Next Article in Journal
The Spectrum of Antimicrobial Activity of Cyadox against Pathogens Collected from Pigs, Chicken, and Fish in China
Next Article in Special Issue
Measurement of the Intestinal pH in Mice under Various Conditions Reveals Alkalization Induced by Antibiotics
Previous Article in Journal
Microbiological and Clinical Aspects of Actinomyces Infections: What Have We Learned?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Antimicrobial Prophylaxis and Modifications of the Gut Microbiota in Children with Cancer

Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, 43126 Parma, Italy
*
Author to whom correspondence should be addressed.
Submission received: 31 December 2020 / Revised: 27 January 2021 / Accepted: 29 January 2021 / Published: 3 February 2021

Abstract

:
In children with cancer, chemotherapy can produce cytotoxic effects, resulting in immunosuppression and an augmented risk of febrile neutropenia and bloodstream infections. This has led to widespread use of antibiotic prophylaxis which, combined with intensive chemotherapy treatment, could have a long-term effect on the gastrointestinal microbiome. In this review, we aimed to analyze the current literature about the widespread use of antibiotic prophylaxis in children experiencing infectious complications induced by chemotherapy and its effects on the gut microbiome. Our review of the literature shows that antimicrobial prophylaxis in children with cancer is still a trending topic and, at the moment, there are not enough data to define universal guidelines. Children with cancer experience long and painful medical treatments and side effects, which are associated with great economic and social burdens, important psychological consequences, and dysbiosis induced by antibiotics and also by chemotherapy. Considering the importance of a healthy gut microbiota, studies are needed to understand the impact of dysbiosis in response to therapy in these children and to define how to modulate the microbiome to favor a positive therapeutic outcome.

1. Introduction

Cancer diagnoses in patients younger than 20 years old are rare, representing only approximately 1% of all new cancer cases every year, and the survival rate increased to 84.8% from 2008 to 2013 in all age groups 0–19 years [1]. The epidemiology of cancer in children differs from that in adulthood: lymphohematopoietic cancers account for approximately 40%, central nervous system (CNS) cancers account for approximately 30%, and embryonal tumors and sarcomas account for approximately 10% of cases. The most frequent cancer in childhood, in particular, is leukemia (acute lymphoblastic leukemia [ALL] and acute myeloid leukemia [AML]), with ALL accounting for 26% of all cancers in children up to 14 years of age and 75% of all pediatric leukemia cases [2]. Children with cancer undergo a treatment protocol that may include chemotherapy, radiotherapy (RT), surgery, or hematopoietic cell transplantation (HCT) alone or in combination depending on the type of cancer, its location, and its stage.
The use of some chemotherapeutics can produce cytotoxic effects, resulting in immunosuppression and an augmented risk of febrile neutropenia (i.e., the total number of blood neutrophils ≤ 500/mm3) and bloodstream infections [2]. This has led to widespread use of antibiotic prophylaxis, which, combined with intensive chemotherapy treatment, could have a long-term effect on the gastrointestinal microbiome [3]. The effects of this alteration have been studied but remain mostly unknown; it has been proven that this change leads to colonization by opportunistic pathogens, impairs the gastrointestinal barrier, increases vulnerability to Clostridium difficile infections [3], and is also linked to the development of a variety of diseases. In this review, we aimed to analyze the current literature about the widespread use of antibiotic prophylaxis in children experiencing infectious complications induced by chemotherapy and its effects on the gut microbiome. A further purpose was to show the current known effects of alterations of the microbiome on children’s responses to chemotherapy and their future health outcomes. Eventually, we wanted to focus briefly on probiotics, their actual use, and their possible more routine application in clinical practice. PubMed was used to search for studies published mainly in the last 15 years using the key words “cancer”, “microbiome” or “microbiota”, “antibiotic prophylaxis”, and “children” or “pediatric”. More than 150 articles were found, but only those written in English were taken into consideration.

2. Overview of Antibiotic Usage in Children with Cancer

2.1. Infectious Complications in Children with Cancer

Infectious complications are among the most common and life-threatening complications; they are associated with significant morbidity and mortality and lead to treatment delays and dose reductions of chemotherapy [4]. Their development in children with cancer mainly occurs during periods of neutropenia [5,6], and is favored by the presence of skin and mucosal damage, central venous catheters, or the use of immunosuppressant drugs [5].
Fever is the first and most frequent symptom of bacterial infection, especially during periods of neutropenia. Castagnola et al. carried out a three-year observational study showing that the highest frequency of neutropenic periods with primary febrile episodes was observed after autologous HCT (58%), during induction treatment for ALL or non-Hodgkin lymphoma (48%), and after allogeneic HCT (44%) [7]. In a study conducted by Gil et al., 92.3% of patients diagnosed with cancer developed infectious complications after high-dose chemotherapy or HCT [8]. A more recent observational study by Zawitkowska et al., focusing on children with newly diagnosed ALL, showed that 53.2% of them had a microbiologically documented bacterial infection during chemotherapy.

2.2. Etiology of Bacterial Infections

The most common infections during periods of neutropenia are bacterial infections, and there has been a clear shift in the type of organisms involved during the past three decades [4]: until the beginning of the 1980s, gram-negative rods (particularly Escherichia coli, Klebsiella spp., and Pseudomonas aeruginosa) were the most frequent causes of bacterial infections in cancer patients [5,6,7,9], but recently, an increase in the frequency of infections caused by gram-positive organisms in these patients occurred [9]. Gram-negative bacteria are the main causes of bloodstream infections, whereas gram-positive bacteria cause mainly central venous catheter (CVC)-related infectious complications. However, infections caused by gram-negative bacteria are associated with higher morbidity and remain the most common cause of mortality during periods of myelosuppression [4,10,11].

3. Antibiotic Prophylaxis

The severity of bacterial infections during periods of neutropenia in children with cancer makes essential the appropriate use of broad-spectrum antibiotics to resolve this kind of complication and to reduce associated morbidity and mortality. Many different approaches have been discussed in recent years, particularly concerning the correct classes of antibiotics to use on these occasions and whether to prevent infectious complications with systemic antibiotic prophylaxis or to treat them when microbiologically documented [12,13]. In the current literature, there is a lack of clinical guidelines concerning the routine use of antibacterial prophylaxis. A recent study by Lehrnbecher et al. reviewed the works on this argument to develop a guideline for the administration of antibiotic prophylaxis in pediatric clinical practice, proving that there are not enough studies supporting the routine use of antibiotics in children with cancer or HCT recipients because the benefits of prophylaxis are balanced by its unknown and potential impacts and resistance [12]. However, antimicrobial prophylaxis for neutropenic patients undergoing cytotoxic therapy reduces mortality, as supported by a meta-analysis published by Gafter-Gvili et al. [13], showing the importance of appropriate administration and selection of the correct class of antibiotic.

3.1. Beta-Lactam Antibiotics

Beta-lactam antibiotics work by inhibiting cell wall biosynthesis in the bacterial organism and are the most widely used group of antibiotics. At first, beta-lactam antibiotics were mainly active only against gram-positive bacteria, yet the recent development of broad-spectrum beta-lactam antibiotics active against various gram-negative organisms has increased their usefulness [5]. The cephalosporin class and amoxicillin/clavulanate have been widely studied and used, as proven by Castagnola et al., who employed prophylaxis with amoxicillin/clavulanate at a dose of 25 mg every 12 h and showed a reduction in febrile episodes [14]. Feng et al. studied the incidence of infection-related fever in children with AML and the outcome of prophylactic use of vancomycin/cefepime or piperacillin/tazobactam versus a control group with no antibiotic prophylaxis: the result was a significant reduction in infection-related fever in those children undergoing antibiotic prophylaxis during periods of chemotherapy-induced neutropenia [15]. However, in the last decade there has been a worldwide increase in multidrug-resistant (MDR) bacteria, making the oral therapy approach with beta-lactams ineffective in many cases [16,17,18,19].

