in neutropenia with acute myeloid leukemia and myelodysplastic syndromes receiving induction or reinduction chemotherapy (Figure 4).8 The role of antifungal prophylaxis in patients with acute leukemia receiving consolidation chemotherapy has not been adequately evaluated. Voriconazole, isavuconazole, fluconazole, echinocandin, or amphotericin B products are all category 2B recommendations in this disease setting. Antifungal prophylaxis should be continued until resolution of neutropenia.
In patients with mucositis receiving autologous HCT, antifungal prophylaxis with fluconazole or echinocandin (both category 1) is recommended until resolution of neutropenia. No prophylaxis is recommended in autologous HCT recipients without mucositis.
The NCCN panel recognizes that strong evidence exists for the use of fluconazole or echinocandin as prophylaxis in neutropenic allogeneic HCT recipients (category 1).8 However, it should be noted that fluconazole use can predispose patients to colonization and bloodstream infection by fluconazole-resistant Candida strains.109,110 Posaconazole as prophylaxis has not been evaluated during the neutropenic period after conditioning in allogeneic HCT recipients; thus, the safety of this approach is unknown. Drug–drug interactions during conditioning for HCT, specifically with posaconazole, itraconazole, or voriconazole, complicate treatment of fungal infections in these patients. Prophylaxis may need to be tailored after consultation with an infectious disease expert. Posaconazole, isavuconazole, voriconazole, echinocandins, and amphotericin B products are all considered category 2B recommendations. Antifungal prophylaxis should be considered until at least day 75 or count recovery (ANC >500) after allogeneic HCT.8,111–113
Patients with hematologic malignancies are at high risk of developing invasive fungal disease. The German Society of Hematology and Medical Oncology strongly recommends administration of antifungal prophylaxis in patients with hematologic malignancies with prolonged neutropenia (ie, <500 cells/mcL for >7 days) with posaconazole as the drug of choice for mold-active prophylaxis.113
Although many centers reasonably use antifungal prophylaxis in non-neutropenic allogeneic HCT recipients with GVHD, this practice was only evaluated in a single, properly designed study. In the prospective, randomized, double-blind study, posaconazole was compared with fluconazole as prophylaxis in allogeneic HCT recipients with severe GVHD requiring intensive IST.28 Inclusion criteria included grade 2 to 4 GVHD, chronic extensive GVHD, or intensive IST consisting of either high-dose corticosteroids, antithymocyte globulin, or a combination of two or more immunosuppressive agents or types of treatment. Prophylaxis with posaconazole resulted in reduced incidences of invasive aspergillosis, total invasive fungal infections while on treatment, and deaths attributed to fungal infection.28 Posaconazole is recommended (category 1) as prophylaxis in patients with GVHD receiving intensive IST, as defined by the inclusion criteria in this trial, although the benefit/risk ratio of mold-active prophylaxis in patients receiving less intensive IST has not been established.
Patients with chronic severe neutropenia (ANC <500 neutrophils/mcL) due to an underlying disease (such as aplastic anemia) are at substantial risk for invasive aspergillosis.114 Although this population has not been evaluated in clinical trials of antifungal prophylaxis, some panel members advise the use of a prophylactic mold-active agent (eg, posaconazole or voriconazole).
Secondary antifungal prophylaxis is defined as administration of antifungal therapy in a patient with a prior fungal infection to prevent recrudescence. The panel recommends secondary prophylaxis with an appropriate antifungal agent in patients with prior chronic disseminated candidiasis115 or with invasive filamentous fungal infection116 during subsequent cycles of chemotherapy or HCT. In patients with invasive aspergillosis before HCT, antifungal therapy for >1 month and resolution of radiologic abnormalities correlate with a lower likelihood of posttransplant recurrence of infection.117 Secondary prophylaxis with a mold-active agent is advised for the entire period of immunosuppression.
Antiviral Prophylaxis and Pre-emptive Antiviral Therapy
Herpes Simplex Virus
HSV is an important pathogen in patients who develop neutropenia and mucositis. HSV infections primarily result from reactivation of latent virus. The presence of latent HSV can be determined by pretreatment HSV serology. Reactivation and infection with HSV occur in 60%–80% of HCT recipients and patients (without prophylaxis) with acute leukemia undergoing induction or reinduction therapy who are seropositive for HSV.118–120 Among allogeneic HCT recipients, HSV disease is most likely to occur within the first month posttransplant, but may occur in later stages during intense immunosuppression.121,122 Although disseminated HSV infection is uncommon, infection from viral reactivation is frequently associated with increased mucosal damage, resulting in increased pain, limited ability to maintain oral hydration and nutrition, and an increased risk of bacterial and fungal superinfections.
NCCN Recommendations for HSV Prophylaxis
Antiviral prophylaxis against HSV is advised during the period of neutropenia in patients who are HSV-seropositive who are receiving chemotherapy (induction or consolidation) for acute leukemia, and during neutropenia and possibly longer in allogeneic and autologous HCT recipients depending on the degree of immunosuppression (Figure 5). A longer period of prophylaxis should be considered in allogeneic HCT recipients with GVHD or with frequent HSV reactivations before transplantation.123 Acyclovir, valacyclovir, or famciclovir are the initial agents of choice for HSV prophylaxis.8,124 Foscarnet is typically reserved for patients with acyclovir-resistant HSV infection.8,124 In patients receiving antiviral prophylaxis with ganciclovir or foscarnet for prevention of CMV reactivation, additional prophylaxis with acyclovir is not necessary given that these agents are active against HSV.124 However, it is required in patients receiving CMV prophylaxis with letermovir as this agent lacks activity against HSV.
HSV and herpes zoster infections are common in patients with chronic lymphocytic leukemia treated with the CD52 monoclonal antibody alemtuzumab. For these patients, antiviral prophylaxis is advised until at least 2 months after completion of alemtuzumab therapy and until CD4+ cell counts are ≥200 cells/mcL.125
Prophylaxis against HSV should be considered in other patients at intermediate risk for HSV reactivation, including those with hematologic malignancies with prolonged neutropenia or those receiving high-dose corticosteroids or T-cell–depleting agents (eg, fludarabine). Once a patient has had HSV reactivation requiring treatment, the panel recommends HSV prophylaxis for that patient during all future episodes of neutropenia induced by cytotoxic therapy. HSV prophylaxis is also indicated in children who are seropositive for the virus.
Varicella Zoster Virus
Impaired cellular immunity is the principal risk factor for VZV disease. In allogeneic HCT recipients with a history of VZV infection, about 30% have reactivation of VZV disease without antiviral prophylaxis.126 In patients with a history of chicken pox, oral acyclovir administered from 1 to 2 months until 1 year after allogeneic HCT significantly decreased the incidence of VZV disease compared with placebo (5% vs 26%, respectively).126 The frequency of VZV disease in the postprophylactic period was similar between the groups and predominantly occurred in patients who required systemic immunosuppression. This prolonged course of acyclovir prophylaxis is likely to also prevent HSV reactivations. Subsequent studies have consistently demonstrated the benefit of long-term antiviral prophylaxis against VZV disease in recipients of allogeneic HCT. Patients who received anti-VZV prophylaxis with acyclovir or valacyclovir for 1 year post-HCT had significantly reduced VZV disease compared with those who did not receive long-term prophylaxis (9% vs 25%; P<.001); no evidence of rebound VZV disease was observed.127 Long-term (1 year postallogeneic HCT) prophylaxis with lower doses of acyclovir or valacyclovir was associated with a higher cumulative incidence of VZV reactivation.128,129
NCCN Recommendations for VZV Prophylaxis
The NCCN Guidelines panel recommends prophylaxis against VZV for at least 1 year after allogeneic HCT in patients seropositive for VZV pretransplant (Figure 5) and recommends considering the extension of prophylaxis in patients who continue to receive systemic IST. Although higher doses are necessary, the same agents used as HSV prophylaxis are also active against VZV. For pediatric patients, prophylaxis for VZV should not be routinely given unless there is a history of recurrent zoster infections or incidence of first zoster infection while on myelosuppressive therapy, even if they are seropositive or vaccinated.
Among recipients of autologous HCT, HSV reactivation is more likely to occur in the early neutropenic phase, whereas the risk of VZV reactivation extends through the first year.130 Thus, VZV prophylaxis for at least 6 to 12 months posttransplant should be considered in autologous HCT recipients. Prophylaxis against VZV should be considered in other patients at intermediate risk for viral reactivation, including patients with hematologic malignancies with prolonged neutropenia or those receiving T-cell–depleting agents (eg, fludarabine, alemtuzumab). Bortezomib is associated with an increased risk of VZV reactivation during active therapy131–134; carfilzomib may also be associated with VZV reactivation. Prophylaxis with acyclovir, valacyclovir, or famciclovir should be protective and can be considered in these settings.135,136 As previously discussed, among patients with chronic lymphocytic leukemia receiving alemtuzumab treatment, antiviral prophylaxis is recommended until 2 months after completion of treatment and until the CD4+ cell counts reach 200 cells/mcL or more.125
Cytomegalovirus
CMV infections most frequently occur in patients with cancer who undergo allogeneic HCT or who receive alemtuzumab therapy or other T-cell depleting therapies. CMV is a common cause of opportunistic infections during the early postengraftment phase following allogeneic HCT, but can also occur in the late postengraftment phase (particularly for patients with GVHD during the latter phase).121,122 Infection can result from viral reactivation (in patients who are immunocompromised CMV-seropositive) or primary infection (in CMV-seronegative). The risk for CMV reactivation and disease is highest among HCT recipients with CMV-seropositive status prior to transplant.137 Among patients who are CMV-seropositive undergoing allogeneic HCT (with graft sources from peripheral blood, bone marrow, or umbilical cord blood), the incidence of CMV reactivation ranged from 50% to 60% (with CMV disease in about 10%–30% of seropositive recipients) even with routine surveillance and antiviral prophylaxis or pre-emptive therapy.137–140
In a randomized phase II prophylaxis trial, oral letermovir was administered to allogeneic HCT recipients with CMV-seropositive status daily at 60 mg, 120 mg, or 240 mg for 12 weeks posttransplant. The incidence of prophylaxis failure (defined as discontinuation of the study drug because of CMV detection, end-organ disease, or any other cause) was significantly lower in the letermovir groups than in the placebo group (32% for the 120 mg group, 29% for the 240 mg group vs 64%). Moreover, letermovir was well tolerated, with no indication of hematologic toxicity or nephrotoxicity.141 In a randomized, double-blind, phase III trial, prophylaxis with daily oral or intravenous dose of letermovir, 480 mg/day (or 240 mg/day in patients taking ciclosporin), was administered to recipients of allogeneic HCT for 14 weeks after transplantation. Fewer patients developed clinically significant CMV infection in the letermovir group than in the placebo group (38% vs 61% in placebo; P<.001). The frequency of adverse events, like vomiting and edema, were mildly higher in the letermovir group.142 Despite the lower toxicity profile of letermovir compared with other CMV-targeted drugs, rapid emergence of resistant mutants under letermovir has been described in cases where treatment is interrupted or underdosed,143 or in patients with other risk factors.144,145
Valganciclovir and ganciclovir are the agents of choice for first-line pre-emptive therapy; foscarnet is more commonly used for patients who cannot tolerate ganciclovir or for second-line pre-emptive therapy.124 Foscarnet and ganciclovir had similar efficacy as pre-emptive CMV therapies in recipients of allogeneic HCT, but ganciclovir was associated with a higher rate of early discontinuation because of neutropenia or thrombocytopenia.146 Although ganciclovir had a higher rate of early discontinuation, there remains a paucity of data to recommend foscarnet as first-line treatment of CMV.147–149
Pharmacokinetic studies have demonstrated the feasibility and safety of using oral valganciclovir, a prodrug of ganciclovir, in place of ganciclovir in patients who underwent allogeneic HCT.150,151 Oral valganciclovir used as pre-emptive anti-CMV therapy was shown to have acceptable oral bioavailability and was safe and effective in controlling CMV infection in allogeneic HCT recipients, including patients with grades 1 and 2 gastrointestinal GVHD.150,152–154 Thus, valganciclovir is a highly acceptable oral option for pre-emptive therapy for CMV in the absence of substantial gastrointestinal GVHD. Reports of higher rates of CMV disease with oral valganciclovir compared with intravenous ganciclovir in patients with hepatic dysfunction restricted approval for solid tumor transplant patients by specifically excluding liver transplant patients.155,156 It is postulated that hepatic dysfunction allows bioabsorption of valganciclovir but decreases cleavage of the valine ester, thereby limiting conversion to the active form.156
Cidofovir has been evaluated as both primary and secondary pre-emptive therapy in recipients of allogeneic HCT.157–160 In a retrospective study of recipients of allogeneic HCT (n=82) treated for CMV disease (n=20), primary pre-emptive therapy (n=24) or secondary pre-emptive therapy (n=38) with cidofovir demonstrated an observed response in 50% of patients treated for CMV disease (mainly CMV pneumonia) and in 62% of patients treated with primary pre-emptive therapy.159 Moreover, secondary pre-emptive therapy with cidofovir resulted in a response rate of 66% in patients where disease progressed or relapse occurred (defined as continued presence or recurrence of pp65 antigenemia or viral DNA after at least 1 week of antivirals) following initial pre-emptive therapy with ganciclovir, foscarnet, or a combination of these agents.159
For CMV infections that are refractory or resistant to available antivirals (valganciclovir, ganciclovir, foscarnet, cidofovir), maribavir has shown promise in some studies. In a phase II, randomized, double-blind trial, ≥400 mg of maribavir twice daily was found to be active against refractory or resistant CMV infections in transplant recipients, with a higher proportion of patients experiencing undetectable CMV DNA within 6 weeks of treatment compared with placebo.161 Dysgeusia was the most common treatment-related adverse event. In another phase III randomized trial, maribavir was superior to other anti-CMV agents in HCT and solid organ transplant recipients, with 56% of patients experiencing CMV viremia clearance under maribavir compared with 24% under other agents (P<.001). Maribavir was also associated with less acute kidney injury versus foscarnet (8.5% vs 21.3%) and with less neutropenia versus valganciclovir/ganciclovir (9.4% vs 33.9%).162 On the other hand, in another phase III randomized double-blind trial, noninferiority of maribavir to valganciclovir for the primary endpoint of confirmed CMV viremia clearance was not achieved based on the prespecified noninferiority margin (69.6% vs 77.4%).163 However, maribavir demonstrated comparable CMV viremia clearance to valganciclovir during posttreatment follow-up (52.7% vs 48.5%), with fewer discontinuations due to neutropenia.
