Acessibilidade / Reportar erro

Could paraprobiotics be a safer alternative to probiotics for managing cancer chemotherapy-induced gastrointestinal toxicities?

Abstract

Clinical oncology has shown outstanding progress improving patient survival due to the incorporation of new drugs. However, treatment success may be reduced by the emergency of dose-limiting side effects, such as intestinal mucositis and diarrhea. Mucositis and diarrhea management is symptomatic, and there is no preventive therapy. Bacterial and fungal-based compounds have been suggested as an alternative for preventing the development of diarrhea in cancer patients. Using probiotics is safe and effective in immunocompetent individuals, but concerns remain during immunosuppressive conditions. Paraprobiotics, formulations composed of non-viable microorganisms, have been proposed to overcome such limitation. The present literature review discusses current evidence regarding the possible use of paraprobiotics as an alternative to probiotics to prevent gastrointestinal toxicity of cancer chemotherapy.

Diarrhea; Probiotics; Paraprobiotics; Cancer; Chemotherapy


Introduction

The survival of patients undergoing chemotherapy or radiotherapy has increased in the last decades due to considerable advances in drug therapy, a better understanding of cancer pathogenesis, and increased awareness of the population's importance of early disease diagnosis (11. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin 2016; 66: 7-30, doi: 10.3322/caac.21332.
https://doi.org/10.3322/caac.21332...
). The evolvement of cancer treatment led to the development of personalized and more selective therapies. However, the high cost of some modern therapeutic approaches limits patient access to precision medicine in specific cancer centers, especially in developing countries, where most cancer-related deaths are concentrated (22. Lee YT, Tan YJ, Oon CE. Molecular targeted therapy: treating cancer with specificity. Eur J Pharmacol 2018; 834: 188-196, doi: 10.1016/j.ejphar.2018.07.034.
https://doi.org/10.1016/j.ejphar.2018.07...
,33. WHO (World Health Organization). Cancer. Geneva: World Health Organization; 2021.).

Therapeutic regimens used worldwide are still based on non-selective cytotoxic drugs and promote unwanted side effects that compromise therapeutic efficacy and can contribute to increased mortality due to toxicities (44. Wallington M, Saxon EB, Bomb M, Smittenaar R, Wickenden M, McPhail S, et al. 30-day mortality after systemic anticancer treatment for breast and lung cancer in England: a population-based, observational study. Lancet Oncol 2016; 17: 1203-1216, doi: 10.1016/S1470-2045(16)30383-7.
https://doi.org/10.1016/S1470-2045(16)30...
). Among toxicities, mucositis affects the entire gastrointestinal tract and is accompanied by pain, vomiting, and diarrhea, usually leading to treatment interruption or reduction in the chemotherapy dose intensity. Clinical management of gastrointestinal mucositis is symptomatic, and there is still no preventive or curative treatment (55. Van Sebille YZA, Stansborough R, Wardill HR, Bateman E, Gibson RJ, Keefe DM. Management of mucositis during chemotherapy: from pathophysiology to pragmatic therapeutics. Curr Oncol Rep 2015; 17: 50, doi: 10.1007/s11912-015-0474-9.
https://doi.org/10.1007/s11912-015-0474-...
).

The intestinal microbiota in healthy individuals plays an essential role in maintaining intestinal homeostasis, with protective effects on epithelial integrity. The mechanism seems to involve bacterial interaction with toll-like receptors and activation of the NF-κB signaling pathway, avoiding mucosal damage and stimulating cell repair and regeneration (66. Stringer AM. Interaction between host cells and microbes in chemotherapy-induced mucositis. Nutrients 2013; 5: 1488-1499, doi: 10.3390/nu5051488.
https://doi.org/10.3390/nu5051488...
,77. Touchefeu Y, Montassier E, Nieman K, Gastinne T, Potel G, Bruley Des Varannes S, et al. 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, doi: 10.1111/apt.12878.
https://doi.org/10.1111/apt.12878...
). Additionally, the mucus produced by the intestinal epithelial cells and the expression of intercellular junction proteins are essential components of the intestinal barrier against pathogens (88. Ulluwishewa D, Anderson RC, McNabb WC, Moughan PJ, Wells JM, Roy NC. Regulation of tight junction permeability by intestinal bacteria and dietary components. J Nutr 2011; 141: 769-776, doi: 10.3945/jn.110.135657.
https://doi.org/10.3945/jn.110.135657...
). Anticancer drugs promote dysbiosis that induces changes in intestinal permeability and inflammation. The use of bacteria-based formulations to balance the intestinal microbiota is recognized for its health benefits and can mitigate the deleterious impact of chemotherapy on the gastrointestinal tract (99. Lu D, Yan J, Liu F, Ding P, Chen B, Lu Y, et al. Probiotics in preventing and treating chemotherapy-induced diarrhea: a meta-analysis. Asia Pac J Clin Nutr 2019; 28: 701-710, doi: 10.6133/apjcn.201912_28(4).0005.
https://doi.org/10.6133/apjcn.201912_28(...
).