3.2. Fluoroquinolones (FQLs)

FQLs interfere with DNA replication by preventing bacterial DNA from unwinding and duplicating. They are effective against both gram-negative and gram-positive bacteria [5]. FQLs were formerly used in the prophylaxis of adult patients with cancer because of their great efficacy, as proven by two large studies conducted in 2005 [20,21]. Since then, there has been growing interest concerning the routine use of FQLs in antibiotic prophylaxis because they have a broad antibacterial spectrum, good bioavailability and bactericidal activity, are well tolerated, and do not have any myelosuppressive effects. Indeed, a recent review by Lehrnbecher showed that FQL prophylaxis significantly reduced bacteremia, fever, and neutropenia; it was not significantly associated with C. difficile infection, invasive fungal disease, or muscle–skeletal toxicities, but it was significantly associated with more FQL resistance in bacteremia isolates [12]. One of the first works on this topic was performed by Cruciani et al., who studied the effects of norfloxacin compared with that of trimethoprim-sulfamethoxazole (TMP-SMX) in 44 neutropenic children with various malignancies, proving that FQL was superior in preventing febrile episodes, but the mean number of febrile days was similar in the two groups [5,22].
Recent studies focused on ciprofloxacin, whose effects have been widely studied in children with cancer, and levofloxacin, which seems to be the preferred agent if antibacterial prophylaxis is planned [12]. Alexander et al. studied the effects of prophylaxis with levofloxacin at a prophylactic dosage of 10 mg/kg twice daily (in children aged six months to five years) or 10 mg/kg once daily (for children aged more than five years) in 624 patients with AML, with relapsed ALL, or who underwent HCT, proving that among children with acute leukemia receiving intensive chemotherapy, the administration of levofloxacin prophylaxis compared with no prophylaxis resulted in a significant reduction in bacteremia; however, there was no significant reduction in bacteremia for levofloxacin prophylaxis among children undergoing HCT [23]. Ciprofloxacin also has a good efficacy profile, as proven by Laoprasopwattana et al., who compared his effects with a placebo group in 95 patients with lymphoma and ALL and observed a reduction in fever and bacteremia in those receiving FQL [24] during induction but not during the consolidation phase. Widjajanto et al. conducted a similar study in 110 children with ALL who were undergoing induction treatment and compared the effects of prophylaxis with ciprofloxacin with those of placebo; the result was disappointing, with a greater risk of fever and sepsis and increased mortality among those who received ciprofloxacin [25]. The dosage of ciprofloxacin has varied among the studies: Yousef et al. used ciprofloxacin at a prophylactic dosage of 25 mg/kg/day [26], Laoprasopwattana at a dosage of 20 mg/kg/day [24], Choeyprasert at 20–30 mg/kg/day in two divided doses in combination with penicillin sodium V 25–50 mg/kg/day in four doses [27], while Al Omar et al. administered it at a dosage of 10 mg/kg/day divided into two doses every 12 h [28].

3.3. Trimethoprim-Sulfamethoxazole (TMP-SMX)

TMP-SMX, also known as co-trimoxazole, is a combination of two antimicrobial agents that act synergistically against a wide variety of bacteria. The two components, TMP and SMX, work sequentially to inhibit enzyme systems involved in the bacterial synthesis of tetrahydrofolic acid [5]. TMP-SMX prophylaxis is a valid alternative to FQLs that reduces bacteremia and infection-related mortality, but it increases resistance in bacteremia isolates [12]. It has broad-spectrum activity, including gram-positive bacteria, gram-negative bacteria, Nocardia, and Pneumocystis jirovecii. Its efficacy profile was supported by some studies conducted by Goorin et al. [29], which showed fewer fever episodes in children treated with TMP-SMX than in children treated with the placebo [29], and by Kovatch et al., which found a reduction in fever and bacteremia related to its use [30]. However, these positive aspects are balanced by a series of drawbacks, including the development of hypersensitivity, breakthrough infections due to resistant gram-negative and gram-positive pathogens, fungal infections, and C. difficile colitis, as evidenced by Gualtieri et al. [29,30,31]. For the appropriate prophylactic dose, Schroder et al. used it at a dosage of 10–30 mg/kg/day [32], Cullen et al. employed a fixed dosage of 20 mg/kg/day [21], Chastagner et al. a prophylactic dosage of 25 mg/kg/day every two days [33], while Al Omar et al. used TMP-SMX at a dosage of 2.5 mg/kg/day in combination with ciprofloxacin for two consecutive days per week [28].
In Table 1, we report the main studies on antibiotic prophylaxis performed by the authors cited above, who administered different agents at different dosages, showing that there are no official and standard guidelines for antibiotic prophylaxis in children with cancer. As demonstrated, antibiotic prophylaxis in these patients may have a fundamental role in preventing and reducing infection-related mortality in children with a high risk of febrile neutropenia and infectious complications, but more studies are required to define the best treatment, to ensure a correct balance of positive and adverse effects, to show the feasibility of all the different molecules available, and to establish a widely accepted guideline [28,34].

4. Dysbiosis and Cancer in Children

The human gut microbiota consists of thousands of different species, each sharing a symbiotic relationship with the human host [35]. It appears that each person has his or her own individual suite of microbial strains [36,37]; this bacterial “panel” is acquired in the early stages of life [38] and can remain unaltered or go through different transitions [39,40]. Dysbiosis, an imbalance in microbial taxa, has been implicated in various aspects of human health. One of the main factors that can disrupt a healthy gut microbiota is antibiotic exposure [41]: even short-term antibiotic usage has been linked to the loss of certain taxa (i.e., a reduction in the diversity of Firmicutes and Bacteroidetes, growth of the family Enterobacteriaceae) [42,43], impairment of the gastrointestinal barrier [44], and increased vulnerability to Clostridium difficile [45] and other vancomycin-resistant Enterococci [46]. Even within days of the administration of antibiotics, a significant upregulation of resistance genes has been shown [41]. In children, it has been shown that a disrupted intestinal microbiota is linked to the development of a range of diseases (such as inflammatory bowel diseases [47], Kawasaki syndrome [48], asthma [49], autism [50,51,52]), and most importantly, dysbiosis itself plays an important role in cancerogenesis, alongside environmental and genetic factors [53,54]. Since antibiotics are largely used to prevent infectious complications in neutropenic children undergoing chemotherapy [55] and represent some of the most frequently prescribed drugs in pediatric patients [56,57], a better understanding of the dynamic interaction between gut microbiota dysbiosis and cancer pathogenesis may be helpful to improve the standard of care in children with cancer. On the other hand, chemotherapy itself can further worsen the microbiota composition.

4.1. Gut Microbiome Alterations in Children with Acute Leukemia

Acute lymphoblastic (ALL) and myeloid (AML) leukemia are the most frequent childhood blood malignancies [58]; therefore, alterations in the microbiota at the time of diagnosis and during chemotherapy have been analyzed mostly in children with leukemia. It has been shown that in both ALL and AML patients, the amount of bacterial flora is reduced in comparison with that in healthy controls: specifically, a significant decrease in Bifidobacteria, Lactobacillus, and E. coli has been found in children with leukemia [59,60]. Rajagopala et al. [3] collected stool samples from 51 children, both pediatric and adolescent patients with ALL and healthy siblings, to identify possible variations in the gut microbiota before and during chemotherapeutic treatment; Bacteroides, Prevotella, and Faecalibacterium were found in both groups at the time of diagnosis, but the overall microbial diversity of the ALL group was lower than that of the healthy sibling group (p < 0.01). Microbial diversity was not significantly different at the end of chemotherapy but increased significantly at different visits after the end of chemotherapy (p < 0.01). Van Vliet et al. [60] collected stool samples from pediatric patients with AML who were undergoing chemotherapy and receiving antimicrobial prophylaxis against gram-negative bacteria and fungi (oral colistin, neomycin, and amphotericin B with ciprofloxacin and itraconazole), viridans group streptococci (oral pheneticillin), and Pneumocystis jirovecii (oral cotrimoxazole); they found a large decrease in anaerobic bacteria, compensated by an increase in potential pathogenic Enterococci. These studies show that antibiotic-induced dysbiosis could be dangerous in critically ill patients but is not solely responsible for such disruption of the gut microbiota; the interaction between chemotherapy and different factors (genetics, diet) [61] in shaping dysbiosis may be the key to obtaining a better understanding of the dynamics underlying cancerogenesis.

4.2. Microbiota and the Patient’s Outcome: Infections, Adverse Effects, and Response to Treatment

Infections are a common and dangerous complication in children with cancer [4,62]. In addition to bloodstream infections (generally caused by gram-negative bacteria [63]), CVC-related infectious complications are also a relevant cause of morbidity. It has been estimated that 14–51% of CVCs implanted in children with malignancies may be complicated by bacteremia [64]. Galloway et al. [65] collected buccal and fecal specimens twice weekly from 34 AML patients undergoing induction chemotherapy to define a possible link between antimicrobial composition and infection outcomes. They observed that a low baseline α-diversity in stool samples was associated with the development of infections during chemotherapy; patients before leukemia treatment had a wide range of α-diversity, indicating that the factors causing this variability are numerous and may include previous antibiotic exposure, diet, and genetics. Interestingly, it was noted that higher antibiotic exposure during induction chemotherapy was significantly associated with an increased risk of infection after treatment.
While early administration of broad-spectrum antibiotics has been shown to reduce mortality, there is some evidence that prophylactic antibiotic regimens may alter the human microbiota, thus inducing antibacterial resistance and the proliferation of MDR bacteria [62]. Two similar nonrandomized studies [66,67] evaluated blood cultures from pediatric cancer patients presenting with febrile neutropenia; in both studies, some patients underwent antibiotic prophylaxis with ciprofloxacin (a small portion of the patients in one study were not treated, but interestingly, all adult patients from the same center received FQL prophylaxis). The results were similar: in children receiving ciprofloxacin, gram-negative bacteria found in blood cultures had increased rates of resistance towards multiple antibiotics. Even children who did not receive ciprofloxacin but were hospitalized in the same institution where adults underwent FQL prophylaxis had this pattern of resistance, suggesting that factors in addition to antibiotic exposure must be involved. TMP-SMX prophylaxis does not seem to cause these alterations, but fewer data are available [68].
A frequent gastrointestinal complication in children with cancer is diarrhea, which can seriously impair the patient’s quality of life as it leads to malnutrition and fatigue [69,70]. While chemotherapy and radiation are proven risk factors for mucositis and therefore acute diarrhea [71,72], the gut microbiota has recently been proposed as a potential factor in the onset of this complication [73,74,75,76,77,78,79,80,81,82,83]. In a study by Manichanh et al. [73], fecal samples from 10 patients undergoing radiotherapy for abdominal tumors were collected at different times during treatment; six of them suffered from diarrhea and showed a progressive reduction in microbial diversity, while the other four had a “stable” gut microbiota compared with that of a healthy control group. Similar results have been seen in post-chemotherapy diarrhea, with the gut microbiota changing towards Escherichia coli and Staphylococcus domination with decreases in lactobacilli, Bifidobacteria, Bacteroides, and Enterococci [74]. Even if very few data are available concerning the role of microbiota in pediatric patients with cancer presenting diarrhea [60], probiotics are now taken into consideration in different studies as a possible tool to prevent chemotherapy- and radiotherapy-related mucositis [75]. Finally, some authors have suggested a potential interaction between the gut microbiota and anticancer drugs, thus influencing antineoplastic treatment. In murine models, there is increasing evidence of this particular interaction. Iida et al. [76] showed that mice with subcutaneous EL4 lymphoma who received an antibiotic cocktail prior to oxaliplatin treatment had a significant reduction in cancer regression and survivability when compared with an “antibiotic-free” control group. These findings are replicated in another interesting study by Gui et al. [77], in which animals with lung cancer receiving a combination of cisplatin and antibiotics had a decreased survival rate in comparison to mice treated with cisplatin alone. On the other hand, the administration of cisplatin and Lactobacillus bacteria improved the response to therapy.
The mechanism that may underlie the relationship between gut microbiota and chemotherapy is summarized in Table 2. Several studies on this topic are still ongoing, and with novel drugs coming out every year and the growing usage of monoclonal antibodies in different stages of cancer, many more relationships are yet to be discovered.