Late CMV disease, defined as occurring after day 100 of HCT, remains a persistent problem in the era of CMV prophylaxis and pre-emptive therapy. CMV infections are also common among patients with lymphoproliferative malignancies, patients receiving T-cell suppressive therapy with purine analogs, and patients receiving alemtuzumab therapy, and occurs most frequently between 3 to 6 weeks after initiation of therapy when T-cell counts reach a nadir.164–169 Several studies of alemtuzumab in patients with chronic lymphocytic leukemia have demonstrated the effectiveness of using routine CMV monitoring coupled with pre-emptive anti-CMV therapy with ganciclovir in preventing overt CMV disease.164–166,170 A small randomized study in patients with lymphoproliferative disease treated with alemtuzumab-containing regimens (n=40) showed that upfront CMV prophylaxis with oral valganciclovir significantly reduced the incidence of CMV reactivation compared with oral valacyclovir (0% vs 35%; P=.004).167
NCCN Recommendations for CMV Prophylaxis
Based on the available data that predict the risk of CMV disease, the NCCN Guidelines panel recommends routine surveillance for CMV reactivation after allogeneic HCT, consisting of weekly monitoring by PCR, especially during alemtuzumab therapy and at least 2 months after completion of treatment.171 On confirmation of CMV viremia (defined as PCR positivity for CMV in ≥2 consecutive samples obtained 1 week apart), the panel recommends pre-emptive therapy with oral valganciclovir, intravenous ganciclovir or intravenous foscarnet for at least 2 weeks and until CMV is no longer detectable (Figure 6).
In cases of ganciclovir-resistant CMV or when ganciclovir is not tolerated (eg, ganciclovir-induced myelosuppression), intravenous foscarnet or intravenous cidofovir may be used. Additionally, in cases of posttransplant CMV infection that is refractory to ganciclovir/valganciclovir, foscarnet, or cidofovir, oral maribavir may be used. However, infectious disease consult is highly recommended due to the possibility of drug resistance or interaction. Primary prophylaxis with oral or intravenous letermovir may be considered for CMV-seropositive recipients who undergo allogeneic HCT, with a dose reduction recommended if coadministered with cyclosporine due to drug interactions. In patients receiving allogeneic HCT, the strategy of CMV surveillance testing by PCR followed by pre-emptive anti-CMV therapy for a positive result is favored over universal long-term prophylaxis. Some centers consider the use of letermovir through day 100 post-HCT and continue CMV surveillance for patients at high risk for CMV reactivation. In certain circumstances, up to day 200 can be considered.172 Letermovir lacks HSV and VZV coverage and HSV/VZV prophylaxis should be continued.
Surveillance should typically occur for at least 3 to 6 months posttransplant and during chronic GVHD requiring IST. Higher risk transplant subgroups may exist and require different management strategies. Note that the CD4+ count will be reduced by systemic corticosteroids and by other lymphocyte-depleting agents. The majority of cases of late CMV disease occur within the first year of transplant and <5% occur after the second year.173 Therefore, the value of CMV surveillance beyond 2 years after HCT is unknown but can be considered in patients with significant chronic GVHD. There is debate about how to treat patients after a negative test for CMV. There are not enough data to determine whether patients should be transitioned to surveillance or continue with chronic maintenance therapy, and if so, for how long. The benefits must be weighed against the potential toxicity associated with long-term antiviral use. Ganciclovir and valganciclovir are associated with bone marrow suppression that may increase the risk of common opportunistic infections. Foscarnet can cause nephrotoxicity and electrolyte abnormalities but is tolerated.146,174 Cidofovir can be associated with substantial nephrotoxicity; although less frequent, ocular toxicity has been reported.159,160 Acyclovir and valacyclovir have excellent safety profiles but are only weakly active against CMV and are not recommended as prophylaxis or treatment of CMV infection.
For the prevention and treatment of CMV pneumonitis, adjunctive intravenous immunoglobulin (IVIG) can be administered; however, IVIG is generally not recommended for prophylactic use except in limited situations due to cost and the limited evidence of activity of this treatment. Although no optimal dosing regimen has been determined, IVIG is commonly administered every other day for 3 to 5 doses. CMV-specific IVIG has not been shown to be any more efficacious than standard IVIG.
Hepatitis B Virus
The risk factors for HBV infection include personal or parental history of an intermediate to high prevalence of HBV infection in one’s birthplace (defined as a prevalence of hepatitis B surface antigen [HBsAg] positivity in >2% of the population); household and sexual contact with HBsAg+ persons; individuals with multiple sexual partners or history of sexually transmitted diseases; individuals who have been inmates of correctional facilities; patients with chronically elevated aspartate aminotransferase or alanine aminotransferase levels; patients with a history of injection drug use; males who have sex with other males; and patients positive for HCV or HIV.
A positive HBsAg is associated with active infection or a window period before the development of protective immunity in patients exposed to HBV. An individual who has been vaccinated for HBV typically has the following serology: negative HBsAg, positive hepatitis B surface antibody (HBsAb), and negative hepatitis B core antibody (HBcAb).175 False-negative HBsAg results may occur in patients with chronic liver disease.176 HBsAb positivity is generally equated with protective immunity, although reactivated HBV disease may occur in the setting of significant immunosuppression in individuals who are HBcAb-positive.177 A patient with resolved hepatitis B infection will be HBcAb positive but HBsAg negative. As mentioned above, some patients with cancer are at increased risk for HBV reactivation due to profound immunosuppression stemming from cytotoxic regimens, high-dose corticosteroids, tyrosine kinase inhibitors, anti-CD20/CD52 monoclonal antibodies, and/or the underlying malignancy (eg, leukemia, lymphoma).
Patients with malignancies who are HBsAg positive and/or HBcAb positive are at risk for HBV reactivation with cytotoxic chemotherapy. Approximately 20%–50% of patients with HBsAg positivity and 3%–45% with HBcAb positivity develop HBV reactivation.175,178–187 The risk of HBV reactivation for patients who are HBsAg negative, HBcAb positive varies widely based on the virological profile, disease, and immunosuppressive regimen. Serum HBV DNA testing before the start of therapy may help define their risk of reactivation. If viremic, they may receive similar prophylaxis as patients who are HBsAg positive.188 Complications of HBV reactivation can range from self-limited hepatitis to fulminant hepatic failure and death.187,189–193 HBV reactivation can lead to early discontinuation or delayed initiation of treatment.194,195 A systematic review and meta-analysis by Zhang et al.196 revealed that patients with B-cell non-Hodgkin lymphoma (NHL) who are HBsAg+ had worse prognosis and higher incidence of hepatic dysfunction during chemotherapy. In a meta-analysis and evaluation of the United States FDA safety reports, it was reported that HBcAb positivity correlated with increased incidence of rituximab-associated HBV reactivation.179 After allogeneic HCT, loss of HBV-specific immunity may occur (ie, loss of HBsAb and development of HBsAg and HBV PCR positivity). This has been observed in up to 40% of susceptible individuals in one report197 and may be confused with hepatic GVHD. A retrospective study showed that recipients of allogeneic HCT who were HBsAg negative but HBcAb positive had a high risk of seroconversion to HBsAg positivity and HBV reactivation (subsequently leading to hepatitis) following allogeneic HCT.198
There are several nucleos(t)ide analogs approved by the FDA for the prevention and treatment of HBV. Historically, data supporting the use of these analogs have been based on lamivudine, a reverse transcription inhibitor. In a meta-analysis of clinical trials evaluating lamivudine prophylaxis in patients with HBsAg-positive lymphoma treated with IST, prophylaxis resulted in a significant reduction in HBV reactivation (risk ratio, 0.21; 95% CI, 0.13–0.35) and a trend for reduced HBV-related deaths (risk ratio, 0.68; 95% CI, 0.19–2.49) compared with no prophylaxis.199 However, despite its initial effectiveness, virologic breakthrough was high, with reports of resistance in 80% of patients after 5 years of therapy.200 Thus, lamivudine monotherapy has fallen out of favor. Studies suggest one of the newer agents (such as entecavir or tenofovir) may be preferable or combination therapy may have a possible role for patients with lamivudine-resistant HBV infections.201–203
As of 2015, tenofovir is available in 2 different prodrug forms, tenofovir disoproxil fumarate (TDF) and tenofovir alafenamide (AF). Tenofovir AF has greater plasma stability than TDF, allowing use of a lower dose and lesser systemic exposure to the drug.204 TDF has demonstrated superior antiviral efficacy compared with adefovir in a phase III randomized double-blind study in patients with chronic HBV infection, making tenofovir preferred over adefovir in this setting.205 Two randomized, phase III, double-blind studies comparing tenofovir AF to TDF in patients with HBeAg-negative206 or HBeAg-positive207 chronic HBV infection showed that the efficacy of tenofovir AF was noninferior to TDF, with better bone and renal safety for tenofovir AF. Although these data support the use of tenofovir for HBV infection, limited data are available regarding its use in patient populations with cancer. A systematic review and meta-analysis showed that in patients with HBV-associated hepatocellular carcinoma, tenofovir was associated with better overall survival and reduced late recurrence compared with entecavir.208 No detectable resistance to TDF was reported in patients with chronic hepatitis B after 6 years of treatment.209 In another study, sequencing of the HBV polymerase/reverse transcription indicated sequence changes at polymorphic sites, though none resulted in drug resistance.210 In total, there were only 16 cases of virologic breakthrough, 12 of which were associated with nonadherence to study medication. Resistance for TDF remained undetectable throughout a 5-year span. By comparison, lamivudine resistance was calculated to be 24% in the first year, and this number steeply climbed to 70% by year 5.210
Entecavir has shown improved antiviral activity compared with adefovir in randomized open-label studies in patients with chronic hepatitis B.211 A few small case studies have evaluated entecavir in the prevention212 or treatment of HBV in patients with cancer (reviewed by Liu et al.213). Entecavir had a low drug resistance of 1.2% at 5 years214 compared with adefovir, which had an intermediate resistance that increased from 0% in the first year to 29% by year 5.205,215,216
In addition to drug resistance, the safety profile of the nucleos(t)ide analogs should affect drug selection. Nephrotoxicity has been seen with adefovir217,218 and tenofovir (specifically TDF).219 No significant side effects have been reported with lamivudine or entecavir; however, it is recommended that all patients be monitored for lactic acidosis and severe hepatomegaly with steatosis.