Currently, there is a wide variety of bacteria-composed products, like probiotics, prebiotics, symbiotics, postbiotics, and paraprobiotics. Table 1 shows the definitions and potential applications of these compounds. Probiotics, composed of live organisms, exert their effects through modulation of the immune response, activation of regulatory cytokines, maintenance of the epithelial barrier integrity, and competition with pathogenic microorganisms. It is a complementary therapy that quantitatively and qualitatively alters intestinal microbiota composition (1010. Molska M, Reguła J. Potential mechanisms of probiotics action in the prevention and treatment of colorectal cancer. Nutrients 2019; 11: 2453, doi: 10.3390/nu11102453.
https://doi.org/10.3390/nu11102453...
).

Table 1
Probiotic, paraprobiotic, and postbiotic definitions and uses.

Probiotics have not been associated with the appearance of significant adverse effects in immunocompetent individuals. On the other hand, the safety of such compounds in immunocompromised patients should be carefully analyzed (1111. Vesterlund S, Vankerckhoven V, Saxelin M, Goossens H, Salminen S, Ouwehand AC. Safety assessment of Lactobacillus strains: presence of putative risk factors in faecal, blood and probiotic isolates. Int J Food Microbiol 2007; 116: 325-331, doi: 10.1016/j.ijfoodmicro.2007.02.002.
https://doi.org/10.1016/j.ijfoodmicro.20...
). Recent studies have evaluated whether bacterial viability is essential for their health benefits. The verification that these positive effects can also be achieved with dead bacteria, known as paraprobiotics, without certain risks of administering a live organism is of great importance. The present review explores the perspectives of using paraprobiotics as an alternative to probiotics to prevent the gastrointestinal toxicities of cancer chemotherapy.

Material and Methods

The search strategy was performed using MEDLINE (via PubMed) database, where the search terms were “Paraprobiotics” OR “Heat-killed Probiotic” OR “Non-viable Probiotic” OR “Ghost Probiotic” OR “Probiotics”. We combined the terms, through the Boolean operator AND, with the MeSH descriptors “Cancer”, “Adverse Effects”, “Safety”, “Diarrhea”, and “Clinical Trial”. We selected original articles or case reports in English according to their relevance to the review topic.