4.3. The Gut Microbiota Plays a Key Role as Trigger for Gut Graft Versus Host Disease in the Context of Hematopoietic Stem Cell Transplantation (HCT)

When cancer or cancer treatments destroy the stem cells, HCT may be the best treatment option. The possible link between gut microbial disruption and HCT has been widely studied, and it is currently clear that dysbiosis is one of the many actors in the genesis of graft versus host disease (GvHD), a potentially lethal complication of HCT.
The first studies on this topic were performed in the 1970s [84,85], when analyses in mice showed a lower incidence of gut GvhD in germ-free animals. Since then, much progress has been made, especially thanks to the use of next-generation sequencing. The work of Holler et al. [86] is particularly relevant: stool specimens from 31 patients receiving HCT were analyzed before and after the procedure through next-generation sequencing. Before HCT, all patients showed a similar balance of commensal bacteria. After HCT, a shift towards Enterococcus domination was observed, which was particularly evident in patients under antibiotic treatment who developed gut GvHD. This was one of the first studies to provide evidence that alteration of the microbiota by HCT-related procedures (such as antibiotic exposure) is directly linked to GvHD, as confirmed in more recent analyses [87,88,89,90]. Italian researchers have been particularly active in defining how microbiota dysbiosis impacts pediatric patients undergoing HCT. A first work published in 2015 [91] showed that changes seen in adult patients also occurred in children. Pre- and post-HCT samples were collected from 26 pediatric patients undergoing HCT and analyzed with next-generation sequencing to define possible variations in microbiota structure. Interestingly, children who suffered from GvHD had specific gut microbiota signatures after HCT: overgrowth of Enterococcus and Clostridiales, decreases in Faecalibacterium and Ruminococcus, and a substantial decrease in Bacteroides, a group of bacteria associated with the production of propionate, which might have the ability to activate T helper type 2 cells [92,93]. Moreover, Biagi et al. [94] tried to determine with more accuracy if these differences in the gut microbiota population were already established before HCT. Stool specimens from 36 pediatric patients undergoing HCT were collected before transplantation, at the time of engraftment and after 30 days. Next-generation sequencing showed that children who suffered from GvHD had dysbiosis before HCT (a lower number of Blautia, a higher abundance of Fusobacterium, and decreased overall diversity), thus implying that the gut microbiota may be a tool to stratify GvhD risk. Finally, in a recent study by d’Amico et al. [95], the gut resistome, meaning the pattern of antibiotic resistance derived from the gut microbiome, was taken into consideration. A comparison was made between 8 pediatric patients undergoing HCT and 10 healthy adults. Interestingly, an amplification of the resistome was noted, especially in four patients who developed acute GvHD, even though antibiotics were not routinely administered in the course of transplantation (aminoglycosides, macrolides, and tetracyclines). All this evidence suggests that microbiota and HCT outcomes are deeply intertwined, even in pediatric patients, in whom the microbiota is often still taking shape and in some sense immature. Prebiotics, probiotics, or fecal microbiota transplantations might be fundamental tools for preventing major damage associated with this procedure.

4.4. Efficacy of Probiotics in Children with Cancer

Probiotics are live microorganisms, which, when administered in adequate amounts, confer a health benefit on the host [96,97]. Probiotic supplementation has been widely studied in children presenting various health conditions [98,99,100,101,102,103,104], whereas there have been very few studies regarding gut microbiota dysbiosis in pediatric patients with cancer. The first one was published in 1980 [105] and was also one of the first works that examined the concept of intestinal decontamination, that is, a prophylactic strategy that consists of the administration of antimicrobials with limited anaerobicidal activity in order to reduce the burden of aerobic gram-negative bacteria and/or yeast in the intestinal tract and so prevent infections caused by these organisms. In a group of 68 children with leukemia and solid tumors, 35 neutropenic episodes in 33 children were treated with framycetin, colymycin, nystatin, and metronidazole, while the other 35 episodes in the remaining 35 children were cured with TMP-SMX and Lactobacillus preparations [105]. Even if there was no significant difference in the incidence of infections during the period of neutropenia, the second group had better tolerance to the medication, so the authors concluded that a TMP-SMX and Lactobacillus preparation may improve quality of life in neutropenic children and is also relatively inexpensive [105]. Finally, Wada et al. [106] demonstrated that children with cancer receiving Bifidobacterium breve strain Yakult had decreased levels of Enterobacteriaceae in their stool and, more importantly, children assigned to the probiotic group had fewer febrile episodes (0.5 ± 0.62, 95% confidence interval [CI]: 0.21–0.79 and 1.06 ± 1.80, 95% CI: 0.19–1.93) than did children assigned to the placebo group (0.95 ± 0.79, 95% CI: 0.62–1.28 and 3.00 ± 3.84, 95% CI: 1.39–4.61), thus indicating lower usage of antibiotics. Nevertheless, there is much more work to be done, especially since some researchers have shown concerns about the possibility of administering living microorganisms to patients with compromised immunity and gut defenses after reports of sepsis caused by probiotic pathogens [107,108,109]. Further studies are needed to define the feasibility and correct duration of probiotic administration, when it should be suspended, and if it has a real impact in preventing major gut comorbidities in children with solid or hematological malignancies.

5. Conclusions

Our review of the literature shows that antimicrobial prophylaxis in children with cancer is still a trending topic and, at the moment, there are not enough data to define universal guidelines. The reasons for this challenge are numerous. First, a child is an “evolving” being: the immune system changes rapidly in pediatric age and drastically over time, as does its response to certain drugs. Table 3 summarizes the effects of different antibiotics on intestinal microbiota. Finding a standard prophylaxis for children of all ages might be hard, especially because most studies take into account the use of FQLs, drugs not registered for children. Moreover, different factors come into play when discussing antibiotic prophylaxis. Not only is the right dosage fundamental but also the feasibility of the prophylaxis itself is an element that needs to be taken into consideration in future studies; when we discuss pediatric patients, the number of administrations and the palatability [110] of a drug are key points, as these are among the major factors affecting pediatric compliance.
In addition to defining the optimal antibiotics needed for prophylaxis, there is still uncertainty about patients who do not need treatment. With antimicrobial resistance being a global health security threat [111], more accurate antibiotic prescriptions should be implemented, especially in patients with cancer, to prevent serious complications and, as we have described in our review, disruption of the intestinal microbiota, an emerging player in maintaining human health. Many researchers around the world have focused on obtaining a better understanding of the influence of the gut microbiota in different health conditions, but only in recent years has the idea of modulating the microbiome to modify the outcome of hematological/oncological diseases started to attract attention. Novel strategies for preventing antibiotic-mediated dysbiosis have been proposed: some authors have suggested the use of beta lactamase enzymes for the degradation of antibiotics in the gut [112], while others proposed that changing nutritional strategies in oncological patients, such as promoting enteral nutrition rather than parenteral nutrition or the administration of specific molecules (probiotics), might be the key to preventing gut complications [113]. Another approach for the prevention of dysbiosis is the possibility of intervening in the nutritional state of the patients. Children undergoing chemotherapy exhibit several variations in their body composition, with a higher percentage of fat mass and a lower body cell index [114] or a general reduction in bone density [115]. Probiotics may be helpful in these patients since some of them can regulate protein absorption and utilization [116,117].
In conclusion, children with cancer experience long and painful medical treatments and side effects, which are associated with great economic and social burdens, important psychological consequences, and dysbiosis induced by antibiotics and also by chemotherapy [118,119,120]. Considering that a healthy gut microbiota keeps the gut epithelium intact, studies are needed to understand the impact of dysbiosis in response to therapy in children with cancer and to define how to modulate the microbiome to favor a positive outcome.