NCCN Recommendations for HBV Prophylaxis
Risk-based screening is recommended by ASCO220 and the American Association for the Study of Liver Disease (AASLD).221 Although it is possible that risk-based screening may be more cost-effective than universal screening, there are currently no validated risk tools that could easily be implemented into clinical practice. Furthermore, <60% of patients with HBV infection may have obvious risk factors,222 and only 10%–35% of these patients may be aware of their own HBV infection.223,224 Therefore, any patient expected to receive IST or chemotherapy should be screened. Implementation of universal screening, as recommended by the CDC, should be considered.225
In patients undergoing intensive IST, including HCT, both patient and donor should be screened for HBV, HCV, and HIV prior to treatment. Evaluation of HBsAg, HBcAb, and HBsAb should be considered at baseline.124,175 Vaccination against HBV should be strongly considered in patients who are HBV-naïve (ie, negative for HBsAg, HBsAb, and HBcAb) (see later section on “Vaccination”).124,175 In patients who are HBV-naïve undergoing allogeneic HCT, grafts from donors who are HBsAg-positive or HBV DNA-positive should be avoided wherever possible. Donors who have not been exposed to HBV should be considered for HBV vaccination before hematopoietic cell collection.
In individuals who are HBsAg-positive or HBcAb-positive, baseline quantitative PCR for HBV DNA should be obtained. In allogeneic HCT candidates with evidence of active HBV infection (chronic hepatitis based on biopsy or positive HBsAg or high levels of HBV DNA), transplant procedure should be delayed when possible, and antiviral therapy should be given for 3 to 6 months prior to conditioning.124 In HCT candidates who are HBsAg-positive or HBcAb-positive but without evidence of active HBV replication, antiviral prophylaxis should be considered (starting shortly before the transplant procedure). All allogeneic HCT recipients should continue surveillance for at least 12 months after transplant or during GVHD (Figure 7). For details on the management of HBV infection in patients with B-cell NHL, see the NCCN Guidelines for B-Cell Lymphomas (available at NCCN.org)
The optimal choice of antiviral agents for prophylaxis (or pre-emptive approaches) will primarily be driven by institutional standards. The NCCN panel recommends consultation with an expert in hepatitis treatment to determine appropriate antiviral prophylaxis for patients who test positive for HBV. Preferred agents for HBV prophylaxis are entecavir and tenofovir. Although data were originally obtained with lamivudine, entecavir and tenofovir are preferred, especially when treating patients with active HBV infections due to the low threshold of resistance with lamivudine. Monitoring of HBsAg and HBV DNA and transaminases should be considered for patients without active HBV infection who are not receiving prophylaxis.
Hepatitis C Virus
Studies for HCV reactivation in patients with cancer are not as expansive as studies for hepatitis B; however, an increase in mortality was reported in patients with cancer who had HCV infection compared with patients with cancer who were HCV negative.226 A review by Yazici et al227 summarized studies of HCV reactivation in patients receiving targeted therapies, and the data correlated an increase in HCV reactivation with these therapies.227 Differences in outcomes between patients who are HCV positive with cancer versus HCV positive without cancer were reported to include higher occurrence of occult infection, higher risk of developing early cirrhosis, higher rate of fibrosis progression, development of viral reactivation, and poorer virologic outcomes (reviewed by Borchardt et al).228 The guidelines from the joint IDSA and AASLD panels for the testing, management, and treatment of hepatitis C recommend that treatment of HCV be considered for patients with chronic HCV with a life expectancy of >12 months.229
NCCN Recommendations for HCV Screening and Management
All patients who are expected to receive chemotherapy or IST should be screened for HCV. The data are limited regarding the treatment of HCV in patients with cancer, but it is generally not recommended that HCV treatment and cancer therapy be given concurrently.228 The IDSA/AASLD guidelines can provide additional guidance for antiviral therapy, but an infectious diseases consult is necessary to evaluate the use of concomitant or sequential anti-HCV and cancer therapy.229 Monitoring of ALT levels and HCV viral load monthly, or as clinically indicated, should be initiated as part of surveillance (Figure 7). For details on the management of HCV infection in patients with HCV-associated lymphomas, see the NCCN Guidelines for B-Cell Lymphomas (available at NCCN.org).
Human Immunodeficiency Virus
The CDC surveillance report estimates that 1.1 million persons are living with HIV in the United States. This includes the estimated 166,000 persons whose infection has not yet been diagnosed.230 There is support for HIV testing in all patients treated for cancer.231 Patients who are HIV-positive and have cancer are classified as having either AIDS-defining cancer (ADC) or non–AIDS-defining cancer (NADC). ADC includes Kaposi sarcoma, NHL, and cervical cancer. There is a higher incidence of these cancers in those who are HIV-positive than in those who are HIV-negative.232
The incidence of NADC is increasing, likely due to the longer life expectancy of patients with HIV resulting from the advancement of treatment options.233 Patients with HIV and NADC were shown to have an overall worse cancer outcome when compared with patients who are HIV-negative with the same cancer.234 However, improvement in outcome was seen when patients with HIV received highly active antiretroviral therapy.235 There should be caution regarding the concomitant administration of select antiretroviral therapies (including the protease inhibitors and non-nucleoside reverse transcription inhibitors) with cancer therapy as adverse events through cytochrome P450 3A4 have been documented.236 A publication from MD Anderson Cancer Center retrospectively evaluated the use of HIV screening in patients before systemic cancer therapy.237 Of the 18,874 patients in this study, 3,514 patients tested positive for HIV at the initiation of systemic cancer therapy. Patient histories indicated a higher incidence in patients with sexually transmitted disease (37.7% vs 18.5%; P<.001) or a history of illegal drug use (46.2% vs 18.6%; P<.001). Patients screened for HIV included 12.1% of patients with NADC and 9.4% of patients with cervical cancer. Interestingly, a significantly higher percentage (88.4%) of patients with NHL were screened for HIV, which may be partially attributed to clinician education of the role of HIV in these patients.237
NCCN Recommendations for HIV Screening
In 2006, the CDC published recommendations for routine HIV testing in all patients (13–64 years of age) in the health care setting.238 The testing is intended to be voluntary and conducted only with consent from patients. Under these guidelines, patients are informed either verbally or in written format that HIV testing will be conducted unless the patient declines testing (opt-out screening). The CDC recommends that patients at high risk for HIV infection be screened at least annually.238 The implementation of these guidelines is largely dependent on institutional practices and the prevalence of undiagnosed HIV infections in specific institutions. However, the NCCN panel strongly encourages concordance with the CDC recommendations.
In addition to the CDC recommendations, the NCCN panel emphasizes that all patients receiving chemotherapy or IST be screened for HIV.231 Patients coinfected with hepatitis pose an additional complication. Select antiretroviral therapies including the integrase strand inhibitors and nucleoside/nucleotide reverse transcription inhibitors have demonstrated fewer drug–drug interactions compared with the protease inhibitors and non-nucleoside reverse transcription inhibitors. However, consultation with an infectious disease expert is necessary for treatment of HIV in patients with cancer as therapies continuously evolve. HIV viral load should be monitored monthly during therapy and then as clinically indicated (Figure 7; see also NCCN Guidelines for Cancer in People with HIV, available at NCCN.org).
Screening for Other Viruses
Rapid PCR panels should be considered for detection of respiratory viruses including respiratory syncytial virus (RSV), influenza, parainfluenza virus, adenovirus, rhinovirus, and metapneumovirus in patients with cough and/or shortness of breath that might indicate a viral infection. Ribavirin and IVIG have been proposed as antiviral therapies239–243; however, data are not sufficient to provide recommendations.
RSV is a major cause of severe infection in patients who are immunocompromised, with mortality rates ≤80% in HCT recipients.244,245 Progression of RSV to the lower respiratory tract occurs in ≤50% of patients receiving HCT or chemotherapy.246–248 The virulent nature of RSV requires hospitalization for treatment. Treatment options are limited to ribavirin and adjunctive IVIG. There is a diversity of practice among the institutions for the treatment of RSV disease. Based on limited data249–251 and strong panel disagreement regarding the use of ribavirin and the best method of delivery, ribavirin has been designated a category 3 recommendation. Recommendations for inhaled versus oral ribavirin should be based on the individual institution.
Rapid screening tests are available for detection of influenza. Clinical benefit is highest when treatment is initiated within the first 48 hours of influenza symptoms, although benefits can still be seen when initiated after the 48-hour window.252 During the influenza season, consider empirical antiviral therapy for patients within 48 hours after symptoms develop that are suggestive of influenza (eg, high fever, coryza, myalgia, dry cough), especially during community outbreaks. Both the IDSA (2007) and CDC guidelines (2011) recommend antiviral treatment with the neuraminidase inhibitors oseltamivir or zanamivir, which are active against both influenza A and B viruses.253,254 Both agents are approved by the FDA for the treatment of influenza within 48 hours of symptomatic onset; the indicated duration of treatment is 5 days. However, longer courses of treatment (eg, 10 days) and treatment until resolution of symptoms can be considered in those who are immunocompromised, though this is controversial. Some centers have used higher doses (eg, 150 mg twice a day) of oseltamivir in these patients with mixed results. Pandemic influenza does not have a predictable seasonal pattern and may spread in the community concurrently with a seasonal influenza strain. Antiviral susceptibility of influenza strains is variable and cannot be predicted based on previous influenza outbreaks. In cases of seasonal influenza and pandemic strains, it is necessary to be familiar with susceptibility patterns and guidelines on appropriate antiviral treatment.255 There are limited data on the activity of peramivir; with comparable clinical outcomes observed in comparison with oseltamivir.256,257 Peramivir, available as an intravenous injection, can be considered for patients who cannot absorb oral oseltamivir or tolerate oseltamivir or inhaled zanamivir.
A few small case studies showed limited efficacy of baloxavir, a polymerase inhibitor active against both influenza A and B viruses, in immunocompromised patients with oseltamivir- or peramivir-resistant influenza.258–260 However, due to limited data and emergence of resistant strains,260,261 this option is not routinely recommended by the CDC.
BK virus is a common polyomavirus that remains dormant in the kidney and urinary tract. In immunosuppressed individuals, BK virus can reactivate. Patients undergoing allogeneic HCT are particularly vulnerable to BK virus, and the development of hemorrhagic cystitis and additional complications such as ureteral stenosis.262,263 Supportive care remains the mainstay of management. While cidofovir demonstrates effectiveness as a treatment option for BK virus, renal toxicity is a significant complication.264 There is currently a lack of data to support recommendations on the treatment of BK virus. Expert consensus on management has been published for pediatric HCT recipients.265
COVID-19 screening, prophylaxis, and treatment is a complex and rapidly evolving area. Please see the CDC guidelines for latest recommendations.