Chemotherapy-associated intestinal microbiota dysbiosis and diarrhea

Cancer treatment induces intestinal mucositis, which considerably worsens the quality of life of patients (1212. Aprile G, Rihawi K, De Carlo E, Sonis ST. Treatment-related gastrointestinal toxicities and advanced colorectal or pancreatic cancer: a critical update. World J Gastroenterol 2015; 21: 11793-11803, doi: 10.3748/wjg.v21.i41.11793.
https://doi.org/10.3748/wjg.v21.i41.1179...
). Diarrhea is one of the most important and debilitating manifestations of intestinal mucositis. The frequency and occurrence of diarrhea depend on the drug used and the therapeutic regimen. For instance, the weekly regimen of irinotecan or 5-fluorouracil (5-FU) causes the highest rate of this adverse effect, with more than 10% of patients developing grade 3 or 4 diarrhea (1313. Stein A, Voigt W, Jordan K. Chemotherapy-induced diarrhea: pathophysiology, frequency and guideline-based management. Ther Adv Med Oncol 2010; 2: 51-63, doi: 10.1177/1758834009355164.
https://doi.org/10.1177/1758834009355164...
). The chemotherapeutic agent irinotecan is a significant aggravating factor. The FOLFIRI regimen (5-FU, leucovorin, and irinotecan) triggers diarrhea in 89% of patients. Additionally, therapy with FOLFOX (5-FU, leucovorin, and oxaliplatin) induces any diarrhea grade in 56% of patients (1414. Ghoreishi Z, Shidfar F, Iravani M, Esfahani A, Ghavamzadeh A. Effect of vitamin E on chemotherapy-induced mucositis and neutropenia in leukemic patients undergoing bone marrow transplantation. Asia Pac J Clin Oncol 2007; 3: 113-118, doi: 10.1111/j.1743-7563.2007.00111.x.
https://doi.org/10.1111/j.1743-7563.2007...
). Conversely, patients exposed to other anticancer drugs, such as doxorubicin and methotrexate, also manifest some degree of gastrointestinal toxicities (1515. Viele CS. Overview of chemotherapy-induced diarrhea. Semin Oncol Nurs 2003; 19: 2-5, doi: 10.1053/j.soncn.2003.09.007.
https://doi.org/10.1053/j.soncn.2003.09....
,1616. Saltz LB. Understanding and managing chemotherapy-induced diarrhea. J Support Oncol 2003; 1: 35-46; discussion 38-41, 45-46.). Despite diarrhea and intestinal mucositis being common side effects in patients undergoing cancer therapy, the mechanisms that trigger them are not fully understood, though some advancements have been seen (1717. Ribeiro RA, Wanderley CWS, Wong DVT, Mota JMSC, Leite CAVG, Souza MHLP, et al. Irinotecan- and 5-fluorouracil-induced intestinal mucositis: insights into pathogenesis and therapeutic perspectives. Cancer Chemother Pharmacol 2016; 78: 881-893, doi: 10.1007/s00280-016-3139-y.
https://doi.org/10.1007/s00280-016-3139-...
).

There are several types of cancer therapy-related diarrhea: secretory, osmotic, exudative, malabsorptive, caused by dysmotility, infectious, inflammatory, and steatorrhea (1818. Gibson RJ, Keefe DMK. Cancer chemotherapy-induced diarrhoea and constipation: mechanisms of damage and prevention strategies. Support Care Cancer 2006; 14: 890-900, doi: 10.1007/s00520-006-0040-y.
https://doi.org/10.1007/s00520-006-0040-...
). Clinical reports suggest that chemotherapy-induced diarrhea may develop due to changes in intestinal absorption accompanied by excessive fluid secretion, or it may be a consequence of chemotherapy's biochemical and inflammatory changes (1818. Gibson RJ, Keefe DMK. Cancer chemotherapy-induced diarrhoea and constipation: mechanisms of damage and prevention strategies. Support Care Cancer 2006; 14: 890-900, doi: 10.1007/s00520-006-0040-y.
https://doi.org/10.1007/s00520-006-0040-...
). Current knowledge is mainly obtained from animal models using irinotecan or 5-FU. Chemotherapy administration damages the intestine, which is accompanied by excessive mucus secretion. Irinotecan induces diarrhea due to malabsorption of water and electrolytes and increases mucin secretion (1919. Ikuno N, Soda H, Watanabe M, Oka M. Irinotecan (CPT-11) and characteristic mucosal changes in the mouse ileum and cecum. J Natl Cancer Inst 1995; 87: 1876-1883, doi: 10.1093/jnci/87.24.1876.
https://doi.org/10.1093/jnci/87.24.1876...
). Several apoptotic cells in the intestinal epithelium and colon, combined with the increase in goblet cells, decrease the absorptive capacity considerably, triggering diarrhea (2020. Gibson RJ, Bowen JM, Inglis MRB, Cummins AG, Keefe DMK. Irinotecan causes severe small intestinal damage, as well as colonic damage, in the rat with implanted breast cancer. J Gastroenterol Hepatol 2003; 18: 1095-1100, doi: 10.1046/j.1440-1746.2003.03136.x.
https://doi.org/10.1046/j.1440-1746.2003...
).