Author Contributions

G.B. and R.D.S. wrote the first draft of the manuscript; A.A. performed the literature review; S.E. supervised the project, gave a scientific contribution, and critically revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

Department of Medicine and Surgery, University of Parma, Parma, Italy.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not needed for a review article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kliegman, R.; Stanton, B.; St. Geme, J.W.; Schor, N.F.; Behrman, R.E.; Nelson, W.E. Nelson Textbook of Pediatrics; Elsevier: Philadelphia, PA, USA, 2016. [Google Scholar]
  2. Rajagopala, S.V.; Singh, H.; Yu, Y.; Zabokrtsky, K.B.; Torralba, M.G.; Moncera, K.J.; Frank, B.; Pieper, R.; Sender, L.; Nelson, K.E. Persistent gut microbial dysbiosis in children with acute lymphoblastic leukemia (ALL) during chemotherapy. Microbial. Ecol. 2020, 79, 1034–1043. [Google Scholar] [CrossRef] [PubMed]
  3. Rajagopala, S.V.; Yooseph, S.; Harkins, D.M.; Moncera, K.J.; Zabokrtsky, K.B.; Torralba, M.G.; Tovchigrechko, A.; Highlander, S.K.; Pieper, R.; Sender, L.; et al. Gastrointestinal microbial populations can distinguish pediatric and adolescent acute lymphoblastic Leukemia (ALL) at the time of disease diagnosis. BMC Genom. 2016, 17, 635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Sulis, M.L.; Blonquist, T.M.; Stevenson, K.E.; Hunt, S.K.; Kay-Green, S.; Athale, U.H.; Clavell, L.A.; Cole, P.D.; Kelly, K.M.; Laverdiere, C.; et al. Effectiveness of antibacterial prophylaxis during induction chemotherapy in children with acute lymphoblastic leukemia. Pediatr. Blood Cancer 2018, 65, e26952. [Google Scholar] [CrossRef] [PubMed]
  5. Cecinati, V.; Principi, N.; Brescia, L.; Esposito, S. Antibiotic prophylaxis in children with cancer or who have undergone hematopoietic cell transplantation. Eur. J. Clin. Microbiol. Infect. Dis. 2014, 33, 1–6. [Google Scholar] [CrossRef] [PubMed]
  6. Vento, S.; Cainelli, F. Infections in patients with cancer undergoing chemotherapy: Aetiology, prevention, and treatment. Lancet Oncol. 2003, 4, 595–604. [Google Scholar] [CrossRef]
  7. Castagnola, E.; Fontana, V.; Caviglia, I.; Caruso, S.; Faraci, M.; Fioredda, F.; Garrè, M.L.; Moroni, C.; Conte, M.; Losurdo, G.; et al. A prospective study on the epidemiology of febrile episodes during chemotherapy-induced neutropenia in children with cancer or after hemopoietic stem cell transplantation. Clin. Infect. Dis. 2007, 45, 1296–1304. [Google Scholar] [CrossRef] [PubMed]
  8. Gil, L.; Styczynski, J.; Komarnicki, M. Infectious complication in 314 patients after high-dose therapy and autologous hematopoietic stem cell transplantation: Risk factors analysis and outcome. Infection 2007, 35, 421–427. [Google Scholar] [CrossRef]
  9. Phillips, R.; Hancock, B.; Graham, J.; Bromham, N.; Jin, H.; Berendse, S. Prevention and management of neutropenic sepsis in patients with cancer: Summary of NICE guidance. BMJ 2012, 345, e5368. [Google Scholar] [CrossRef] [Green Version]
  10. Moon, S.; Williams, S.; Cullen, M. Role of prophylactic antibiotics in the prevention of infections after chemotherapy: A literature review. Support Cancer Ther. 2006, 3, 207–216. [Google Scholar] [CrossRef]
  11. Freifeld, A.G.; Bow, E.J.; Sepkowitz, K.A.; Boeckh, M.J.; Ito, J.I.; Mullen, C.A.; Raad, I.I.; Rolston, K.V.; Young, J.A.; Wingard, J.R. Infectious disease society of America. 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–e93. [Google Scholar] [CrossRef] [Green Version]
  12. Lehrnbecher, T.; Fisher, B.T.; Phillips, B.; Alexander, S.; Ammann, R.A.; Beauchemin, M.; Carlesse, F.; Castagnola, E.; Davis, B.L.; Dupuis, L.L.; et al. Guideline for antibacterial prophylaxis administration in pediatric cancer and hematopoietic stem cell transplantation. Clin. Infect. Dis. 2020, 71, 226–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Gafter-Gvili, A.; Fraser, A.; Paul, M.; Leibovici, L. Meta-analysis: Antibiotic prophylaxis reduces mortality in neutropenic patients. Ann. Int. Med. 2005, 142, 979–995. [Google Scholar] [CrossRef] [PubMed]
  14. Castagnola, E.; Boni, L.; Giacchino, M.; Cesaro, S.; De Sio, L.; Garaventa, A.; Zanazzo, G.; Biddau, P.; Rossi, M.R.; Schettini, F.; et al. Infectious diseases study group of the Italian association of pediatric hematology and oncology. A multicenter, randomized, double blind placebo-controlled trial of amoxicillin/clavulanate for the prophylaxis of fever and infection in neutropenic children with cancer. Pediatr. Infect. Dis. J. 2003, 22, 359–365. [Google Scholar] [CrossRef] [PubMed]
  15. Sezgin, G.; Acipayam, C.; Ozkan, A.; Bayram, I.; Tanyeli, A. Meropenem versus piperacillin-tazobactam as empiric therapy for febrile neutropenia in pediatric oncology patients. Asian Pacific. J. Canc. Prevent 2014, 15, 4549–4553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Ichikawa, M.; Suzuki, D.; Ohshima, J.; Cho, Y.; Kaneda, M.; Iguchi, A.; Ariga, T. Piperacillin/tazobactam versus cefozopran for the empirical treatment of pediatric cancer patients with febrile neutropenia. Pediatr. Blood Canc. 2011, 57, 1159–1162. [Google Scholar] [CrossRef]
  17. Feng, X.; Ruan, Y.; He, Y.; Zhang, Y.; Wu, X.; Liu, H.; Liu, X.; He, L.; Li, C. Prophylactic first-line antibiotics reduce infectious fever and shorten hospital stay during chemotherapy-induced agranulocytosis in childhood acute myeloid leukemia. Acta Haematol. 2014, 132, 112–117. [Google Scholar] [CrossRef]
  18. Sano, H.; Kobayashi, R.; Suzuki, D.; Kishimoto, K.; Yasuda, K.; Kobayashi, K. Comparison between piperacillin/tazobactam and cefepime monotherapies as an empirical therapy for febrile neutropenia in children with hematological and malignant disorders: A prospective, randomized study. Pediatr. Blood Cancer 2015, 62, 356–358. [Google Scholar] [CrossRef]
  19. Gustinetti, G.; Mikulska, M. Bloodstream infections in neutropenic cancer patients: A practical update. Virulence 2016, 7, 280–297. [Google Scholar] [CrossRef]
  20. Bucaneve, G.; Micozzi, A.; Menichetti, F.; Martino, P.; Dionisi, M.S.; Martinelli, G.; Allione, B.; D’Antonio, D.; Buelli, M.; Nosari, A.M.; et al. Gruppo Italiano malattie ematologiche dell’adulto (gimema) infection program. Levofloxacin to prevent bacterial infection in patients with cancer and neutropenia. N. Engl. J. Med. 2005, 353, 977–987. [Google Scholar] [CrossRef] [Green Version]
  21. Cullen, M.; Steven, N.; Billingham, L.; Gaunt, C.; Hastings, M.; Simmonds, P.; Stuart, N.; Rea, D.; Bower, M.; Fernando, I.; et al. Simple investigation in neutropenic individuals of the frequency of infection after chemotherapy +/− antibiotic in a number of tumours (SIGNIFICANT) trial group. Antibacterial prophylaxis after chemotherapy for solid tumors and lymphomas. N. Engl. J. Med. 2005, 353, 988–998. [Google Scholar] [CrossRef] [Green Version]
  22. Cruciani, M.; Concia, E.; Navarra, A.; Perversi, L.; Bonetti, F.; Aricò, M.; Nespoli, L. Prophylactic co-trimoxazole versus norfloxacin in neutropenic children--perspective randomized study. Infection 1989, 17, 65–69. [Google Scholar] [CrossRef]
  23. Alexander, S.; Fisher, B.T.; Gaur, A.H.; Dvorak, C.C.; Villa Luna, D.; Dang, H.; Chen, L.; Green, M.; Nieder, M.L.; Fisher, B.; et al. Children’s Oncology Group. Effect of levofloxacin prophylaxis on bacteremia in children with acute leukemia or undergoing hematopoietic stem cell transplantation: A randomized clinical TRIAL. JAMA 2018, 320, 995–1004. [Google Scholar] [CrossRef] [Green Version]
  24. Laoprasopwattana, K.; Khwanna, T.; Suwankeeree, P.; Sujjanunt, T.; Tunyapanit, W.; Chelae, S. Ciprofloxacin reduces occurrence of fever in children with acute leukemia who develop neutropenia during chemotherapy. Pediatr. Infect. Dis. J. 2013, 32, e94–e98. [Google Scholar] [CrossRef]