Prophylaxis for Pneumocystis jirovecii
TMP/SMX prophylaxis for Pneumocystis jirovecii is highly effective in preventing PJP.266–269 In a systematic review and meta-analysis of 12 randomized studies (n=1,245; primarily in patients with acute leukemias or in HCT recipients), prophylaxis with TMP/SMX resulted in a significant reduction in PJP occurrence by 91% compared with placebo, no treatment, or treatment with non-PJP antibiotics (RR, 0.09; 95% CI, 0.02–0.32). In addition, TMP/SMX prophylaxis significantly reduced PJP-related mortality (RR, 0.17; 95% CI, 0.03–0.94).266 TMP/SMX has the potential advantage of activity against other infectious complications (such as common bacterial infections, listeriosis, nocardiosis, and toxoplasmosis) that may afflict patients with severe T-cell depletion or impairment.270 TMP/SMX is considered the treatment of choice for PJP prophylaxis (preferred, category 1; Figure 8). In cases of intolerance, TMP/SMX desensitization should be considered. Atovaquone, dapsone, and aerosolized or intravenous pentamidine are alternatives to TMP/SMX in cases of intolerance.271–273 For patients receiving dapsone, measurement of G6PD levels is recommended before the start of therapy. Patients who are G6PD deficient may have an increased risk for hemolytic adverse reactions. Methemoglobinemia can also occur with dapsone therapy.274 Atovaquone appears to be equivalent to dapsone in HIV patients who cannot tolerate TMP/SMX.275 In pediatric patients with acute leukemias with intolerance of TMP/SMX, atovaquone was reported to be an effective strategy for PJP prophylaxis.276
Prophylaxis against PJP should be used in allogeneic HCT recipients (category 1) and patients receiving CAR T-cell therapy for at least 6 months and while receiving IST, as well as in patients with ALL (category 1) throughout antileukemic therapy.277,278 Patients should receive prophylaxis against PJP for a minimum of 2 months after alemtuzumab and until the CD4 count is >200 cells/mcL. Other patients who should receive PJP prophylaxis at least through active treatment include (1) those receiving treatment with select phosphatidylinositol-3-kinase inhibitors (copanlisib, idelalisib, or duvelisib) ± rituximab; (2) patients with neoplastic diseases receiving intensive corticosteroid treatment (eg, the equivalent of ≥20 mg of prednisone daily for ≥4 weeks, also depending on the patient’s overall immunologic status); and (3) patients receiving temozolomide (Figure 8).278–281 Some panel members advise prophylaxis against PJP (category 2B) for patients receiving purine analog therapy (eg, fludarabine, cladribine) and other T-cell–depleting agents until CD4 count is >200 cells/mcL and for recipients of autologous HCT until 3 to 6 months posttransplant. Prophylaxis against PJP may also be considered in patients receiving certain bispecific antibodies. For more information on infection risks associated with bispecific antibodies, see the full NCCN Guidelines for Prevention and Treatment of Cancer-Related Infections (available at NCCN.org).
Vaccination
Vaccination in patients with cancer can reduce the morbidity and mortality associated with infection. In general, patients with hematologic malignancies have a greater risk for infection than patients with solid tumors. Patients receiving HCT may lose immunity to pathogens posttransplant. Therefore, vaccination recommendations for these patients are more expansive than the recommendations for the general population of patients with cancer. In any patient who is immunocompromised, live vaccines, including the live attenuated influenza vaccine (LAIV), have the potential to cause disease and should not be administered during chemotherapy or periods of significant immunosuppression such as treatment of GVHD. The safety of vaccines for patients receiving immunostimulatory drugs has not been established. Inactivated vaccines can often be safely administered to patients with cancer. Although the immunogenicity of the vaccines may be reduced in patients who are immunocompromised, the potential for protection conferred by antigen-derived vaccines, even if incomplete, is better than no protection if the vaccine is withheld. While guidelines may provide general recommendations for vaccination schedules, the efficacy and safety of each vaccine should be evaluated to optimize the schedule on a case-by-case basis. For more information on vaccination in cancer survivors, see the NCCN Guidelines for Survivorship (available at NCCN.org).
Influenza Vaccine
Influenza infections cause significant morbidity and mortality in patients with cancer. Among bone marrow transplant recipients, influenza accounts for about 10%–40% of all community-acquired viral respiratory infections.282–284 An increase in both the incidence and duration of influenza infections has been observed in patients with cancer who are immunosuppressed compared with healthy controls.285,286 During community outbreaks, influenza infections may represent a significant proportion of fever and neutropenia episodes.287 Influenza infections in patients with cancer who are severely immunocompromised are often associated with hospitalizations, delays in potentially life-saving chemotherapy, and occasionally death.285–287 As a result, annual vaccination against influenza with the inactivated influenza virus is recommended for all individuals at increased risk due to immunosuppression.288 A randomized study of 97 patients receiving cytotoxic chemotherapy (3-week cycles) for solid tumors found that the immunogenicity of the influenza vaccine was similar when administered at the time of chemotherapy administration (day 1) or within the cytopenic period (day 11).289 The Advisory Committee on Immunization Practices (ACIP) for the CDC guidelines includes health care professionals and household members or caregivers in their target group for annual immunization to prevent transmission of influenza to patients at high risk.288
The intranasal vaccine should be avoided in patients with immunosuppression, because a LAIV is still capable of replication, which could theoretically lead to infection in immunocompromised individuals.288,290 Because no data are available assessing the risk for person-to-person transmission of the LAIV from vaccine recipients to immunosuppressed contacts, the CDC recommends that inactivated influenza vaccine should be used in household contacts, health care workers, and others who have close contact with patients who are severely immunocompromised (ie, persons requiring a protected environment). Persons with close contact to patients with a lesser degree of immunosuppression (eg, patients receiving chemotherapy or corticosteroids, patients with HIV) may receive the LAIV.288,290
There are not yet sufficient data for the panel to recommend the high-dose influenza vaccine over the standard-dose influenza vaccine. Preliminary data have shown that the high-dose influenza vaccine is safe for patients with cancer and may show more immunogenicity compared with the standard-dose influenza vaccine for this patient population.291,292 A randomized, single-blind, controlled trial of influenza vaccine in autologous HCT recipients found comparable seroprotection and seroconversion rates against all influenza strains with both the high-dose and the standard-dose vaccines when used as part of a 2-dose regimen.293 Further data are needed to assess whether the high-dose influenza vaccine confers a clinical benefit compared with the standard-dose vaccine for patients with cancer.
Pneumococcal Vaccine
The pneumococcal conjugate vaccine can be given in newly diagnosed adults with hematologic or solid tumor malignancies following assessment of their immune status. The conjugate pneumococcal vaccine (PCV20) should be administered to newly diagnosed adults with cancer who are pneumococcal vaccine-naïve. Alternatively, PCV15 can be given, followed by the polysaccharide pneumococcal vaccine (PPSV23) at least 8 weeks later. Additional PPSV23 is not needed for those receiving PCV20. For patients who have previously received PPSV23, PCV20 (preferred) or PCV15 can be given. Patients who have previously received PCV13 only can receive PCV20 (rather than PPSV23) at least 1 year later. Patients who have previously received PCV13 and 1 to 2 doses of PPSV23 can receive PCV20 at least 5 years later.
A phase III randomized controlled clinical trial found that PCV15 had a comparable safety profile to PCV13 in allo-HCT recipients and induced comparable immune responses to PCV13.294 Based on CDC recommendations, vaccination with the conjugated 20-valent or 15-valent vaccine 3 to 6 months after HCT is recommended. If PCV20 is used, 4 doses should be administered. The first 3 doses are generally 1 to 2 months apart, with the fourth dose 6 months after the third dose. There is no need to give PPSV23. If PCV15 is used, 3 doses should be administered, followed by PPSV23 6 to 12 months post primary series. Following the primary series of 3 PCV doses, a dose of the PPSV23 to broaden the immune response might be given. For patients with chronic GVHD in whom PPSV23 might not be as effective, a fourth dose of PCV20 or PCV15 should be considered instead of PPSV23. Patients with asplenia should receive the pneumococcal vaccine. According to the ACIP Vaccine Recommendations and Guidelines for Altered Immunocompetence, the pneumococcal vaccine should be administered at least 2 weeks before elective splenectomy. Penicillin prophylaxis is advised in patients who are asplenic to prevent pneumococcal disease.295,296
Meningococcal Conjugate Vaccine
The meningococcal vaccine is recommended for patients with increased risk for meningococcal disease including patients with persistent complement component deficiency, patients taking eculizumab, and patients with anatomic or functional asplenia. The ACIP recommends that asplenic persons be immunized with the meningococcal vaccine. The meningococcal vaccine should be administered at least 2 weeks before elective splenectomy. Patients at increased risk for meningococcal disease should receive quadrivalent conjugate vaccine series (MenACWY) and monovalent meningococcal serogroup B vaccine series. Patients at risk include those with persistent complement component deficiencies, those taking a complement C5 inhibitor (eg, eculizumab, ravulizumab), or those with anatomic or functional asplenia. MenACWY vaccine is given in 2 doses ≥8 weeks apart; serogroup B vaccine is available in a 2-or 3-dose series, depending on the vaccine formulation used. The meningococcal vaccine is recommended 6 to 12 months after HCT. Booster doses can be considered in those that remain at risk for meningococcal disease. This includes revaccination every 5 years with MenACWY vaccine, while for MenB vaccine, 1 booster dose is recommended 1 year after primary series with revaccination every 2–3 years if risk remains.
Human Papillomavirus Vaccine
The HPV vaccine is a recombinant 3-dose vaccine that is recommended for patients ≤26 years of age, and can be considered for those up to age 45. The lower age limit for this vaccine is 9 years of age. Although it does not treat existing HPV infection or disease, HPV vaccination is still recommended in individuals with prior HPV since it can still provide protection against HPV serotypes not already acquired.
Haemophilus Influenzae Type b Vaccine
Immunization of adults with the pediatric H. influenzae type b (Hib) vaccine is considered optional because of limited data on efficacy in older children and adults, although studies suggest good immunogenicity in patients who are immunocompromised. The Hib vaccine is recommended 6 to 12 months post-HCT. For patients with planned splenectomy, immunization is ideally performed at least 2 weeks in advance. If this is not feasible, immunization is advisable after splenectomy, because such patients are still capable of mounting a protective antibody response.
Varicella/Zoster Vaccines
The recombinant zoster vaccine (RZV) should be given to patients 50 to 70 days after autologous HCT. RZV may be considered after allogeneic HCT; however, its efficacy in allogeneic HCT, in the presence of GVHD, or during ongoing immunosuppression has not been established. The CDC recommends administration of RZV for patients aged ≥50 years, and those ≥18 years who are at increased risk for herpes zoster. The RZV vaccine is given in 2 doses 2 to 6 months apart. For adults who are immunocompromised and aged ≥18 years, a second dose can be given 1 to 2 months after the first dose if they will benefit from a shorter vaccination schedule. For patients who have previously received the live-attenuated herpes zoster vaccine, RZV should be given at least 2 months after the last live-attenuated herpes zoster dose.
COVID-19 Vaccine
The NCCN Guidelines recommend COVID-19 vaccination for all persons with cancer or who have been previously treated for cancer. According to the CDC, individuals aged ≥6 months who are moderately or severely immunocompromised (including recipients of HCT) and not previously vaccinated against COVID-19 should get 2 or 3 doses of the same brand of updated COVID-19 vaccine. Additionally, updated vaccines should be administered at least 2 weeks before initiation or resumption of immunosuppressive therapies. The NCCN Guidelines recommend vaccination with COVID-19 vaccine 6 months post-HCT, with a consideration for early vaccination at 3 months during community outbreaks and high disease activity. For further details of the most up-to-date vaccination recommendations, refer to CDC guidance.
Tetanus/Diphtheria/Pertussis (Tdap) Vaccine
As per the CDC, individuals ≥7 years should receive one dose of Tdap every 10 years. The FDA/ACIP approved a 3-dose series of Tdap/Tdap/Tdap or Tdap/Td/Td (tetanus/diphtheria) for individuals ≥7 years. For recipients of HCT, NCCN recommends administration of the 3-dose vaccine series, with the first dose 6 to 12 months post-HCT.
RSV Vaccine
The RSV vaccine is available for adults aged ≥60 years. However, its effectiveness in patients with cancer is unknown. The long-acting RSV monoclonal antibody (nirsevimab) is approved for infants <24 months of age to prevent RSV infection.