Such toxicity is primarily associated with intestinal microbiota dysbiosis (2121. Deleemans JM, Chleilat F, Reimer RA, Baydoun M, Piedalue KA, Lowry DE, et al. The chemo-gut pilot study: associations between gut microbiota, gastrointestinal symptoms, and psychosocial health outcomes in a cross-sectional sample of young adult cancer survivors. Curr Oncol 2022; 29: 2973-2994, doi: 10.3390/curroncol29050243.
https://doi.org/10.3390/curroncol2905024...
,2222. Akbarali HI, Muchhala KH, Jessup DK, Cheatham S. Chemotherapy induced gastrointestinal toxicities. Adv Cancer Res 2022; 155: 131-166, doi: 10.1016/bs.acr.2022.02.007.
https://doi.org/10.1016/bs.acr.2022.02.0...
). It is not yet clear how the drugs alter gut microbiota composition. Indeed, animal models and clinical trials indicate that irinotecan, 5-FU, and radiotherapy modify the gut microbiota. After treatment with irinotecan, for example, the number of beneficial bacteria such as Lactobacillus spp. and Bifidobacterium spp. decreases, while the amount of Staphylococcus spp., Clostridium spp., and E. coli increases (2323. Stringer AM, Gibson RJ, Logan RM, Bowen JM, Yeoh AS, Burns J, et al. Chemotherapy-induced diarrhea is associated with changes in the luminal environment in the DA rat. Exp Biol Med (Maywood) 2007; 232: 96-106.). Additionally, 5-FU increases anaerobic bacteria in the oral cavity and facultative anaerobes in the large intestine, culminating with mucositis development (77. Touchefeu Y, Montassier E, Nieman K, Gastinne T, Potel G, Bruley Des Varannes S, et al. 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, doi: 10.1111/apt.12878.
https://doi.org/10.1111/apt.12878...
,2424. Secombe KR, Crame EE, Tam JSY, Wardill HR, Gibson RJ, Coller JK, et al. Intestinal toll-like receptor 4 knockout alters the functional capacity of the gut microbiome following irinotecan treatment. Cancer Chemother Pharmacol 2022; 89: 275-281, doi: 10.1007/s00280-021-04382-3.
https://doi.org/10.1007/s00280-021-04382...
). Notably, the presence of bacteria increases in the cervical and mesenteric lymph nodes (2525. Von Bültzingslöwen I, Adlerberth I, Wold AE, Dahlén G, Jontell M. Oral and intestinal microflora in 5-fluorouracil treated rats, translocation to cervical and mesenteric lymph nodes and effects of probiotic bacteria. Oral Microbiol Immunol 2003; 18: 278-284, doi: 10.1034/j.1399-302X.2003.00075.x.
https://doi.org/10.1034/j.1399-302X.2003...
). Stringer et al. (2323. Stringer AM, Gibson RJ, Logan RM, Bowen JM, Yeoh AS, Burns J, et al. Chemotherapy-induced diarrhea is associated with changes in the luminal environment in the DA rat. Exp Biol Med (Maywood) 2007; 232: 96-106.), treating mice with a single dose of irinotecan, observed an increase in Escherichia spp., Clostridium spp., Enterococcus spp., Serratia spp., and Staphylococcus spp. in the microbiota of the stomach, intestine, and colon. In stool, there was an increase of Proteus spp., Clostridium spp., and Peptostreptococcus spp., along with a decrease in Bacillus spp., and Bifidobacterium spp.

The incidence of dysbiosis, though, is described for other antitumor agents. In a study of patients with non-Hodgkin's lymphoma treated with Carmustine, Etoposide, Aracytine, or Melphalan, the feces of these patients were analyzed before and after chemotherapy. Chemotherapy caused qualitative changes in the composition of the microbiota, with a reduction in bacteria of the Firmicutes and Actinobacteria phylum and an increase in the genera Clostridium, Bifidobacterium, Citrobacter, Klebsiella, Enterococcus, Megasphaera, and Parabacteroides (2626. Montassier E, Gastinne T, Vangay P, Al-Ghalith GA, Bruley des Varannes S, Massart S, et al. Chemotherapy-driven dysbiosis in the intestinal microbiome. Aliment Pharmacol Ther 2015; 42: 515-528, doi: 10.1111/apt.13302.
https://doi.org/10.1111/apt.13302...
). In another study, paclitaxel-treated mice showed reduced fecal and colonic microbiota diversity but a relative increase in the presence of the Lactobacillus genus (2727. Loman BR, Jordan KR, Haynes B, Bailey MT, Pyter LM. Chemotherapy-induced neuroinflammation is associated with disrupted colonic and bacterial homeostasis in female mice. Sci Rep 2019; 9: 16490, doi: 10.1038/s41598-019-52893-0.
https://doi.org/10.1038/s41598-019-52893...
). The relationship between unbalanced gut microbiota and chemotherapy-derived gastrointestinal toxicities strongly points toward solving dysbiosis as a strategy to prevent or treat such side effects.