  25. Widjajanto, P.H.; Sumadiono, S.; Cloos, J.; Purwanto, I.; Sutaryo, S.; Veerman, A.J. Randomized double blind trial of ciprofloxacin prophylaxis during induction treatment in childhood acute lymphoblastic leukemia in the WK-ALL protocol in Indonesia. J. Blood Med. 2013, 4, 1–9. [Google Scholar] [CrossRef] [Green Version]
  26. Yousef, A.A.; Fryer, C.J.; Chedid, F.D.; Abbas, A.A.; Felimban, S.K.; Khattab, T.M. A pilot study of prophylactic ciprofloxacin during delayed intensification in children with acute lymphoblastic leukemia. Pediatr. Blood Canc. 2004, 43, 637–643. [Google Scholar] [CrossRef] [PubMed]
  27. Choeyprasert, W.; Hongeng, S.; Anurathapan, U.; Pakakasama, S. Bacteremia during neutropenic episodes in children undergoing hematopoietic stem cell transplantation with ciprofloxacin and penicillin prophylaxis. Int. J. Hematol. 2017, 105, 213–220. [Google Scholar] [CrossRef] [PubMed]
  28. Al Omar, S.; Anabtawi, N.; Al Qasem, W.; Rihani, R. Bacterial infections in children with acute myeloid leukemia receiving ciprofloxacin prophylaxis. J. Pediatr. Hematol. Oncol. 2017, 39, e131–e135. [Google Scholar] [CrossRef] [PubMed]
  29. Goorin, A.M.; Hershey, B.J.; Levin, M.J.; Siber, G.R.; Gelber, R.D.; Flynn, K.; Lew, M.; Beckett, K.; Blanding, P.; Sallan, S.E. Use of trimethoprim-sulfamethoxazole to prevent bacterial infections in children with acute lymphoblastic leukemia. Pediatr. Infect. Dis. 1985, 4, 265–269. [Google Scholar] [CrossRef] [PubMed]
  30. Kovatch, A.L.; Wald, E.R.; Albo, V.C.; Prin, W.; Orlando, S.J.; Wollman, M.R.; Phebus, C.K.; Shapiro, E.D. Oral trimethoprim/sulfamethoxazole for prevention of bacterial infection during the induction phase of cancer chemotherapy in children. Pediatrics 1985, 76, 754–760. [Google Scholar] [PubMed]
  31. Gualtieri, R.J.; Donowitz, G.R.; Kaiser, D.L.; Hess, C.E.; Sande, M.A. Double-blind randomized study of prophylactic trimethoprim/sulfamethoxazole in granulocytopenic patients with hematologic malignancies. Am. J. Med. 1983, 74, 934–940. [Google Scholar] [CrossRef]
  32. Schrøder, H.; Agger, K.E.; Rosthøj, S.; Carlsen, N.T.; Schmiegelow, K. Antibacterial prophylaxis with trimethoprim-sulfamethoxazole during induction treatment for acute lymphoblastic leukemia. Danish. Med. Bull. 2001, 48, 275–277. [Google Scholar] [PubMed]
  33. Chastagner, P.; Michel, D.; Contet, A.; Lozniewski, A.; Hadou, T.; Schmitt, C.; Phulpin, A.; Fouyssac, F.; Mansuy, L. Effectiveness of antibacterial prophylaxis in children with acute leukemia: A report from a single institution over a 20-year period. Arch. Pediatr. 2018, 25, 464–468. [Google Scholar] [CrossRef] [PubMed]
  34. Inaba, H.; Gaur, A.H.; Cao, X.; Flynn, P.M.; Pounds, S.B.; Avutu, V.; Marszal, L.N.; Howard, S.C.; Pui, C.H.; Ribeiro, R.C.; et al. Feasibility, efficacy, and adverse effects of outpatient antibacterial prophylaxis in children with acute myeloid leukemia. Cancer 2014, 120, 1985–1992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Proctor, L.M.; Creasy, H.H.; Fettweis, J.M.; Lloyd-Price, J.; Mahurkar, A.; Zhou, W.; Buck, G.A.; Snyder, M.P.; Strauss, J.F.; Weinstock, G.M.; et al. Integrative HMP (iHMP) research network consortium. The integrative human microbiome project. Nature 2019, 569, 641–648. [Google Scholar] [CrossRef] [Green Version]
  36. Franzosa, E.A.; Huang, K.; Meadow, J.F.; Gevers, D.; Lemon, K.P.; Bohannan, B.J.; Huttenhower, C. Identifying personal microbiomes using metagenomic codes. Proc. Natl. Acad. Sci. USA 2015, 112, E2930–E2938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Byrd, A.L.; Segre, J.A. Elucidating microbial codes to distinguish individuals. Proc. Natl. Acad. Sci. USA 2015, 112, 6778–6779. [Google Scholar] [CrossRef] [Green Version]
  38. Koenig, J.E.; Spor, A.; Scalfone, N.; Fricker, A.D.; Stombaugh, J.; Knight, R.; Angenent, L.T.; Ley, R.E. Succession of microbial consortia in the developing infant gut microbiome. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. 1), 4578–4585. [Google Scholar] [CrossRef] [Green Version]
  39. Pasolli, E.; Asnicar, F.; Manara, S.; Zolfo, M.; Karcher, N.; Armanini, F.; Beghini, F.; Manghi, P.; Tett, A.; Ghensi, P.; et al. Extensive unexplored human microbiome diversity revealed by over 150,000 genomes from metagenomes spanning age, geography, and lifestyle. Cell 2019, 176, 649–662. [Google Scholar] [CrossRef] [Green Version]
  40. Faith, J.J.; Guruge, J.L.; Charbonneau, M.; Subramanian, S.; Seedorf, H.; Goodman, A.L.; Clemente, J.C.; Knight, R.; Heath, A.C.; Leibel, R.L.; et al. The long-term stability of the human gut microbiota. Science 2013, 341, 1237439. [Google Scholar] [CrossRef] [Green Version]
  41. Dethlefsen, L.; Huse, S.; Sogin, M.L.; Relman, D.A. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol. 2008, 6, e280. [Google Scholar] [CrossRef]
  42. Jernberg, C.; Löfmark, S.; Edlund, C.; Jansson, J.K. Long-term ecological impacts of antibiotic administration on the human intestinal microbiota. ISME J. 2007, 1, 56–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Yin, J.; Prabhakar, M.; Wang, S.; Liao, S.X.; Peng, X.; He, Y.; Chen, Y.R.; Shen, H.F.; Su, J.; Chen, Y.; et al. Different dynamic patterns of β-lactams, quinolones, glycopeptides and macrolides on mouse gut microbial diversity. PLoS ONE 2015, 10, e0126712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Jakobsson, H.E.; Rodríguez-Piñeiro, A.M.; Schütte, A.; Ermund, A.; Boysen, P.; Bemark, M.; Sommer, F.; Bäckhed, F.; Hansson, G.C.; Johansson, M.E. The composition of the gut microbiota shapes the colon mucus barrier. EMBO 2015, 16, 164–177. [Google Scholar] [CrossRef]
  45. Owens, R.C., Jr.; Donskey, C.J.; Gaynes, R.P.; Loo, V.G.; Muto, C.A. Antimicrobial-associated risk factors for Clostridium difficile infection. Clin. Infect. Dis. 2008, 46 (Suppl. 1), S19–S31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Costello, E.K.; Stagaman, K.; Dethlefsen, L.; Bohannan, B.J.; Relman, D.A. The application of ecological theory toward an understanding of the human microbiome. Science 2012, 336, 1255–1262. [Google Scholar] [CrossRef] [Green Version]
  47. Frank, D.N.; Robertson, C.E.; Hamm, C.M.; Kpadeh, Z.; Zhang, T.; Chen, H.; Zhu, W.; Sartor, R.B.; Boedeker, E.C.; Harpaz, N.; et al. Disease phenotype and genotype are associated with shifts in intestinal-associated microbiota in inflammatory bowel diseases. Inflamm. Bowel. Dis. 2011, 17, 179–184. [Google Scholar] [CrossRef]
  48. Esposito, S.; Polinori, I.; Rigante, D. The gut Microbiota-host partnership as a potential driver of Kawasaki syndrome. Front Pediatr. 2019, 7, 124. [Google Scholar] [CrossRef] [Green Version]
  49. Abrahamsson, T.R.; Jakobsson, H.E.; Andersson, A.F.; Björkstén, B.; Engstrand, L.; Jenmalm, M.C. Low gut microbiota diversity in early infancy precedes asthma at school age. Clin. Experim. Allergy 2014, 44, 842–850. [Google Scholar] [CrossRef] [Green Version]
  50. Vuong, H.E.; Hsiao, E.Y. Emerging roles for the gut microbiome in autism spectrum disorder. Biol. Psychiatr. 2017, 81, 411–423. [Google Scholar] [CrossRef] [Green Version]
  51. Pulikkan, J.; Mazumder, A.; Grace, T. Role of the gut microbiome in autism spectrum disorders. Adv. Experim. Med. Biol. 2019, 1118, 253–269. [Google Scholar] [CrossRef]
  52. Li, Q.; Zhou, J.M. The microbiota-gut-brain axis and its potential therapeutic role in autism spectrum disorder. Neuroscience 2016, 324, 131–139. [Google Scholar] [CrossRef] [PubMed]
  53. Garrett, W.S. Cancer and the microbiota. Science 2015, 348, 80–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Dąbrowska, K.; Witkiewicz, W. Correlations of host genetics and gut microbiome composition. Front Microbiol. 2016, 7, 1357. [Google Scholar] [CrossRef] [Green Version]