Travel Vaccines
Vaccines have variable risk and efficacy in patients receiving cancer care; therefore, the panel recommends consultation with an infectious disease expert before the administration of travel vaccines (eg, typhoid, yellow fever). Additional information on travel vaccines may be found in the CDC Yellow Book.
Vaccine Summary
Although efficacy data are lacking for the use of vaccines in patients with cancer, recommendations for their use are based on the principles of immunization and safety data. Persons receiving chemotherapy or radiation therapy for malignancies should not receive live vaccines for at least 3 months after cessation of therapy and until they are presumed to be immunocompetent.297 Live vaccines are contraindicated during treatment and for a period of at least 6 to 12 months in patients receiving IST (eg, blinatumomab, CAR T-cell therapy, monoclonal antibodies). These patients may also have a blunted response to inactivated vaccines. Certain live vaccines can be safely administered to household members of patients who are severely immunocompromised (eg, measles, mumps, rubella), whereas others cannot (eg, smallpox vaccine) because of the potential risk of transmission. The package insert for the vaccine should be reviewed prior to administration. The NCCN panel recommends that all household members be up to date on vaccinations.
Ideally, patients should be vaccinated at least 2 weeks before receiving cytotoxic therapy or IST; however, this timing is often not feasible in patients with cancer. In general, vaccination should not be given on the same day as cytotoxic therapy as cytotoxic therapy may reduce the proliferative lymphocytic responses required for protective immunity. In patients receiving chemotherapy, immunization between cytotoxic chemotherapy courses is likely to be associated with higher response rates than during chemotherapy administration.298,299 Patients vaccinated <2 weeks before starting cytotoxic therapy or IST or while receiving these agents may have a limited response to vaccination. These patients should be revaccinated at least 3 months after therapy is discontinued and once immune competence has been restored.297
In summary, the NCCN panel recommends that patients with cancer receive the influenza, COVID-19, pneumococcal, meningococcal, and HPV vaccines (Figure 9). HCT recipients should also receive the inactivated vaccines for Tdap, Hib, hepatitis A and B, RZV, and polio (Figure 10 and Figure 11). The live vaccines for measles, mumps, rubella, and varicella may be given if no GVHD or ongoing immunosuppression is seen 2 years posttransplant in patients who are seronegative. Consultation with an infectious disease expert is recommended before administration of travel vaccines.
Protected Environments
Although well-designed clinical trials have not validated the use of high-efficiency particulate air (HEPA) filtration, the CDC recommends that recipients of allogeneic HCT be placed in rooms with HEPA filters.123,300 It is also reasonable to use HEPA filtration in patients who are nontransplant recipients with prolonged neutropenia. The principal benefit of HEPA filtration is likely to be related to the prevention of mold infections. In a retrospective analysis, HEPA filters were protective in those who are highly immunocompromised with hematologic malignancies in the setting of an outbreak of aspergillosis.301 The value of laminar airflow in preventing infections is unclear and generally is not recommended.302
Summary
Substantial progress has been made in the prevention and treatment of infectious complications associated with neutropenia and IST in patients with cancer. However, infectious complications remain an important cause of morbidity and mortality in patients with cancer. The extent of infectious risk is highly dependent on an individual patient’s underlying malignancy, degree of neutropenia, past history of infections and exposure to pathogens, treatment with myelosuppressive regimens, and the overall status of immune function in the patient. In summary, prevention and treatment of infections in patients with cancer is a complex and continuously evolving field. Individualized risk evaluation for infections, incorporation of preventative measures, and prompt identification and treatment of active infections are essential components of the overall spectrum of care in cancer management and can contribute to optimizing treatment outcomes in patients with cancer.
References
- 1.↑
Kampouri E, Little JS, Rejeski K, et al. Infections after chimeric antigen receptor (CAR)-T-cell therapy for hematologic malignancies. Transpl Infect Dis 2023;25(Suppl 1):e14157.
- 2.↑
Shahid Z, Jain T, Dioverti V, et al. Best practice considerations by the American Society of Transplant and Cellular Therapy: infection prevention and management after chimeric antigen receptor T cell therapy for hematological malignancies. Transplant Cell Ther 2024;30:955–969.
- 3.↑
Hayden PJ, Roddie C, Bader P, et al. Management of adults and children receiving CAR T-cell therapy: 2021 best practice recommendations of the European Society for Blood and Marrow Transplantation (EBMT) and the Joint Accreditation Committee of ISCT and EBMT (JACIE) and the European Haematology Association (EHA). Ann Oncol 2022;33:259–275.
- 4.↑
Reynolds G, Sim B, Anderson MA, et al. Predicting infections in patients with haematological malignancies treated with chimeric antigen receptor T-cell therapies: a systematic scoping review and narrative synthesis. Clin Microbiol Infect 2023;29:1280–1288.
- 5.↑
Bucaneve G, Micozzi A, Menichetti F, et al. Levofloxacin to prevent bacterial infection in patients with cancer and neutropenia. N Engl J Med 2005;353:977–987.
- 6.↑
Cullen M, Steven N, Billingham L, et al. Antibacterial prophylaxis after chemotherapy for solid tumors and lymphomas. N Engl J Med 2005;353:988–998.
- 7.↑
Eleutherakis-Papaiakovou E, Kostis E, Migkou M, et al. Prophylactic antibiotics for the prevention of neutropenic fever in patients undergoing autologous stem-cell transplantation: results of a single institution, randomized phase 2 trial. Am J Hematol 2010;85:863–867.
- 8.↑
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:e56–93.
- 9.↑
Gafter-Gvili A, Fraser A, Paul M, et al. Antibiotic prophylaxis for bacterial infections in afebrile neutropenic patients following chemotherapy. Cochrane Database Syst Rev 2012;1:CD004386.
- 10.↑
Gafter-Gvili A, Paul M, Fraser A, Leibovici L. Effect of quinolone prophylaxis in afebrile neutropenic patients on microbial resistance: systematic review and meta-analysis. J Antimicrob Chemother 2007;59:5–22.
- 11.↑
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;65:1819–1828.
- 12.↑
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;76:20–37.
- 13.↑
Cullen MH, Billingham LJ, Gaunt CH, Steven NM. Rational selection of patients for antibacterial prophylaxis after chemotherapy. J Clin Oncol 2007;25:4821–4828.
- 14.↑
Hoffman T, Atamna A, Litchevsky V, et al. Fluoroquinolone prophylaxis during conventional chemotherapy or hematopoietic stem cell transplantation for acute leukemia - pros and cons. Acta Haematol 2024;147:186–197.
- 15.↑
Bartlett JG, Perl TM. The new Clostridium difficile–what does it mean? N Engl J Med 2005;353:2503–2505.
- 16.↑
Cook PP, Catrou P, Gooch M, Holbert D. Effect of reduction in ciprofloxacin use on prevalence of meticillin-resistant Staphylococcus aureus rates within individual units of a tertiary care hospital. J Hosp Infect 2006;64:348–351.
- 17.↑
Loo VG, Poirier L, Miller MA, et al. A predominantly clonal multi-institutional outbreak of Clostridium difficile-associated diarrhea with high morbidity and mortality. N Engl J Med 2005;353:2442–2449.
- 18.↑
McDonald LC, Killgore GE, Thompson A, et al. An epidemic, toxin gene-variant strain of Clostridium difficile. N Engl J Med 2005;353:2433–2441.
- 19.↑
Lehrnbecher T, Fisher BT, Phillips B, et al. Guideline for antibacterial prophylaxis administration in pediatric cancer and hematopoietic stem cell transplantation. Clin Infect Dis 2020;71:226–236.
- 20.↑
Engelhard D, Cordonnier C, Shaw PJ, et al. Early and late invasive pneumococcal infection following stem cell transplantation: a European Bone Marrow Transplantation survey. Br J Haematol 2002;117:444–450.
- 21.↑
Kulkarni S, Powles R, Treleaven J, et al. Chronic graft versus host disease is associated with long-term risk for pneumococcal infections in recipients of bone marrow transplants. Blood 2000;95:3683–3686.
- 22.↑
Youssef S, Rodriguez G, Rolston KV, et al. Streptococcus pneumoniae infections in 47 hematopoietic stem cell transplantation recipients: clinical characteristics of infections and vaccine-breakthrough infections, 1989–2005. Medicine (Baltimore) 2007;86:69–77.
- 23.↑
Engelhard D, Akova M, Boeckh MJ, et al. Bacterial infection prevention after hematopoietic cell transplantation. Bone Marrow Transplant 2009;44:467–470.
- 24.↑
Brown SD, Rybak MJ. Antimicrobial susceptibility of Streptococcus pneumoniae, Streptococcus pyogenes and Haemophilus influenzae collected from patients across the USA, in 2001–2002, as part of the PROTEKT US study. J Antimicrob Chemother 2004;54(Suppl 1):i7–15.
- 25.↑
Kumashi P, Girgawy E, Tarrand JJ, et al. Streptococcus pneumoniae bacteremia in patients with cancer: disease characteristics and outcomes in the era of escalating drug resistance (1998–2002). Medicine (Baltimore) 2005;84:303–312.
- 26.↑
Wingard JR, Carter SL, Walsh TJ, et al. Randomized, double-blind trial of fluconazole versus voriconazole for prevention of invasive fungal infection after allogeneic hematopoietic cell transplantation. Blood 2010;116:5111–5118.
- 27.↑
Cornely OA, Maertens J, Winston DJ, et al. Posaconazole vs. fluconazole or itraconazole prophylaxis in patients with neutropenia. N Engl J Med 2007;356:348–359.
- 28.↑
Ullmann AJ, Lipton JH, Vesole DH, et al. Posaconazole or fluconazole for prophylaxis in severe graft-versus-host disease. N Engl J Med 2007;356:335–347.
- 29.↑
Vazquez JA, Skiest DJ, Nieto L, et al. A multicenter randomized trial evaluating posaconazole versus fluconazole for the treatment of oropharyngeal candidiasis in subjects with HIV/AIDS. Clin Infect Dis 2006;42:1179–1186.
- 30.↑
Maertens JA, Rahav G, Lee DG, et al. Posaconazole versus voriconazole for primary treatment of invasive aspergillosis: a phase 3, randomised, controlled, non-inferiority trial. Lancet 2021;397:499–509.
- 31.↑
Wang J, Zhou M, Xu JY, et al. Comparison of antifungal prophylaxis drugs in patients with hematological disease or undergoing hematopoietic stem cell transplantation: a systematic review and network meta-analysis. JAMA Netw Open 2020;3:e2017652.
- 32.↑
Kato H, Hagihara M, Asai N, et al. A systematic review and meta-analysis of efficacy and safety of isavuconazole for the treatment and prophylaxis of invasive fungal infections. Mycoses 2023;66:815–824.
- 33.↑
Cowen EW, Nguyen JC, Miller DD, et al. Chronic phototoxicity and aggressive squamous cell carcinoma of the skin in children and adults during treatment with voriconazole. J Am Acad Dermatol 2010;62:31–37.
- 34.↑
Gerber B, Guggenberger R, Fasler D, et al. Reversible skeletal disease and high fluoride serum levels in hematologic patients receiving voriconazole. Blood 2012;120:2390–2394.
- 35.↑
Miller DD, Cowen EW, Nguyen JC, et al. Melanoma associated with long-term voriconazole therapy: a new manifestation of chronic photosensitivity. Arch Dermatol 2010;146:300–304.
- 36.↑
Tarlock K, Johnson D, Cornell C, et al. Elevated fluoride levels and periostitis in pediatric hematopoietic stem cell transplant recipients receiving long-term voriconazole. Pediatr Blood Cancer 2015;62:918–920.
- 37.↑
Williams K, Mansh M, Chin-Hong P, et al. Voriconazole-associated cutaneous malignancy: a literature review on photocarcinogenesis in organ transplant recipients. Clin Infect Dis 2014;58:997–1002.
- 38.↑
Marr KA, Crippa F, Leisenring W, et al. Itraconazole versus fluconazole for prevention of fungal infections in patients receiving allogeneic stem cell transplants. Blood 2004;103:1527–1533.
- 39.↑
Marr KA, Leisenring W, Crippa F, et al. Cyclophosphamide metabolism is affected by azole antifungals. Blood 2004;103:1557–1559.