Effects of probiotics on the gastrointestinal toxicities of cancer chemotherapy

To re-establish the balance of the gut microbiota, the use of probiotics is an option. Some studies reveal that the incorporation of probiotics as a therapeutic alternative to prevent or improve the gastrointestinal symptoms of chemotherapy is an approach that shows favorable outcomes. In order to deliver good effects, probiotic use must start at least one month before the beginning of antineoplastic therapy. Among the types of therapy, probiotics have shown to be more effective in preventing the intestinal effects induced by radiotherapy than chemotherapy (2828. Thomsen M, Vitetta L. Adjunctive treatments for the prevention of chemotherapy- and radiotherapy-induced mucositis. Integr Cancer Ther 2018; 17: 1027-1047, doi: 10.1177/1534735418794885.
https://doi.org/10.1177/1534735418794885...
).

The Colon DophilusTM probiotic (composed of Bifidobacterium breve, Bifidobacterium bifidum, Bifidobacterium longum, Lactobacillus rhamnosus, Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus plantarum, Streptococcus thermophilus, Lactobacillus brevis, and Bifidobacterium infantis) given for 12 weeks to patients with metastatic colorectal cancer treated with irinotecan reduced diarrhea severity and decreased the overall incidence of diarrhea and enterocolitis (2929. Mego M, Chovanec J, Vochyanova-Andrezalova I, Konkolovsky P, Mikulova M, Reckova M, et al. Prevention of irinotecan induced diarrhea by probiotics: A randomized double blind, placebo controlled pilot study. Complement Ther Med 2015; 23: 356-362, doi: 10.1016/j.ctim.2015.03.008.
https://doi.org/10.1016/j.ctim.2015.03.0...
). Additionally, abdominal bloating was more frequently reported by patients receiving a placebo compared to the probiotic-treated group. Patients who ingested the formula also reported less use of loperamide and atropine for the symptomatic treatment of diarrhea (2929. Mego M, Chovanec J, Vochyanova-Andrezalova I, Konkolovsky P, Mikulova M, Reckova M, et al. Prevention of irinotecan induced diarrhea by probiotics: A randomized double blind, placebo controlled pilot study. Complement Ther Med 2015; 23: 356-362, doi: 10.1016/j.ctim.2015.03.008.
https://doi.org/10.1016/j.ctim.2015.03.0...
). In children with acute leukemia presenting chemotherapy-related gastrointestinal symptoms, using a Lactobacillus rhamnosus probiotic at a concentration of 5×109 was found to be safe and effective, preventing the development of diarrhea and reducing abdominal distention, constipation, nausea, and meteorism (3030. Reyna-Figueroa J, Barrón-Calvillo E, García-Parra C, Galindo-Delgado P, Contreras-Ochoa C, Lagunas-Martínez A, et al. Probiotic supplementation decreases chemotherapy-induced gastrointestinal side effects in patients with acute leukemia. J Pediatr Hematol Oncol 2019; 41: 468-472, doi: 10.1097/MPH.0000000000001497.
https://doi.org/10.1097/MPH.000000000000...
).

Another study followed up 52 patients with colorectal cancer who consumed probiotics containing six strains of Lactobacillus and Bifidobacterium for six months, starting one month after surgery (3131. Zaharuddin L, Mokhtar NM, Muhammad Nawawi KN, Raja Ali RA. A randomized double-blind placebo-controlled trial of probiotics in post-surgical colorectal cancer. BMC Gastroenterol 2019; 19: 131, doi: 10.1186/s12876-019-1047-4.
https://doi.org/10.1186/s12876-019-1047-...
). It demonstrated that despite probiotics reducing the cytokines TNF-α, IL-17A, IL-17C, IL-22, IL-10, and IL-12, there was no difference in diarrhea incidence compared with the group that received a placebo. However, using probiotics was shown to be safe for those patients (3131. Zaharuddin L, Mokhtar NM, Muhammad Nawawi KN, Raja Ali RA. A randomized double-blind placebo-controlled trial of probiotics in post-surgical colorectal cancer. BMC Gastroenterol 2019; 19: 131, doi: 10.1186/s12876-019-1047-4.
https://doi.org/10.1186/s12876-019-1047-...
). In another study, 150 patients treated with 5-FU using a Lactobacillus rhamnosus GG probiotic showed reduced diarrhea frequency and less abdominal discomfort, contributing to sustaining the chemotherapeutic dose intensity. Furthermore, the probiotic did not trigger adverse effects, and no bacteria were detected in the blood cultures of the patients (3232. Osterlund P, Ruotsalainen T, Korpela R, Saxelin M, Ollus A, Valta P, et al. Lactobacillus supplementation for diarrhoea related to chemotherapy of colorectal cancer: a randomised study. Br J Cancer 2007; 97: 1028-1034, doi: 10.1038/sj.bjc.6603990.
https://doi.org/10.1038/sj.bjc.6603990...
).