  55. Calitri, C.; Ruberto, E.; Castagnola, E. Antibiotic prophylaxis in neutropenic children with acute leukemia: Do the presently available data really support this practice? Eur. J. Haematol. 2018, 101, 721–727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Chai, G.; Governale, L.; McMahon, A.W.; Trinidad, J.P.; Staffa, J.; Murphy, D. Trends of outpatient prescription drug utilization in US children, 2002–2010. Pediatrics 2012, 130, 23–31. [Google Scholar] [CrossRef] [Green Version]
  57. De Luca, M.; Donà, D.; Montagnani, C.; Lo Vecchio, A.; Romanengo, M.; Tagliabue, C.; Centenari, C.; D’Argenio, P.; Lundin, R.; Giaquinto, C.; et al. Antibiotic prescriptions and prophylaxis in Italian children. Is it time to change? Data from the ARPEC project. PLoS ONE 2016, 11, e0154662. [Google Scholar] [CrossRef] [Green Version]
  58. Steliarova-Foucher, E.; Colombet, M.; Ries, L.; Moreno, F.; Dolya, A.; Bray, F.; Hesseling, P.; Shin, H.Y.; Stiller, C.A. IICC-3 contributors. International incidence of childhood cancer, 2001-10: A population-based registry study. Lancet Oncol. 2017, 18, 719–731. [Google Scholar] [CrossRef]
  59. Huang, Y.; Yang, W.; Liu, H.; Duan, J.; Zhang, Y.; Liu, M.; Li, H.; Hou, Z.; Wu, K.K. Effect of high-dose methotrexate chemotherapy on intestinal Bifidobacteria, Lactobacillus and E. coli in children with acute lymphoblastic leukemia. Exp. Biol. Med. 2012, 237, 305–311. [Google Scholar] [CrossRef]
  60. van Vliet, M.J.; Tissing, W.J.; Dun, C.A.; Meessen, N.E.; Kamps, W.A.; de Bont, E.S.; Harmsen, H.J. Chemotherapy treatment in pediatric patients with acute myeloid leukemia receiving antimicrobial prophylaxis leads to a relative increase of colonization with potentially pathogenic bacteria in the gut. Clin. Infect. Dis. 2009, 49, 262–270. [Google Scholar] [CrossRef] [Green Version]
  61. Rothschild, D.; Weissbrod, O.; Barkan, E.; Kurilshikov, A.; Korem, T.; Zeevi, D.; Costea, P.I.; Godneva, A.; Kalka, I.N.; Bar, N.; et al. Environment dominates over host genetics in shaping human gut microbiota. Nature 2018, 555, 210–215. [Google Scholar] [CrossRef]
  62. Levene, I.; Castagnola, E.; Haeusler, G.M. Antibiotic-resistant gram-negative blood stream infections in children with cancer: A review of epidemiology, risk factors, and outcome. Pediatr. Infect. Dis. J. 2018, 37, 495–498. [Google Scholar] [CrossRef] [PubMed]
  63. Groll, A.H.; Castagnola, E.; Cesaro, S.; Dalle, J.H.; Engelhard, D.; Hope, W.; Roilides, E.; Styczynski, J.; Warris, A.; Lehrnbecher, T. Fourth European conference on infections in Leukaemia, infectious diseases working party of the European group for blood marrow transplantation (EBMT-IDWP), infectious diseases group of the European organisation for research and treatment of cancer (EORTC-IDG), international immunocompromised host society (ICHS), & European Leukaemia net (ELN) fourth European Conference on infections in Leukaemia (ECIL-4): Guidelines for diagnosis, prevention, and treatment of invasive fungal diseases in paediatric patients with cancer or allogeneic haemopoietic stem-cell transplantation. Lancet Oncol. 2014, 15, e327–e340. [Google Scholar] [CrossRef] [PubMed]
  64. Cecinati, V.; Brescia, L.; Tagliaferri, L.; Giordano, P.; Esposito, S. Catheter-related infections in pediatric patients with cancer. Eur. J. Clin. Microbiol. Infect. Dis. 2012, 31, 2869–2877. [Google Scholar] [CrossRef] [PubMed]
  65. Galloway-Peña, J.R.; Smith, D.P.; Sahasrabhojane, P.; Ajami, N.J.; Wadsworth, W.D.; Daver, N.G.; Chemaly, R.F.; Marsh, L.; Ghantoji, S.S.; Pemmaraju, N.; et al. The role of the gastrointestinal microbiome in infectious complications during induction chemotherapy for acute myeloid leukemia. Cancer 2016, 122, 2186–2196. [Google Scholar] [CrossRef] [Green Version]
  66. Miedema, K.G.; Winter, R.H.; Ammann, R.A.; Droz, S.; Spanjaard, L.; de Bont, E.S.; Kamps, W.A.; van de Wetering, M.D.; Tissing, W.J. Bacteria causing bacteremia in pediatric cancer patients presenting with febrile neutropenia--species distribution and susceptibility patterns. Support. Care Cancer 2013, 21, 2417–2426. [Google Scholar] [CrossRef]
  67. Castagnola, E.; Haupt, R.; Micozzi, A.; Caviglia, I.; Testi, A.M.; Giona, F.; Parodi, S.; Girmenia, C. Differences in the proportions of fluoroquinolone-resistant Gram-negative bacteria isolated from bacteraemic children with cancer in two Italian centres. Clin. Microbiol. Infect. 2005, 11, 505–507. [Google Scholar] [CrossRef] [Green Version]
  68. Ariffin, H.; Navaratnam, P.; Mohamed, M.; Arasu, A.; Abdullah, W.A.; Lee, C.L.; Peng, L.H. Ceftazidime-resistant Klebsiella pneumoniae bloodstream infection in children with febrile neutropenia. Int. J. Infect. Dis. 2000, 4, 21–25. [Google Scholar] [CrossRef] [Green Version]
  69. Gaynor, E.P.; Sullivan, P.B. Nutritional status and nutritional management in children with cancer. Arch. Dis. Child. 2015, 100, 1169–1172. [Google Scholar] [CrossRef]
  70. Robinson, P.D.; Oberoi, S.; Tomlinson, D.; Duong, N.; Davis, H.; Cataudella, D.; Culos-Reed, N.; Gibson, F.; Götte, M.; Hinds, P.; et al. Management of fatigue in children and adolescents with cancer and in paediatric recipients of haemopoietic stem-cell transplants: A clinical practice guideline. Lancet Child Adolesc. Health 2018, 2, 371–378. [Google Scholar] [CrossRef]
  71. Andreyev, J.; Ross, P.; Donnellan, C.; Lennan, E.; Leonard, P.; Waters, C.; Wedlake, L.; Bridgewater, J.; Glynne-Jones, R.; Allum, W.; et al. Guidance on the management of diarrhoea during cancer chemotherapy. Lancet Oncol. 2014, 15, e447–e460. [Google Scholar] [CrossRef]
  72. Thomsen, M.; Vitetta, L. Adjunctive treatments for the prevention of chemotherapy- and radiotherapy-induced mucositis. Integr. Cancer Ther. 2018, 17, 1027–1047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Manichanh, C.; Varela, E.; Martinez, C.; Antolin, M.; Llopis, M.; Doré, J.; Giralt, J.; Guarner, F.; Malagelada, J.R. The gut microbiota predispose to the pathophysiology of acute postradiotherapy diarrhea. Am. J. Gastroenterol. 2008, 103, 1754–1761. [Google Scholar] [CrossRef] [PubMed]
  74. Stringer, A.M.; Al-Dasooqi, N.; Bowen, J.M.; Tan, T.H.; Radzuan, M.; Logan, R.M.; Mayo, B.; Keefe, D.M.; Gibson, R.J. Biomarkers of chemotherapy-induced diarrhoea: A clinical study of intestinal microbiome alterations, inflammation and circulating matrix metalloproteinases. Support Care Canc. 2013, 21, 1843–1852. [Google Scholar] [CrossRef] [PubMed]
  75. Touchefeu, Y.; Montassier, E.; Nieman, K.; Gastinne, T.; Potel, G.; Bruley des Varannes, S.; Le Vacon, F.; de La Cochetière, M.F. Systematic review: The role of the gut microbiota in chemotherapy- or radiation-induced gastrointestinal mucositis - current evidence and potential clinical applications. Aliment. Pharmacol. Ther. 2014, 40, 409–421. [Google Scholar] [CrossRef] [PubMed]
  76. Iida, N.; Dzutsev, A.; Stewart, C.A.; Smith, L.; Bouladoux, N.; Weingarten, R.A.; Molina, D.A.; Salcedo, R.; Back, T.; Cramer, S.; et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 2013, 342, 967–970. [Google Scholar] [CrossRef] [PubMed]
  77. Gui, Q.F.; Lu, H.F.; Zhang, C.X.; Xu, Z.R.; Yang, Y.H. Well-balanced commensal microbiota contributes to anti-cancer response in a lung cancer mouse model. Gen. Mol. Res. 2015, 14, 5642–5651. [Google Scholar] [CrossRef]
  78. Huang, S.; Li, J.Y.; Wu, J.; Meng, L.; Shou, C.C. Mycoplasma infections and different human carcinomas. World J. Gastroenterol. 2001, 7, 266–269. [Google Scholar] [CrossRef]
  79. Vande Voorde, J.; Balzarini, J.; Liekens, S. Mycoplasmas and cancer: Focus on nucleoside metabolism. EXCLI J. 2014, 13, 300–322. [Google Scholar]