- 41.↑
Kably B, Launay M, Derobertmasure A, et al. Antifungal drugs TDM: trends and update. Ther Drug Monit 2022;44:166–197.
- 42.↑
Andes D, van Ogtrop M. Characterization and quantitation of the pharmacodynamics of fluconazole in a neutropenic murine disseminated candidiasis infection model. Antimicrob Agents Chemother 1999;43:2116–2120.
- 43.↑
Clancy CJ, Yu VL, Morris AJ, et al. Fluconazole MIC and the fluconazole dose/MIC ratio correlate with therapeutic response among patients with candidemia. Antimicrob Agents Chemother 2005;49:3171–3177.
- 44.↑
Csajka C, Decosterd LA, Buclin T, et al. Population pharmacokinetics of fluconazole given for secondary prevention of oropharyngeal candidiasis in HIV-positive patients. Eur J Clin Pharmacol 2001;57:723–727.
- 45.↑
Rex JH, Pfaller MA, Galgiani JN, et al. Development of interpretive breakpoints for antifungal susceptibility testing: conceptual framework and analysis of in vitro-in vivo correlation data for fluconazole, itraconazole, and candida infections. Subcommittee on Antifungal Susceptibility Testing of the National Committee for Clinical Laboratory Standards. Clin Infect Dis 1997;24:235–247.
- 46.↑
Krishna G, Moton A, Ma L, et al. Pharmacokinetics and absorption of posaconazole oral suspension under various gastric conditions in healthy volunteers. Antimicrob Agents Chemother 2009;53:958–966.
- 47.↑
Gross BN, Ihorst G, Jung M, et al. Posaconazole therapeutic drug monitoring in the real-life setting: a single-center experience and review of the literature. Pharmacotherapy 2013;33:1117–1125.
- 48.↑
Bryant AM, Slain D, Cumpston A, Craig M. A post-marketing evaluation of posaconazole plasma concentrations in neutropenic patients with haematological malignancy receiving posaconazole prophylaxis. Int J Antimicrob Agents 2011;37:266–269.
- 49.↑
Bruggemann RJ, Touw DJ, Aarnoutse RE, et al. International interlaboratory proficiency testing program for measurement of azole antifungal plasma concentrations. Antimicrob Agents Chemother 2009;53:303–305.
- 50.↑
Kraft WK, Chang PS, van Iersel ML, et al. Posaconazole tablet pharmacokinetics: lack of effect of concomitant medications altering gastric pH and gastric motility in healthy subjects. Antimicrob Agents Chemother 2014;58:4020–4025.
- 51.↑
Heinz WJ, Egerer G, Lellek H, et al. Posaconazole after previous antifungal therapy with voriconazole for therapy of invasive aspergillus disease, a retrospective analysis. Mycoses 2013;56:304–310.
- 52.↑
Maertens J, Cornely OA, Ullmann AJ, et al. Phase 1B study of the pharmacokinetics and safety of posaconazole intravenous solution in patients at risk for invasive fungal disease. Antimicrob Agents Chemother 2014;58:3610–3617.
- 53.↑
Jang SH, Colangelo PM, Gobburu JV. Exposure-response of posaconazole used for prophylaxis against invasive fungal infections: evaluating the need to adjust doses based on drug concentrations in plasma. Clin Pharmacol Ther 2010;88:115–119.
- 54.↑
Tonini J, Thiebaut A, Jourdil JF, et al. Therapeutic drug monitoring of posaconazole in allogeneic hematopoietic stem cell transplantation patients who develop gastrointestinal graft-versus-host disease. Antimicrob Agents Chemother 2012;56:5247–5252.
- 55.↑
Shields RK, Clancy CJ, Vadnerkar A, et al. Posaconazole serum concentrations among cardiothoracic transplant recipients: factors impacting trough levels and correlation with clinical response to therapy. Antimicrob Agents Chemother 2011;55:1308–1311.
- 56.↑
Eiden C, Meniane JC, Peyriere H, et al. Therapeutic drug monitoring of posaconazole in hematology adults under posaconazole prophylaxis: influence of food intake. Eur J Clin Microbiol Infect Dis 2012;31:161–167.
- 57.↑
Hoenigl M, Raggam RB, Salzer HJ, et al. Posaconazole plasma concentrations and invasive mould infections in patients with haematological malignancies. Int J Antimicrob Agents 2012;39:510–513.
- 58.↑
Lebeaux D, Lanternier F, Elie C, et al. Therapeutic drug monitoring of posaconazole: a monocentric study with 54 adults. Antimicrob Agents Chemother 2009;53:5224–5229.
- 59.↑
Howard SJ, Cerar D, Anderson MJ, et al. Frequency and evolution of Azole resistance in Aspergillus fumigatus associated with treatment failure. Emerg Infect Dis 2009;15:1068–1076.
- 60.↑
Mavridou E, Bruggemann RJ, Melchers WJ, et al. Efficacy of posaconazole against three clinical Aspergillus fumigatus isolates with mutations in the cyp51A gene. Antimicrob Agents Chemother 2010;54:860–865.
- 61.↑
Boogaerts MA, Verhoef GE, Zachee P, et al. Antifungal prophylaxis with itraconazole in prolonged neutropenia: correlation with plasma levels. Mycoses 1989;32(Suppl 1):103–108.
- 62.↑
Tricot G, Joosten E, Boogaerts MA, et al. Ketoconazole vs. itraconazole for antifungal prophylaxis in patients with severe granulocytopenia: preliminary results of two nonrandomized studies. Rev Infect Dis 1987;9(Suppl 1):S94–99.
- 63.↑
Glasmacher A, Hahn C, Leutner C, et al. Breakthrough invasive fungal infections in neutropenic patients after prophylaxis with itraconazole. Mycoses 1999;42:443–451.
- 64.↑
Glasmacher A, Hahn C, Molitor E, et al. Itraconazole trough concentrations in antifungal prophylaxis with six different dosing regimens using hydroxypropyl-beta-cyclodextrin oral solution or coated-pellet capsules. Mycoses 1999;42:591–600.
- 65.↑
Denning DW, Tucker RM, Hanson LH, et al. Itraconazole therapy for cryptococcal meningitis and cryptococcosis. Arch Intern Med 1989;149:2301–2308.
- 66.↑
Denning DW, Tucker RM, Hanson LH, Stevens DA. Treatment of invasive aspergillosis with itraconazole. Am J Med 1989;86:791–800.
- 67.↑
Sharkey PK, Rinaldi MG, Dunn JF, et al. High-dose itraconazole in the treatment of severe mycoses. Antimicrob Agents Chemother 1991;35:707–713.
- 68.↑
Wheat J, Hafner R, Korzun AH, et al. Itraconazole treatment of disseminated histoplasmosis in patients with the acquired immunodeficiency syndrome. AIDS Clinical Trial Group. Am J Med 1995;98: 336–342.
- 69.↑
Lestner JM, Roberts SA, Moore CB, et al. Toxicodynamics of itraconazole: implications for therapeutic drug monitoring. Clin Infect Dis 2009;49:928–930.
- 70.↑
Barone JA, Moskovitz BL, Guarnieri J, et al. Food interaction and steady-state pharmacokinetics of itraconazole oral solution in healthy volunteers. Pharmacotherapy 1998;18:295–301.
- 71.↑
Lindsay J, Othman J, Kong Y, et al. SUBA-itraconazole for primary antifungal prophylaxis after allogeneic hematopoietic cell transplantation. Open Forum Infect Dis 2021;8:ofab502.
- 72.↑
Liu J, Vanderwyk KA, Donnelley MA, Thompson GR III. SUBA-itraconazole in the treatment of systemic fungal infections. Future Microbiol 2024;19:1171–1175.
- 73.↑
Pascual A, Calandra T, Bolay S, et al. Voriconazole therapeutic drug monitoring in patients with invasive mycoses improves efficacy and safety outcomes. Clin Infect Dis 2008;46:201–211.
- 74.↑
Jeans AR, Howard SJ, Al-Nakeeb Z, et al. Combination of voriconazole and anidulafungin for treatment of triazole-resistant aspergillus fumigatus in an in vitro model of invasive pulmonary aspergillosis. Antimicrob Agents Chemother 2012;56:5180–5185.
- 75.↑
Smith J, Safdar N, Knasinski V, et al. Voriconazole therapeutic drug monitoring. Antimicrob Agents Chemother 2006;50:1570–1572.
- 76.↑
Troke PF, Hockey HP, Hope WW. Observational study of the clinical efficacy of voriconazole and its relationship to plasma concentrations in patients. Antimicrob Agents Chemother 2011;55:4782–4788.
- 77.↑
Dolton MJ, Ray JE, Chen SC, et al. Multicenter study of voriconazole pharmacokinetics and therapeutic drug monitoring. Antimicrob Agents Chemother 2012;56:4793–4799.
- 78.↑
Miyakis S, van Hal SJ, Ray J, Marriott D. Voriconazole concentrations and outcome of invasive fungal infections. Clin Microbiol Infect 2010;16:927–933.
- 79.↑
Gomez-Lopez A, Cendejas-Bueno E, Cuesta I, et al. Voriconazole serum levels measured by high-performance liquid chromatography: a monocentric study in treated patients. Med Mycol 2012;50:439–445.
- 80.↑
Ueda K, Nannya Y, Kumano K, et al. Monitoring trough concentration of voriconazole is important to ensure successful antifungal therapy and to avoid hepatic damage in patients with hematological disorders. Int J Hematol 2009;89:592–599.
- 81.↑
Trifilio S, Singhal S, Williams S, et al. Breakthrough fungal infections after allogeneic hematopoietic stem cell transplantation in patients on prophylactic voriconazole. Bone Marrow Transplant 2007;40:451–456.
- 82.↑
Mitsani D, Nguyen MH, Shields RK, et al. Prospective, observational study of voriconazole therapeutic drug monitoring among lung transplant recipients receiving prophylaxis: factors impacting levels of and associations between serum troughs, efficacy, and toxicity. Antimicrob Agents Chemother 2012;56:2371–2377.
- 83.↑
Pascual A, Csajka C, Buclin T, et al. Challenging recommended oral and intravenous voriconazole doses for improved efficacy and safety: population pharmacokinetics-based analysis of adult patients with invasive fungal infections. Clin Infect Dis 2012;55:381–390.
- 84.↑
Zonios DI, Gea-Banacloche J, Childs R, Bennett JE. Hallucinations during voriconazole therapy. Clin Infect Dis 2008;47:e7–10.
- 85.↑
Hamada Y, Seto Y, Yago K, Kuroyama M. Investigation and threshold of optimum blood concentration of voriconazole: a descriptive statistical meta-analysis. J Infect Chemother 2012;18:501–507.
- 86.↑
Kim KH, Lee S, Lee S, et al. Voriconazole-associated severe hyponatremia. Med Mycol 2012;50:103–105.
- 87.↑
Matsumoto K, Ikawa K, Abematsu K, et al. Correlation between voriconazole trough plasma concentration and hepatotoxicity in patients with different CYP2C19 genotypes. Int J Antimicrob Agents 2009;34:91–94.
- 88.↑
Purkins L, Wood N, Kleinermans D, et al. Effect of food on the pharmacokinetics of multiple-dose oral voriconazole. Br J Clin Pharmacol 2003;56(Suppl 1):17–23.
- 89.↑
McCreary EK, Davis MR, Narayanan N, et al. Utility of triazole antifungal therapeutic drug monitoring: insights from the Society of Infectious Diseases Pharmacists: endorsed by the Mycoses Study Group Education and Research Consortium. Pharmacotherapy 2023;43:1043–1050.
- 90.↑
Koh LP, Kurup A, Goh YT, et al. Randomized trial of fluconazole versus low-dose amphotericin B in prophylaxis against fungal infections in patients undergoing hematopoietic stem cell transplantation. Am J Hematol 2002;71:260–267.
- 91.↑
Winston DJ, Maziarz RT, Chandrasekar PH, et al. Intravenous and oral itraconazole versus intravenous and oral fluconazole for long-term antifungal prophylaxis in allogeneic hematopoietic stem-cell transplant recipients. A multicenter, randomized trial. Ann Intern Med 2003;138:705–713.