Rodents administered with 5-FU present intestinal damage in a mechanism dependent on the full activation of toll-like receptor types 2 and 4 (TLR2 and TLR4) by pathogenic Gram-negative bacteria. TLR2 and TLR4, in turn, stimulate NF-κB and the release of pro-inflammatory cytokines that culminates with the manifestation of gastrointestinal symptoms such as nausea, pain, and diarrhea (2323. Stringer AM, Gibson RJ, Logan RM, Bowen JM, Yeoh AS, Burns J, et al. Chemotherapy-induced diarrhea is associated with changes in the luminal environment in the DA rat. Exp Biol Med (Maywood) 2007; 232: 96-106.,3333. Justino PFC, Melo LFM, Nogueira AF, Costa JVG, Silva LMN, Santos CM, et al. Treatment with Saccharomyces boulardii reduces the inflammation and dysfunction of the gastrointestinal tract in 5-fluorouracil-induced intestinal mucositis in mice. Br J Nutr 2014; 111: 1611-1621, doi: 10.1017/S0007114513004248.
https://doi.org/10.1017/S000711451300424...
). Conversely, prophylactic use of Saccharomyces boulardii probiotic prevents gastrointestinal dysfunction, such as changes in gastric emptying, absorption, permeability, and intestinal transit (3333. Justino PFC, Melo LFM, Nogueira AF, Costa JVG, Silva LMN, Santos CM, et al. Treatment with Saccharomyces boulardii reduces the inflammation and dysfunction of the gastrointestinal tract in 5-fluorouracil-induced intestinal mucositis in mice. Br J Nutr 2014; 111: 1611-1621, doi: 10.1017/S0007114513004248.
https://doi.org/10.1017/S000711451300424...
). It also reduces neutrophil infiltration and expression of inflammatory markers (3434. Justino PFC, Franco AX, Pontier-Bres R, Monteiro CES, Barbosa ALR, Souza MHLP, et al. Modulation of 5-fluorouracil activation of toll-like/MyD88/NF-κB/MAPK pathway by Saccharomyces boulardii CNCM I-745 probiotic. Cytokine 2020; 125: 154791, doi: 10.1016/j.cyto.2019.154791.
https://doi.org/10.1016/j.cyto.2019.1547...
). Similar effects were obtained with Lactobacillus acidophilus probiotic, whose administration started on the day of intestinal mucositis induction with 5-FU (3535. Justino PFC, Melo LFM, Nogueira AF, Morais CM, Mendes WO, Franco AX, et al. Regulatory role of Lactobacillus acidophilus on inflammation and gastric dysmotility in intestinal mucositis induced by 5-fluorouracil in mice. Cancer Chemother Pharmacol 2015; 75: 559-567, doi: 10.1007/s00280-014-2663-x.
https://doi.org/10.1007/s00280-014-2663-...
). The probiotic VSL#3 (a combination of Lactobacillus plantarum, Lactobacillus casei, Lactobacillus acidophilus, Lactobacillus delbrueckii subspecies bulgaricus, Bifidobacterium infantis, Bifidobacterium longum, Bifidobacterium breve, and Streptococcus salivarius subspecies thermophilus) exerts benefits when administered before and after treatment of mice with irinotecan. It reduces chemotherapy-induced diarrhea through three mechanisms: increasing proliferation of epithelial cells, reducing apoptosis, and preventing the increase in caliciform cells and mucin secretion (3636. Bowen JM, Stringer AM, Gibson RJ, Yeoh ASJ, Hannam S, Keefe DMK. VSL#3 probiotic treatment reduces chemotherapy-induced diarrhea and weight loss. Cancer Biol Ther 2007; 6: 1449-1454, doi: 10.4161/cbt.6.9.4622.
https://doi.org/10.4161/cbt.6.9.4622...
). Probiotics containing Lactobacillus acidophilus and Bifidobacterium animalis subspecies lactis have also been shown to reduce the incidence and severity of radiotherapy-induced diarrhea and abdominal pain. Remarkably, loperamide is the only anti-diarrheic medication used for controlling chemotherapy-associated diarrhea, but with limited effectiveness (3737. Linn YH, Thu KK, Win NHH. Effect of probiotics for the prevention of acute radiation-induced diarrhoea among cervical cancer patients: a randomized double-blind placebo-controlled study. Probiotics Antimicrob Proteins 2019; 11: 638-647, doi: 10.1007/s12602-018-9408-9.
https://doi.org/10.1007/s12602-018-9408-...
).