  80. Vande Voorde, J.; Sabuncuoğlu, S.; Noppen, S.; Hofer, A.; Ranjbarian, F.; Fieuws, S.; Balzarini, J.; Liekens, S. Nucleoside-catabolizing enzymes in mycoplasma-infected tumor cell cultures compromise the cytostatic activity of the anticancer drug gemcitabine. J. Biol. Chem. 2014, 289, 13054–13065. [Google Scholar] [CrossRef] [Green Version]
  81. Lehouritis, P.; Cummins, J.; Stanton, M.; Murphy, C.T.; McCarthy, F.O.; Reid, G.; Urbaniak, C.; Byrne, W.L.; Tangney, M. Local bacteria affect the efficacy of chemotherapeutic drugs. Sci. Rep. 2015, 5, 14554. [Google Scholar] [CrossRef] [Green Version]
  82. Viaud, S.; Saccheri, F.; Mignot, G.; Yamazaki, T.; Daillère, R.; Hannani, D.; Enot, D.P.; Pfirschke, C.; Engblom, C.; Pittet, M.J.; et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science 2013, 342, 971–976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Frank, M.; Hennenberg, E.M.; Eyking, A.; Rünzi, M.; Gerken, G.; Scott, P.; Parkhill, J.; Walker, A.W.; Cario, E. TLR signaling modulates side effects of anticancer therapy in the small intestine. J. Immunol. 2015, 194, 1983–1995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Zhou, B.; Xia, X.; Wang, P.; Chen, S.; Yu, C.; Huang, R.; Zhang, R.; Wang, Y.; Lu, L.; Yuan, F.; et al. Induction and amelioration of methotrexate-Induced gastrointestinal toxicity are related to immune response and gut microbiota. eBioMed 2018, 33, 122–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. van Bekkum, D.W.; Roodenburg, J.; Heidt, P.J.; van der Waaij, D. Mitigation of secondary disease of allogeneic mouse radiation chimeras by modification of the intestinal microflora. J. Nat. Cancer Inst. 1974, 52, 401–404. [Google Scholar] [CrossRef] [PubMed]
  86. Holler, E.; Butzhammer, P.; Schmid, K.; Hundsrucker, C.; Koestler, J.; Peter, K.; Zhu, W.; Sporrer, D.; Hehlgans, T.; Kreutz, M.; et al. Metagenomic analysis of the stool microbiome in patients receiving allogeneic stem cell transplantation: Loss of diversity is associated with use of systemic antibiotics and more pronounced in gastrointestinal graft-versus-host disease. Biol. Blood Marrow Transpl. 2014, 20, 640–645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Hidaka, D.; Hayase, E.; Shiratori, S.; Hasegawa, Y.; Ishio, T.; Tateno, T.; Okada, K.; Goto, H.; Sugita, J.; Onozawa, M.; et al. The association between the incidence of intestinal graft-vs-host disease and antibiotic use after allogeneic hematopoietic stem cell transplantation. Clin. Transpl. 2018, 32, e13361. [Google Scholar] [CrossRef] [PubMed]
  88. Shono, Y. Rinsho ketsueki. Jpn. J. Clin. Hematol. 2017, 58, 835–842. [Google Scholar] [CrossRef]
  89. Shono, Y.; van den Brink, M. Gut microbiota injury in allogeneic haematopoietic stem cell transplantation. Nat. Rev. Cancer 2018, 18, 283–295. [Google Scholar] [CrossRef]
  90. Jenq, R.R.; Taur, Y.; Devlin, S.M.; Ponce, D.M.; Goldberg, J.D.; Ahr, K.F.; Littmann, E.R.; Ling, L.; Gobourne, A.C.; Miller, L.C.; et al. Intestinal blautia is associated with reduced death from graft-versus-host disease. Biol. Blood Marrow Transpl. 2015, 21, 1373–1383. [Google Scholar] [CrossRef] [Green Version]
  91. Biagi, E.; Zama, D.; Nastasi, C.; Consolandi, C.; Fiori, J.; Rampelli, S.; Turroni, S.; Centanni, M.; Severgnini, M.; Peano, C.; et al. Gut microbiota trajectory in pediatric patients undergoing hematopoietic SCT. Bone Marrow Transpl. 2015, 50, 992–998. [Google Scholar] [CrossRef] [Green Version]
  92. Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; de Roos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef] [PubMed]
  93. Trompette, A.; Gollwitzer, E.S.; Yadava, K.; Sichelstiel, A.K.; Sprenger, N.; Ngom-Bru, C.; Blanchard, C.; Junt, T.; Nicod, L.P.; Harris, N.L.; et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat. Med. 2014, 20, 159–166. [Google Scholar] [CrossRef] [PubMed]
  94. Biagi, E.; Zama, D.; Rampelli, S.; Turroni, S.; Brigidi, P.; Consolandi, C.; Severgnini, M.; Picotti, E.; Gasperini, P.; Merli, P.; et al. Early gut microbiota signature of a GvHD in children given allogeneic hematopoietic cell transplantation for hematological disorders. BMC Med. Genom. 2019, 12, 49. [Google Scholar] [CrossRef] [PubMed]
  95. D’Amico, F.; Soverini, M.; Zama, D.; Consolandi, C.; Severgnini, M.; Prete, A.; Pession, A.; Barone, M.; Turroni, S.; Biagi, E.; et al. Gut resistome plasticity in pediatric patients undergoing hematopoietic stem cell transplantation. Sci. Rep. 2019, 9, 5649. [Google Scholar] [CrossRef] [PubMed]
  96. Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef] [Green Version]
  97. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document. The international scientific association for probiotics and prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Rianda, D.; Agustina, R.; Setiawan, E.A.; Manikam, N. Effect of probiotic supplementation on cognitive function in children and adolescents: A systematic review of randomised trials. Beneficial. Microb. 2019, 10, 873–882. [Google Scholar] [CrossRef]
  99. Mantegazza, C.; Molinari, P.; D’Auria, E.; Sonnino, M.; Morelli, L.; Zuccotti, G.V. Probiotics and antibiotic-associated diarrhea in children: A review and new evidence on Lactobacillus rhamnosus GG during and after antibiotic treatment. Pharmacol. Res. 2018, 128, 63–72. [Google Scholar] [CrossRef]
  100. Ananthan, A.; Balasubramanian, H.; Rao, S.; Patole, S. Probiotic supplementation in children with cystic fibrosis-a systematic review. Eur. J. Pediatr. 2016, 175, 1255–1266. [Google Scholar] [CrossRef]
  101. Huang, R.; Ning, H.; Shen, M.; Li, J.; Zhang, J.; Chen, X. Probiotics for the treatment of atopic dermatitis in children: A systematic review and meta-analysis of randomized controlled trials. Front Cell. Infect. Microbiol. 2017, 7, 392. [Google Scholar] [CrossRef]
  102. Scott, A.M.; Clark, J.; Julien, B.; Islam, F.; Roos, K.; Grimwood, K.; Little, P.; Del Mar, C.B. Probiotics for preventing acute otitis media in children. Cochrane Database Syst. Rev. 2019, 6, CD012941. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Grimaldi, R.; Gibson, G.R.; Vulevic, J.; Giallourou, N.; Castro-Mejía, J.L.; Hansen, L.H.; Leigh Gibson, E.; Nielsen, D.S.; Costabile, A. A prebiotic intervention study in children with autism spectrum disorders (ASDs). Microbiome 2018, 6, 133. [Google Scholar] [CrossRef] [PubMed]
  104. Hume, M.P.; Nicolucci, A.C.; Reimer, R.A. Prebiotic supplementation improves appetite control in children with overweight and obesity: A randomized controlled trial. Am. J. Clin. Nutr. 2017, 105, 790–799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Ekert, H.; Jurk, I.H.; Waters, K.D.; Tiedemann, K. Prophylactic co-trimoxazole and lactobacilli preparation in neutropenic patients. Med. Pediatr. Oncol. 1980, 8, 47–51. [Google Scholar] [CrossRef] [PubMed]
  106. Wada, M.; Nagata, S.; Saito, M.; Shimizu, T.; Yamashiro, Y.; Matsuki, T.; Asahara, T.; Nomoto, K. Effects of the enteral administration of Bifidobacterium breve on patients undergoing chemotherapy for pediatric malignancies. Support Care Cancer 2010, 18, 751–759. [Google Scholar] [CrossRef]
  107. Vahabnezhad, E.; Mochon, A.B.; Wozniak, L.J.; Ziring, D.A. Lactobacillus bacteremia associated with probiotic use in a pediatric patient with ulcerative colitis. J. Clin. Gastroenterol. 2013, 47, 437–439. [Google Scholar] [CrossRef] [Green Version]
  108. Ambesh, P.; Stroud, S.; Franzova, E.; Gotesman, J.; Sharma, K.; Wolf, L.; Kamholz, S. Recurrent Lactobacillus bacteremia in a patient with Leukemia. J. Invest. Med. 2017, 5, 2324709617744233. [Google Scholar] [CrossRef] [Green Version]