- 92.↑
Bates DW, Su L, Yu DT, et al. Correlates of acute renal failure in patients receiving parenteral amphotericin B. Kidney Int 2001;60:1452–1459.
- 93.↑
Wingard JR, Kubilis P, Lee L, et al. Clinical significance of nephrotoxicity in patients treated with amphotericin B for suspected or proven aspergillosis. Clin Infect Dis 1999;29:1402–1407.
- 94.↑
Rijnders BJ, Cornelissen JJ, Slobbe L, et al. Aerosolized liposomal amphotericin B for the prevention of invasive pulmonary aspergillosis during prolonged neutropenia: a randomized, placebo-controlled trial. Clin Infect Dis 2008;46:1401–1408.
- 95.↑
Perfect JR. Aerosolized antifungal prophylaxis: the winds of change? Clin Infect Dis 2008;46:1409–1411.
- 96.↑
Espinel-Ingroff A. In vitro antifungal activities of anidulafungin and micafungin, licensed agents and the investigational triazole posaconazole as determined by NCCLS methods for 12,052 fungal isolates: review of the literature. Rev Iberoam Micol 2003;20:121–136.
- 97.↑
Villanueva A, Arathoon EG, Gotuzzo E, et al. A randomized double-blind study of caspofungin versus amphotericin for the treatment of candidal esophagitis. Clin Infect Dis 2001;33:1529–1535.
- 98.↑
Maertens J, Raad I, Petrikkos G, et al. Efficacy and safety of caspofungin for treatment of invasive aspergillosis in patients refractory to or intolerant of conventional antifungal therapy. Clin Infect Dis 2004;39:1563–1571.
- 99.↑
Kartsonis NA, Saah AJ, Joy Lipka C, et al. Salvage therapy with caspofungin for invasive aspergillosis: results from the caspofungin compassionate use study. J Infect 2005;50:196–205.
- 100.↑
van Burik JH, Ratanatharathorn V, Stepan DE, et al. Micafungin versus fluconazole for prophylaxis against invasive fungal infections during neutropenia in patients undergoing hematopoietic stem cell transplantation. Clin Infect Dis 2004;39:1407–1416.
- 101.↑
Yokote T, Akioka T, Oka S, et al. Successful treatment with micafungin of invasive pulmonary aspergillosis in acute myeloid leukemia, with renal failure due to amphotericin B therapy. Ann Hematol 2004;83:64–66.
- 102.↑
Ota S, Tanaka J, Kahata K, et al. Successful micafungin (FK463) treatment of invasive pulmonary aspergillosis in a patient with acute lymphoblastic leukemia in a phase II study. Int J Hematol 2004;79:390–393.
- 103.↑
Singer MS, Seibel NL, Vezina G, et al. Successful treatment of invasive aspergillosis in two patients with acute myelogenous leukemia. J Pediatr Hematol Oncol 2003;25:252–256.
- 104.↑
Krause DS, Simjee AE, van Rensburg C, et al. A randomized, double-blind trial of anidulafungin versus fluconazole for the treatment of esophageal candidiasis. Clin Infect Dis 2004;39:770–775.
- 105.↑
Vazquez JA, Schranz JA, Clark K, et al. A phase 2, open-label study of the safety and efficacy of intravenous anidulafungin as a treatment for azole-refractory mucosal candidiasis. J Acquir Immune Defic Syndr 2008;48:304–309.
- 106.↑
Reboli AC, Rotstein C, Pappas PG, et al. Anidulafungin versus fluconazole for invasive candidiasis. N Engl J Med 2007;356:2472–2482.
- 107.↑
Thompson GR, Soriano A, Cornely OA, et al. Rezafungin versus caspofungin for treatment of candidaemia and invasive candidiasis (ReSTORE): a multicentre, double-blind, double-dummy, randomised phase 3 trial. Lancet 2023;401:49–59.
- 108.↑
Harnicar S, Adel N, Jurcic J. Modification of vincristine dosing during concomitant azole therapy in adult acute lymphoblastic leukemia patients. J Oncol Pharm Pract 2009;15:175–182.
- 109.↑
Bodey GP, Mardani M, Hanna HA, et al. The epidemiology of Candida glabrata and Candida albicans fungemia in immunocompromised patients with cancer. Am J Med 2002;112:380–385.
- 110.↑
Marr KA, Seidel K, White TC, Bowden RA. Candidemia in allogeneic blood and marrow transplant recipients: evolution of risk factors after the adoption of prophylactic fluconazole. J Infect Dis 2000;181:309–316.
- 111.↑
Marr KA, Seidel K, Slavin MA, et al. Prolonged fluconazole prophylaxis is associated with persistent protection against candidiasis-related death in allogeneic marrow transplant recipients: long-term follow-up of a randomized, placebo-controlled trial. Blood 2000;96:2055–2061.
- 112.↑
Ullmann AJ, Schmidt-Hieber M, Bertz H, et al. Infectious diseases in allogeneic haematopoietic stem cell transplantation: prevention and prophylaxis strategy guidelines 2016. Ann Hematol 2016;95:1435–1455.
- 113.↑
Stemler J, Mellinghoff SC, Khodamoradi Y, et al. Primary prophylaxis of invasive fungal diseases in patients with haematological malignancies: 2022 update of the recommendations of the Infectious Diseases Working Party (AGIHO) of the German Society for Haematology and Medical Oncology (DGHO). J Antimicrob Chemother 2023;78:1813–1826.
- 114.↑
Weinberger M, Elattar I, Marshall D, et al. Patterns of infection in patients with aplastic anemia and the emergence of Aspergillus as a major cause of death. Medicine (Baltimore) 1992;71:24–43.
- 115.↑
Walsh TJ, Whitcomb PO, Revankar SG, Pizzo PA. Successful treatment of hepatosplenic candidiasis through repeated cycles of chemotherapy and neutropenia. Cancer 1995;76:2357–2362.
- 116.↑
Offner F, Cordonnier C, Ljungman P, et al. Impact of previous aspergillosis on the outcome of bone marrow transplantation. Clin Infect Dis 1998;26:1098–1103.
- 117.↑
Fukuda T, Boeckh M, Guthrie KA, et al. Invasive aspergillosis before allogeneic hematopoietic stem cell transplantation: 10-year experience at a single transplant center. Biol Blood Marrow Transplant 2004;10:494–503.
- 118.↑
Meyers JD, Flournoy N, Thomas ED. Infection with herpes simplex virus and cell-mediated immunity after marrow transplant. J Infect Dis 1980;142:338–346.
- 119.↑
Saral R, Burns WH, Laskin OL, et al. Acyclovir prophylaxis of herpes-simplex-virus infections. N Engl J Med 1981;305:63–67.
- 120.↑
Saral R, Ambinder RF, Burns WH, et al. Acyclovir prophylaxis against herpes simplex virus infection in patients with leukemia. A randomized, double-blind, placebo-controlled study. Ann Intern Med 1983;99:773–776.
- 121.↑
Mackall C, Fry T, Gress R, et al. Background to hematopoietic cell transplantation, including post transplant immune recovery. Bone Marrow Transplant 2009;44:457–462.
- 122.↑
Tomblyn M, Chiller T, Einsele H, et al. Guidelines for preventing infectious complications among hematopoietic cell transplantation recipients: a global perspective. Biol Blood Marrow Transplant 2009;15:1143–1238.
- 123.↑
Sullivan KM, Dykewicz CA, Longworth DL, et al. Preventing opportunistic infections after hematopoietic stem cell transplantation: the Centers for Disease Control and Prevention, Infectious Diseases Society of America, and American Society for Blood and Marrow Transplantation Practice Guidelines and beyond. Hematology Am Soc Hematol Educ Program 2001:392–421.
- 124.↑
Zaia J, Baden L, Boeckh MJ, et al. Viral disease prevention after hematopoietic cell transplantation. Bone Marrow Transplant 2009;44:471–482.
- 125.↑
Keating M, Coutre S, Rai K, et al. Management guidelines for use of alemtuzumab in B-cell chronic lymphocytic leukemia. Clin Lymphoma 2004;4:220–227.
- 126.↑
Boeckh M, Kim HW, Flowers MED, et al. Long-term acyclovir for prevention of varicella zoster virus disease after allogeneic hematopoietic cell transplantation–a randomized double-blind placebo-controlled study. Blood 2006;107:1800–1805.
- 127.↑
Erard V, Guthrie KA, Varley C, et al. One-year acyclovir prophylaxis for preventing varicella-zoster virus disease after hematopoietic cell transplantation: no evidence of rebound varicella-zoster virus disease after drug discontinuation. Blood 2007;110:3071–3077.
- 128.↑
Asano-Mori Y, Kanda Y, Oshima K, et al. Long-term ultra-low-dose acyclovir against varicella-zoster virus reactivation after allogeneic hematopoietic stem cell transplantation. Am J Hematol 2008;83:472–476.
- 129.↑
Oshima K, Takahashi T, Mori T, et al. One-year low-dose valacyclovir as prophylaxis for varicella zoster virus disease after allogeneic hematopoietic stem cell transplantation. A prospective study of the Japan Hematology and Oncology Clinical Study Group. Transpl Infect Dis 2010;12:421–427.
- 130.↑
Schuchter LM, Wingard JR, Piantadosi S, et al. Herpes zoster infection after autologous bone marrow transplantation. Blood 1989;74:1424–1427.
- 131.↑
Chanan-Khan A, Sonneveld P, Schuster MW, et al. Analysis of herpes zoster events among bortezomib-treated patients in the phase III APEX study. J Clin Oncol 2008;26:4784–4790.
- 132.↑
Reece DE, Rodriguez GP, Chen C, et al. Phase I-II trial of bortezomib plus oral cyclophosphamide and prednisone in relapsed and refractory multiple myeloma. J Clin Oncol 2008;26:4777–4783.
- 133.↑
Tong Y, Qian J, Li Y, et al. The high incidence of varicella herpes zoster with the use of bortezomib in 10 patients. Am J Hematol 2007;82:403–404.
- 134.↑
Varettoni M, Vassallo C, Borroni G, et al. Late onset of bortezomib-associated cutaneous reaction following herpes zoster. Ann Hematol 2007;86:301–302.
- 135.↑
Pour L, Adam Z, Buresova L, et al. Varicella-zoster virus prophylaxis with low-dose acyclovir in patients with multiple myeloma treated with bortezomib. Clin Lymphoma Myeloma 2009;9:151–153.
- 136.↑
Vickrey E, Allen S, Mehta J, Singhal S. Acyclovir to prevent reactivation of varicella zoster virus (herpes zoster) in multiple myeloma patients receiving bortezomib therapy. Cancer 2009;115:229–232.
- 137.↑
George B, Pati N, Gilroy N, et al. Pre-transplant cytomegalovirus (CMV) serostatus remains the most important determinant of CMV reactivation after allogeneic hematopoietic stem cell transplantation in the era of surveillance and preemptive therapy. Transpl Infect Dis 2010;12:322–329.
- 138.↑
Beck JC, Wagner JE, DeFor TE, et al. Impact of cytomegalovirus (CMV) reactivation after umbilical cord blood transplantation. Biol Blood Marrow Transplant 2010;16:215–222.
- 139.↑
Milano F, Pergam SA, Xie H, et al. Intensive strategy to prevent CMV disease in seropositive umbilical cord blood transplant recipients. Blood 2011;118:5689–5696.
- 140.↑
Montesinos P, Sanz J, Cantero S, et al. Incidence, risk factors, and outcome of cytomegalovirus infection and disease in patients receiving prophylaxis with oral valganciclovir or intravenous ganciclovir after umbilical cord blood transplantation. Biol Blood Marrow Transplant 2009;15:730–740.
- 141.↑
Chemaly RF, Ullmann AJ, Stoelben S, et al. Letermovir for cytomegalovirus prophylaxis in hematopoietic-cell transplantation. N Engl J Med 2014;370:1781–1789.
