gastrointestinal mucositis
Recently Published Documents


TOTAL DOCUMENTS

81
(FIVE YEARS 15)

H-INDEX

18
(FIVE YEARS 1)

Biomedicines ◽  
2021 ◽  
Vol 9 (10) ◽  
pp. 1389
Author(s):  
Ana Rita da Silva Ferreira ◽  
Anne-Grete Märtson ◽  
Alyse de Boer ◽  
Hannah R. Wardill ◽  
Jan-Willem Alffenaar ◽  
...  

Antimicrobial prophylaxis is increasingly being used in patients with hematological malignancies receiving high-dose chemotherapy and hematopoietic stem cell transplantation (HSCT). However, few studies have focused on the potential impact of gastrointestinal mucositis (GI-M), a frequently observed side effect of chemotherapy in patients with cancer that affects the gastrointestinal microenvironment, on drug absorption. In this review, we discuss how chemotherapy leads to an overall loss of mucosal surface area and consequently to uncontrolled transport across the barrier. The barrier function is depending on intestinal luminal pH, intestinal motility, and diet. Another factor contributing to drug absorption is the gut microbiota, as it modulates the bioavailability of orally administrated drugs by altering the gastrointestinal properties. To better understand the complex interplay of factors in GI-M and drug absorption we suggest: (i) the longitudinal characterization of the impact of GI-M severity on drug exposure in patients, (ii) the development of tools to predict drug absorption, and (iii) strategies that allow the support of the gut microbiota. These studies will provide relevant data to better design strategies to reduce the severity and impact of GI-M in patients with cancer.


JBMTCT ◽  
2021 ◽  
Vol 2 (2) ◽  
pp. p51
Author(s):  
Cristina De Paula Novaes

Objectives: To compare the severity of oral mucositis and the frequency of gastrointestinal mucositis, and to observe if there is impact of these adverse effects on overall survival (OS), in patients who underwent CBV (carmustine, BCNU, and VP-16) and LEAM (lomustine, etoposide, Ara-C, and melphalan) conditioning for autologous hematopoietic cell transplantation (aHCT). Method: We collected retrospective data from medical records (n = 120) of transplantation and mucositis in the digestive tract of Hodgkin’s and non-Hodgkin’s lymphoma patients. Results: The frequency of OM grade 1 was higher in LEAM (36.76%) than in CBV (19.72%, p=0.038). There were no significant differences between the frequency of gastrointestinal mucositis in the two regimens (CBV - 52.11% and LEAM - 63.27%, p=0.305). CBV regimen exhibited lower 1-year overall survival (OS) than did LEAM (p=0.003). Oral mucositis grade ≥2 was associated with reduced OS in the CBV group (p=0.013). CBV regimen (HR=2.98, p 0.005) and oral mucositis grade ≥2 (HR=2.17, p=0.013) interfered negatively on the OS rate. Conclusion: Oral mucositis was more severe in CBV than in LEAM, decreasing the OS rate. Further studies with comprehensive follow-up and toxicity analyses must be undertaken to clarify the safety of LEAM conditioning in the digestive tract.


2021 ◽  
Vol 16 (1) ◽  
Author(s):  
Jing Liu ◽  
Chao Liu ◽  
Jinbo Yue

AbstractAn ever-growing body of evidence has linked the gut microbiome with both the effectiveness and the toxicity of cancer therapies. Radiotherapy is an effective way to treat tumors, although large variations exist among patients in tumor radio-responsiveness and in the incidence and severity of radiotherapy-induced side effects. Relatively little is known about whether and how the microbiome regulates the response to radiotherapy. Gut microbiota may be an important player in modulating “hot” versus “cold” tumor microenvironment, ultimately affecting treatment efficacy. The interaction of the gut microbiome and radiotherapy is a bidirectional function, in that radiotherapy can disrupt the microbiome and those disruptions can influence the effectiveness of the anticancer treatments. Limited data have shown that interactions between the radiation and the microbiome can have positive effects on oncotherapy. On the other hand, exposure to ionizing radiation leads to changes in the gut microbiome that contribute to radiation enteropathy. The gut microbiome can influence radiation-induced gastrointestinal mucositis through two mechanisms including translocation and dysbiosis. We propose that the gut microbiome can be modified to maximize the response to treatment and minimize adverse effects through the use of personalized probiotics, prebiotics, or fecal microbial transplantation. 16S rRNA sequencing is the most commonly used approach to investigate distribution and diversity of gut microbiome between individuals though it only identifies bacteria level other than strain level. The functional gut microbiome can be studied using methods involving metagenomics, metatranscriptomics, metaproteomics, as well as metabolomics. Multiple ‘-omic’ approaches can be applied simultaneously to the same sample to obtain integrated results. That said, challenges and remaining unknowns in the future that persist at this time include the mechanisms by which the gut microbiome affects radiosensitivity, interactions between the gut microbiome and combination treatments, the role of the gut microbiome with regard to predictive and prognostic biomarkers, the need for multi “-omic” approach for in-depth exploration of functional changes and their effects on host-microbiome interactions, and interactions between gut microbiome, microbial metabolites and immune microenvironment.