  109. Salminen, M.K.; Rautelin, H.; Tynkkynen, S.; Poussa, T.; Saxelin, M.; Valtonen, V.; Järvinen, A. Lactobacillus bacteremia, clinical significance, and patient outcome, with special focus on probiotic L. rhamnosus GG. Clin. Infect. Dis. 2004, 38, 62–69. [Google Scholar] [CrossRef] [Green Version]
  110. Nakama, K.A.; Dos Santos, R.B.; Serpa, P.; Maciel, T.R.; Haas, S.E. Organoleptic excipients used in pediatric antibiotics. Arch. Pediatr. 2019, 26, 431–436. [Google Scholar] [CrossRef]
  111. WHO. Antimicrobial Resistance: Global Report on Surveillance 2014; WHO: Geneva, Switzerland, 2016. [Google Scholar]
  112. Kokai-Kun, J.F.; Roberts, T.; Coughlin, O.; Le, C.; Whalen, H.; Stevenson, R.; Wacher, V.J.; Sliman, J. Use of ribaxamase (SYN-004), a β-lactamase, to prevent Clostridium difficile infection in β-lactam-treated patients: A double-blind, phase 2b, randomised placebo-controlled trial. Lancet Infect Dis. 2019, 19, 487–496. [Google Scholar] [CrossRef]
  113. D’Amico, F.; Biagi, E.; Rampelli, S.; Fiori, J.; Zama, D.; Soverini, M.; Barone, M.; Leardini, D.; Muratore, E.; Prete, A.; et al. Enteral nutrition in pediatric patients undergoing hematopoietic SCT promotes the recovery of gut microbiome homeostasis. Nutrients 2019, 11, 2958. [Google Scholar] [CrossRef] [Green Version]
  114. Murphy, A.J.; White, M.; Elliott, S.A.; Lockwood, L.; Hallahan, A.; Davies, P.S. Body composition of children with cancer during treatment and in survivorship. Am. J. Clin. Nutr. 2015, 102, 891–896. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Chemaitilly, W.; Cohen, L.E.; Mostoufi-Moab, S.; Patterson, B.C.; Simmons, J.H.; Meacham, L.R.; van Santen, H.M.; Sklar, C.A. Endocrine late effects in childhood cancer survivors. J. Clin. Oncol. 2018, 36, 2153–2159. [Google Scholar] [CrossRef] [PubMed]
  116. Jäger, R.; Purpura, M.; Farmer, S.; Cash, H.A.; Keller, D. Probiotic Bacillus coagulans GBI-30, 6086 improves protein absorption and utilization. Probiotics Antimicrob. Proteins 2018, 10, 611–615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Kesika, P.; Sivamaruthi, B.S.; Chaiyasut, C. Do probiotics improve the health status of individuals with diabetes mellitus? A review on outcomes of clinical trials. BioMed Res. Int. 2019, 2019, 1531567. [Google Scholar] [CrossRef]
  118. Million, M.; Diallo, A.; Raoult, D. Gut microbiota and malnutrition. Microb. Pathog. 2017, 106, 127–138. [Google Scholar] [CrossRef] [PubMed]
  119. Guidotti, L.; Solari, F.; Bertolini, P.; Gebennini, E.; Ghiaroni, G.; Corsano, P. Reminiscing on acute and chronic events in children with cancer and their parents: An exploratory study. Child Care Health Dev. 2019, 45, 568–576. [Google Scholar] [CrossRef]
  120. Kaatsch, P. Epidemiology of childhood cancer. Cancer Treat Rev. 2010, 36, 277–285. [Google Scholar] [CrossRef]
Table 1. Main studies regarding antibiotic administration in children with cancer.
Table 1. Main studies regarding antibiotic administration in children with cancer.
Author, YearCriteria for ProphylaxisAntibiotics Employed
Cecinati, 2013Children with cancer and
probably long-lasting
neutropenia, in accordance to the chemotherapics employed
TMP-SMX → 10–30 mg/kg/die or 20 mg/kg/die
CPF → 25 mg/kg/die or 20 mg/kg/die
Amoxicillin + clavulanic acid → 25 mg twice daily for 15 days maximum
Chastagner, 2018Patients with AML or ALL in order to prevent infections related to mortality
Oral TMP-SMX 25 mg/kg/die every two days
Choeyprasert, 2017To all HCT recipients on the day on which conditioning regimens started, until engraftment, and discontinuation was indicated when the patients developed fever, clinically documented infection or suspected infection
Oral Ciprofloxacin 20–30 mg/kg/die in two divided doses + Penicillin V sodium 25–50 mg/kg/die in four doses
Al Omar, 2017To each CT course when the ANC was ≤1000 cells/mm3 and continued until AMC was ≥100 cells/mm3 postnadir
Ciprofloxacin → 10 mg/kg/die every 12 h
Cotrimoxazole → 2.5 mg/kg/die every 12 h for two or consecutive days per week
Alexander, 2018To all patients aged 6 months to 21 years with acute leukemia (any AML or relapsed ALL) or HCT recipients
Levofloxacin → 6 months to 5 years: 10 mg/kg twice daily; >5 years: 10 mg/kg once daily
Table 2. Main studies on the gut microbiota and chemotherapy dynamics.
Table 2. Main studies on the gut microbiota and chemotherapy dynamics.
Author, YearFinding
Huang [78], 2001Certain bacteria reside in the tumor tissue and can directly
modulate chemotherapy by producing nucleoside
analogue-catabolizing enzymes, which can interfere
with antineoplastic drugs.
Lehouritis [81], 2015Escherichia coli nitroreductase activity is able to enhance the
cytotoxicity of the drug CB1954.
Iida [76], 2013Shifts in microbiota decrease the production of ROS and oxidative damage, key mechanisms in many anticancer drugs.
Viaud [82], 2013Gram-positive bacterial decontamination with antibiotics reduces the stimulation of the Th1 and Th17 immune responses, thus
impairing the efficacy of cyclophosphamide.
Frank [83], 2015Methotrexate can induce gastrointestinal toxicity: in murine models, gut microbiota depletion has been linked to poorer TLR2 activation and therefore lower expression of the multidrug resistance pump ABCB1/MDR1. The TLR2 pathway has been proven to reduce the toxic effects of methotrexate on the gut epithelium.
Table 3. Effects of different antibiotics on intestinal microbiota.
Table 3. Effects of different antibiotics on intestinal microbiota.
AntibioticEffects on Intestinal Microbial
Amoxicillin/clavulanate acid
  • Reduction in bacterial diversity
  • Increase in abundance of Enterococcus spp. and Enterobacteriaceae (Citrobacter spp., Klebsiella spp., Proteus spp.)
  • Reduction in Clostridium spp., Bifidobacterium spp., Lactobacillus spp., Roseburia spp.
Cephalosporins
  • Depletion of Enterobacteriaceae spp. and Escherichia coli
  • Increase in the abundance of Enterococcus, Citrobacter spp., Klebsiella spp., Pseudomonas spp.
  • Increase in the colonization of Clostridium difficile for high generation cephalosporins
Piperacillin
  • Increase in abundance of Enterococcus spp. and Escherichia coli
  • Depletion of Bacteroides spp., Bifidobacterium spp., Clostridium spp., Lactospirum spp., Lactobacillus spp.
Trimethoprim-Sulfamethoxazole
  • Reduction in the abundance of Enterobacteriaceae spp. and Escherichia coli
  • Increase of resistant Escherichia coli, Acinetobacter spp. and Pseudomonas spp.
Fluoroquinolones
  • Reduction in the abundance of Enterobacteriaceae, Escherichia coli, Bacillus spp., Corynebacterium spp.
  • Depletion of some anaerobic bacteria (Bacteroides spp., Bifidobacterium spp., Lactobacillus spp., Peptostreptococcus spp., Veilonella spp.)
  • Increase in the abundance of Citrobacter spp., Enterobacter and Klebsiella spp.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bossù, G.; Di Sario, R.; Argentiero, A.; Esposito, S. Antimicrobial Prophylaxis and Modifications of the Gut Microbiota in Children with Cancer. Antibiotics 2021, 10, 152. https://0-doi-org.brum.beds.ac.uk/10.3390/antibiotics10020152

AMA Style

Bossù G, Di Sario R, Argentiero A, Esposito S. Antimicrobial Prophylaxis and Modifications of the Gut Microbiota in Children with Cancer. Antibiotics. 2021; 10(2):152. https://0-doi-org.brum.beds.ac.uk/10.3390/antibiotics10020152

Chicago/Turabian Style

Bossù, Gianluca, Riccardo Di Sario, Alberto Argentiero, and Susanna Esposito. 2021. "Antimicrobial Prophylaxis and Modifications of the Gut Microbiota in Children with Cancer" Antibiotics 10, no. 2: 152. https://0-doi-org.brum.beds.ac.uk/10.3390/antibiotics10020152

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

Article Metrics

Back to TopTop