Are probiotics safe?

The quality control in the development and production of bacteria-derived products merits improvement. Better strain identification is highly desirable to avoid contaminants, like pathogens, that may compromise the safety of these compounds (3838. Kolaček S, Hojsak I, Canani RB, Guarino A, Indrio F, Orel R, et al. Commercial probiotic products: a call for improved quality control. a position paper by the ESPGHAN working group for probiotics and prebiotics. J Pediatr Gastroenterol Nutr 2017; 65: 117-124, doi: 10.1097/MPG.0000000000001603.
https://doi.org/10.1097/MPG.000000000000...
). According to a report jointly published by the World Health Organization (WHO) and the Food and Agriculture Organization of the United Nations (FAO), probiotics can be responsible for four types of side effects: systemic infections, harmful metabolic activities, gene transfer, and excessive immune stimulation in susceptible individuals (3939. Doron S, Snydman DR. Risk and safety of probiotics. Clin Infect Dis 2015; 60: S129-S134, doi: 10.1093/cid/civ085.
https://doi.org/10.1093/cid/civ085...
). Most cases of probiotic-derived adverse events are associated with the genera Lactobacillus and Bifidobacterium. They include bacteremia, endocarditis, and liver abscess (Table 2).

Table 2
Probiotics-related side effects.

Among the species that cause bacteremia, Lactobacillus rhamnosus has been one of the most prevalent (4040. Salminen MK, Rautelin H, Tynkkynen S, Poussa T, Saxelin M, Valtonen V, et al. Lactobacillus bacteremia, clinical significance, and patient outcome, with special focus on probiotic L. rhamnosus GG. Clin Infect Dis 2004; 38: 62-69, doi: 10.1086/380455.
https://doi.org/10.1086/380455...
,4141. Salminen MK, Rautelin H, Tynkkynen S, Poussa T, Saxelin M, Valtonen V, et al. Lactobacillus bacteremia, species identification, and antimicrobial susceptibility of 85 blood isolates. Clin Infect Dis 2006; 42: e35-44, doi: 10.1086/500214.
https://doi.org/10.1086/500214...
). Bacteremia was reported in a patient with severe ulcerative colitis using a probiotic containing L. rhamnosus. The same strain was isolated in the patient's blood and had the same vancomycin resistance profile as the strain in the probiotic (4141. Salminen MK, Rautelin H, Tynkkynen S, Poussa T, Saxelin M, Valtonen V, et al. Lactobacillus bacteremia, species identification, and antimicrobial susceptibility of 85 blood isolates. Clin Infect Dis 2006; 42: e35-44, doi: 10.1086/500214.
https://doi.org/10.1086/500214...
). Bacteremia (4242. Meini S, Laureano R, Fani L, Tascini C, Galano A, Antonelli A, et al. Breakthrough Lactobacillus rhamnosus GG bacteremia associated with probiotic use in an adult patient with severe active ulcerative colitis: case report and review of the literature. Infection 2015; 43: 777-781, doi: 10.1007/s15010-015-0798-2.
https://doi.org/10.1007/s15010-015-0798-...
) was also described in a patient with ulcerative colitis treated with corticosteroid and infliximab, who used an L. rhamnosus GG probiotic (4343. Vahabnezhad E, Mochon AB, Wozniak LJ, Ziring DA. Lactobacillus bacteremia associated with probiotic use in a pediatric patient with ulcerative colitis. J Clin Gastroenterol 2013; 47: 437-439, doi: 10.1097/MCG.0b013e318279abf0.
https://doi.org/10.1097/MCG.0b013e318279...
), and in a child with the short gut syndrome (4444. De Groote MA, Frank DN, Dowell E, Glode MP, Pace NR. Lactobacillus rhamnosus GG bacteremia associated with probiotic use in a child with short gut syndrome. Pediatr Infect Dis J 2005; 24: 278-280, doi: 10.1097/01.inf.0000154588.79356.e6.
https://doi.org/10.1097/01.inf.000015458...
). Sometimes, patients developed epidural and retropharyngeal abscesses (4545. Conen A, Zimmerer S, Trampuz A, Frei R, Battegay M, Elzi L. A pain in the neck: probiotics for ulcerative colitis. Ann Int Med 2009; 151: 895-897, doi: 10.7326/0003-4819-151-12-200912150-00020.
https://doi.org/10.7326/0003-4819-151-12...
) or liver abscesses (4646. Rautio M, Jousimies-Somer H, Kauma H, Pietarinen I, Saxelin M, Tynkkynen S, et al. Liver abscess due to a Lactobacillus rhamnosus strain indistinguishable from L. rhamnosus Strain GG. Clin Infect Dis 1999; 28: 1159-1160, doi: 10.1086/514766.
https://doi.org/10.1086/514766...
).