2020 ◽  
Vol 115 (1) ◽  
pp. S1016-S1017
Author(s):  
Umair M. Nasir ◽  
Alexander Mozeika ◽  
Imraan Jan ◽  
Catherine Choi ◽  
Shima Ghavimi ◽  
...  

2020 ◽  
Vol 40 (9) ◽  
pp. 5291-5294 ◽  
Author(s):  
UMAIR M. NASIR ◽  
ALEXANDER M. MOZEIKA ◽  
MUTLAY SAYAN ◽  
IMRAAN JAN ◽  
NOEL KOWAL ◽  
...  

Gut Microbes ◽  
2020 ◽  
Vol 12 (1) ◽  
pp. 1809332
Author(s):  
H. R. Wardill ◽  
A. R. da Silva Ferreira ◽  
S. Lichtenberg Cloo ◽  
R. Havinga ◽  
H. J. M. Harmsen ◽  
...  

2020 ◽  
Vol 11 (4) ◽  
pp. 305-318
Author(s):  
W.G. Lima ◽  
R.M. Pessoa ◽  
K.D. Vital ◽  
I.K.T.M. Takenaka ◽  
V.N. Cardoso ◽  
...  

Gastrointestinal mucositis (GIM) is an inflammation caused by antitumor therapy, especially after chemotherapy and radiotherapy. Currently in the clinical practice, only palliative measures are taken to treat GIM, representing the main clinical limitation in the management of this condition. Several studies have highlighted the potential benefits of probiotics for the management of GIM, but the actual role of these microorganisms in the maintenance of intestinal homeostasis remains elusive. In this context, here we aimed to realise a systematic review with meta-analysis to evaluate the effect of probiotics on experimental GIM. The meta-analysis showed that probiotics significantly suppressed the body weight loss related to GIM in rodents (95% confidence interval (CI): -2.67 to -0.70; I2=98%, P<0.00). Subgroup analysis showed that pre-treatment (≥7 days before chemotherapy) (95% CI: -8.84 to -0.17; I2=98%, P<0.04) with a high dose of probiotics (≥ 109 cfu/day) (95% CI: -2.58 to -0.28; I2=98%, P<0.00) comprising two or more microorganism species (95% CI: -6.49 to -0.28; I2=96%, P=0.03) remedied GIM more effectively. It was also revealed that fungi (specifically Saccharomyces boullardii) are more effective in remedying GIM than bacteria (P=0.03 vs P<0.00), and the mouse models are more receptive than rats to the enteroprotective effects of probiotics (95% CI: -4.76, -0.69; I2=97%, P=0.01). Qualitative analyses highlighted that probiotics suppress GIM through several mechanisms; they reduce the intestinal permeability, suppress the pro-inflammatory cytokine production while stimulating production and secretion of anti-inflammatory cytokines, inhibit the signalling pathways coupled to inflammation and apoptosis, accelerate the proliferation of enterocytes, reduce the levels of reactive oxygen species, and help maintain the protective mucus layer. In conclusion, this review highlights the therapeutic benefits of probiotics in experimental GIM.


Antioxidants ◽  
2020 ◽  
Vol 9 (8) ◽  
pp. 699 ◽  
Author(s):  
Giacomo Pepe ◽  
Shara Francesca Rapa ◽  
Emanuela Salviati ◽  
Alessia Bertamino ◽  
Giulia Auriemma ◽  
...  

Intestinal epithelial cells (IECs) play a pivotal role in maintaining intestinal homeostasis. Different noxious agents, among them also anticancer therapies, can impair intestinal epithelial integrity triggering inflammation and oxidative stress. A frequent complication of chemotherapy is gastrointestinal mucositis, strongly influencing the effectiveness of therapy, increasing healthcare costs, and impairing patients’ quality of life. Different strategies are used to treat gastrointestinal mucositis, including products from natural sources. Our study focused on the effect of pomegranate (Punica granatum L.) juice extract on IEC-6 cells, both during inflammatory conditions and following treatment with 5-fluorouracil (5-FU). The polyphenolic profile of pomegranate juice was characterized in detail by Online Comprehensive two dimensional Liquid Chromatography-Mass Spectrometry. The evaluation of pomegranate juice extract in IEC-6 indicates a significant inhibition in proinflammatory factors, such as cytokines release, cyclooxygenase-2 and inducible nitric oxide synthase expression, and nitrotyrosine formation. Pomegranate also inhibited oxidative stress and adhesion protein expression. In 5-FU-treated IEC-6, pomegranate also inhibited both inflammatory and oxidative stress parameters and apoptosis. It promoted wound repair and tight junction expression. These results suggest a potential use of pomegranate as an adjuvant in the treatment of intestinal inflammatory and oxidative stress states, which also occur during chemotherapy-induced mucositis.


Sign in / Sign up

Export Citation Format

Share Document