- 142.↑
Marty FM, Ljungman P, Chemaly RF, et al. Letermovir prophylaxis for cytomegalovirus in hematopoietic-cell transplantation. N Engl J Med 2017;377:2433–2444.
- 143.↑
Alain S, Feghoul L, Girault S, et al. Letermovir breakthroughs during the French Named Patient Programme: interest of monitoring blood concentration in clinical practice. J Antimicrob Chemother 2020;75:2253–2257.
- 144.↑
Cherrier L, Nasar A, Goodlet KJ, et al. Emergence of letermovir resistance in a lung transplant recipient with ganciclovir-resistant cytomegalovirus infection. Am J Transplant 2018;18:3060–3064.
- 145.↑
Frietsch JJ, Michel D, Stamminger T, et al. In vivo emergence of UL56 C325Y cytomegalovirus resistance to letermovir in a patient with acute myeloid leukemia after hematopoietic cell transplantation. Mediterr J Hematol Infect Dis 2019;11:e2019001.
- 146.↑
Reusser P, Einsele H, Lee J, et al. Randomized multicenter trial of foscarnet versus ganciclovir for preemptive therapy of cytomegalovirus infection after allogeneic stem cell transplantation. Blood 2002;99:1159–1164.
- 147.↑
Reusser P, Gambertoglio JG, Lilleby K, Meyers JD. Phase I-II trial of foscarnet for prevention of cytomegalovirus infection in autologous and allogeneic marrow transplant recipients. J Infect Dis 1992;166:473–479.
- 148.↑
Bacigalupo A, Tedone E, Van Lint MT, et al. CMV prophylaxis with foscarnet in allogeneic bone marrow transplant recipients at high risk of developing CMV infections. Bone Marrow Transplant 1994;13:783–788.
- 149.↑
Bregante S, Bertilson S, Tedone E, et al. Foscarnet prophylaxis of cytomegalovirus infections in patients undergoing allogeneic bone marrow transplantation (BMT): a dose-finding study. Bone Marrow Transplant 2000;26:23–29.
- 150.↑
Einsele H, Reusser P, Bornhauser M, et al. Oral valganciclovir leads to higher exposure to ganciclovir than intravenous ganciclovir in patients following allogeneic stem cell transplantation. Blood 2006;107:3002–3008.
- 151.↑
Winston DJ, Baden LR, Gabriel DA, et al. Pharmacokinetics of ganciclovir after oral valganciclovir versus intravenous ganciclovir in allogeneic stem cell transplant patients with graft-versus-host disease of the gastrointestinal tract. Biol Blood Marrow Transplant 2006;12:635–640.
- 152.↑
Ayala E, Greene J, Sandin R, et al. Valganciclovir is safe and effective as pre-emptive therapy for CMV infection in allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 2006;37:851–856.
- 153.↑
Busca A, de Fabritiis P, Ghisetti V, et al. Oral valganciclovir as preemptive therapy for cytomegalovirus infection post allogeneic stem cell transplantation. Transpl Infect Dis 2007;9:102–107.
- 154.↑
van der Heiden PLJ, Kalpoe JS, Barge RM, et al. Oral valganciclovir as pre-emptive therapy has similar efficacy on cytomegalovirus DNA load reduction as intravenous ganciclovir in allogeneic stem cell transplantation recipients. Bone Marrow Transplant 2006;37:693–698.
- 155.↑
Kalil AC, Freifeld AG, Lyden ER, Stoner JA. Valganciclovir for cytomegalovirus prevention in solid organ transplant patients: an evidence-based reassessment of safety and efficacy. PLoS One 2009;4:e5512.
- 156.↑
Kalil AC, Mindru C, Botha JF, et al. Risk of cytomegalovirus disease in high-risk liver transplant recipients on valganciclovir prophylaxis: a systematic review and meta-analysis. Liver Transpl 2012;18:1440–1447.
- 157.↑
Bosi A, Bartolozzi B, Vannucchi AM, et al. Polymerase chain reaction-based “pre-emptive” therapy with cidofovir for cytomegalovirus reactivation in allogeneic hematopoietic stem cells transplantation recipients: a prospective study. Haematologica 2002;87:446–447.
- 158.↑
Chakrabarti S, Collingham KE, Osman H, et al. Cidofovir as primary pre-emptive therapy for post-transplant cytomegalovirus infections. Bone Marrow Transplant 2001;28:879–881.
- 159.↑
Ljungman P, Deliliers GL, Platzbecker U, et al. Cidofovir for cytomegalovirus infection and disease in allogeneic stem cell transplant recipients. The Infectious Diseases Working Party of the European Group for Blood and Marrow Transplantation. Blood 2001;97:388–392.
- 160.↑
Platzbecker U, Bandt D, Thiede C, et al. Successful preemptive cidofovir treatment for CMV antigenemia after dose-reduced conditioning and allogeneic blood stem cell transplantation. Transplantation 2001;71:880–885.
- 161.↑
Papanicolaou GA, Silveira FP, Langston AA, et al. Maribavir for refractory or resistant cytomegalovirus infections in hematopoietic-cell or solid-organ transplant recipients: a randomized, dose-ranging, double-blind, phase 2 study. Clin Infect Dis 2019;68:1255–1264.
- 162.↑
Avery RK, Alain S, Alexander BD, et al. Maribavir for refractory cytomegalovirus infections with or without resistance post-transplant: results from a phase 3 randomized clinical trial. Clin Infect Dis 2022;75:690–701.
- 163.↑
Papanicolaou GA, Avery RK, Cordonnier C, et al. Treatment for first cytomegalovirus infection post-hematopoietic cell transplant in the AURORA trial: a multicenter, double-blind, randomized, phase 3 trial comparing maribavir with valganciclovir. Clin Infect Dis 2024;78:562–572.
- 164.↑
Laurenti L, Piccioni P, Cattani P, et al. Cytomegalovirus reactivation during alemtuzumab therapy for chronic lymphocytic leukemia: incidence and treatment with oral ganciclovir. Haematologica 2004;89:1248–1252.
- 165.↑
Moreton P, Kennedy B, Lucas G, et al. Eradication of minimal residual disease in B-cell chronic lymphocytic leukemia after alemtuzumab therapy is associated with prolonged survival. J Clin Oncol 2005;23:2971–2979.
- 166.↑
Nguyen DD, Cao TM, Dugan K, et al. Cytomegalovirus viremia during Campath-1H therapy for relapsed and refractory chronic lymphocytic leukemia and prolymphocytic leukemia. Clin Lymphoma 2002;3:105–110.
- 167.↑
O’Brien S, Ravandi F, Riehl T, et al. Valganciclovir prevents cytomegalovirus reactivation in patients receiving alemtuzumab-based therapy. Blood 2008;111:1816–1819.
- 168.↑
Ljungman P, de la Camara R, Robin C, et al. Guidelines for the management of cytomegalovirus infection in patients with haematological malignancies and after stem cell transplantation from the 2017 European Conference on Infections in Leukaemia (ECIL 7). Lancet Infect Dis 2019;19:e260–272.
- 169.↑
Tay KH, Slavin MA, Thursky KA, et al. Cytomegalovirus DNAemia and disease: current-era epidemiology, clinical characteristics and outcomes in cancer patients other than allogeneic haemopoietic transplantation. Intern Med J 2022;52:1759–1767.
- 170.↑
Montillo M, Schinkoethe T, Elter T. Eradication of minimal residual disease with alemtuzumab in B-cell chronic lymphocytic leukemia (B-CLL) patients: the need for a standard method of detection and the potential impact of bone marrow clearance on disease outcome. Cancer Invest 2005;23:488–496.
- 171.↑
O’Brien SM, Keating MJ, Mocarski ES. Updated guidelines on the management of cytomegalovirus reactivation in patients with chronic lymphocytic leukemia treated with alemtuzumab. Clin Lymphoma Myeloma 2006;7:125–130.
- 172.↑
Russo D, Schmitt M, Pilorge S, et al. Efficacy and safety of extended duration letermovir prophylaxis in recipients of haematopoietic stem-cell transplantation at risk of cytomegalovirus infection: a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Haematol 2024;11:e127–135.
- 173.↑
Nguyen Q, Champlin R, Giralt S, et al. Late cytomegalovirus pneumonia in adult allogeneic blood and marrow transplant recipients. Clin Infect Dis 1999;28:618–623.
- 174.↑
Moretti S, Zikos P, Van Lint MT, et al. Forscarnet vs ganciclovir for cytomegalovirus (CMV) antigenemia after allogeneic hemopoietic stem cell transplantation (HSCT): a randomised study. Bone Marrow Transplant 1998;22:175–180.
- 175.↑
Liang R. How I treat and monitor viral hepatitis B infection in patients receiving intensive immunosuppressive therapies or undergoing hematopoietic stem cell transplantation. Blood 2009;113:3147–3153.
- 176.↑
Brechot C, Degos F, Lugassy C, et al. Hepatitis B virus DNA in patients with chronic liver disease and negative tests for hepatitis B surface antigen. N Engl J Med 1985;312:270–276.
- 177.↑
Dhedin N, Douvin C, Kuentz M, et al. Reverse seroconversion of hepatitis B after allogeneic bone marrow transplantation: a retrospective study of 37 patients with pretransplant anti-HBs and anti-HBc. Transplantation 1998;66:616–619.
- 178.↑
Borentain P, Colson P, Coso D, et al. Clinical and virological factors associated with hepatitis B virus reactivation in HBsAg-negative and anti-HBc antibodies-positive patients undergoing chemotherapy and/or autologous stem cell transplantation for cancer. J Viral Hepat 2010;17:807–815.
- 179.↑
Evens AM, Jovanovic BD, Su YC, et al. Rituximab-associated hepatitis B virus (HBV) reactivation in lymphoproliferative diseases: meta-analysis and examination of FDA safety reports. Ann Oncol 2011;22:1170–1180.
- 180.↑
Fukushima N, Mizuta T, Tanaka M, et al. Retrospective and prospective studies of hepatitis B virus reactivation in malignant lymphoma with occult HBV carrier. Ann Oncol 2009;20:2013–2017.
- 181.↑
Ji D, Cao J, Hong X, et al. Low incidence of hepatitis B virus reactivation during chemotherapy among diffuse large B-cell lymphoma patients who are HBsAg-negative/HBcAb-positive: a multicenter retrospective study. Eur J Haematol 2010;85:243–250.
- 182.↑
Koo YX, Tan DS, Tan IB, et al. Hepatitis B virus reactivation and role of antiviral prophylaxis in lymphoma patients with past hepatitis B virus infection who are receiving chemoimmunotherapy. Cancer 2010;116:115–121.
- 183.↑
Lalazar G, Rund D, Shouval D. Screening, prevention and treatment of viral hepatitis B reactivation in patients with haematological malignancies. Br J Haematol 2007;136:699–712.
- 184.↑
Lau GKK, He ML, Fong DYT, et al. Preemptive use of lamivudine reduces hepatitis B exacerbation after allogeneic hematopoietic cell transplantation. Hepatology 2002;36:702–709.
- 185.↑
Matsue K, Kimura S, Takanashi Y, et al. Reactivation of hepatitis B virus after rituximab-containing treatment in patients with CD20-positive B-cell lymphoma. Cancer 2010;116:4769–4776.
- 186.↑
Yeo W, Chan PK, Zhong S, et al. Frequency of hepatitis B virus reactivation in cancer patients undergoing cytotoxic chemotherapy: a prospective study of 626 patients with identification of risk factors. J Med Virol 2000;62:299–307.
- 187.↑
Yeo W, Chan TC, Leung NW, et al. Hepatitis B virus reactivation in lymphoma patients with prior resolved hepatitis B undergoing anticancer therapy with or without rituximab. J Clin Oncol 2009;27:605–611.
- 188.↑
European Association for the Study of the Liver. EASL 2017 Clinical Practice Guidelines on the management of hepatitis B virus infection. J Hepatol 2017;67:370–398.
- 189.↑
Liang RH, Lok AS, Lai CL, et al. Hepatitis B infection in patients with lymphomas. Hematol Oncol 1990;8:261–270.