One case of endocarditis was associated with probiotics (composed of Lactobacillus rhamnosus, Streptococcus faecalis, and Lactobacillus acidophilus) used to balance the intestinal microbiota after amoxicillin. That patient presented a positive blood culture for L. rhamnosus (4747. Mackay AD, Taylor MB, Kibbler CC, Hamilton-Miller JMT. Lactobacillus endocarditis caused by a probiotic organism. Clin Microbiol Infect 1999; 5: 290-292, doi: 10.1111/j.1469-0691.1999.tb00144.x.
https://doi.org/10.1111/j.1469-0691.1999...
). Despite being one of the most used genera as a probiotic, Lactobacillus is associated with several cases of endocarditis, most of them in patients who had undergone dental surgery or had some gingival damage (4848. Pasala S, Singer L, Arshad T, Roach K. Lactobacillus endocarditis in a healthy patient with probiotic use. IDCases 2020; 22: e00915, doi: 10.1016/j.idcr.2020.e00915.
https://doi.org/10.1016/j.idcr.2020.e009...
). The presence of damage in other organs can also be a gateway for these microorganisms to reach the bloodstream, as in a case of liver cirrhosis (4949. Naqvi SSB, Nagendra V, Hofmeyr A. Probiotic related Lactobacillus rhamnosus endocarditis in a patient with liver cirrhosis. IDCases 2018; 13: e00439, doi: 10.1016/j.idcr.2018.e00439.
https://doi.org/10.1016/j.idcr.2018.e004...
) and in a case of a patient with prostate cancer in remission, who habitually consumed probiotics, who presented positive bacteremia for Lactobacillus paracasei after colonoscopy (5050. Franko B, Vaillant M, Recule C, Vautrin E, Brion JP, Pavese P. Lactobacillus paracasei endocarditis in a consumer of probiotics. Med Mal Infect 2013; 43: 171-173, doi: 10.1016/j.medmal.2013.01.007.
https://doi.org/10.1016/j.medmal.2013.01...
).

There are also some reported cases of bacteremia and sepsis development regarding treatment with Bifidobacterium, including premature newborn babies with necrotizing enterocolitis (5151. Esaiassen E, Cavanagh P, Hjerde E, Simonsen GS, Støen R, Klingenberg C. Bifidobacterium longum subspecies infantis bacteremia in 3 extremely preterm infants receiving probiotics. Emerg Infect Dis 2016; 22: 1664-1666, doi: 10.3201/eid2209.160033.
https://doi.org/10.3201/eid2209.160033...
) and a neonate with multiple abdominal organ abnormalities (5252. Sato S, Uchida T, Kuwana S, Sasaki K, Watanabe T, Saito J, et al. Bacteremia induced by Bifidobacterium breve in a newborn with cloacal exstrophy. Pediatr Int 2016; 58: 1226-1228, doi: 10.1111/ped.13103.
https://doi.org/10.1111/ped.13103...
). Additionally, bacteremia was found in a child with heart disease who used probiotics to prevent antibiotic-induced diarrhea (5353. Pruccoli G, Silvestro E, Napoleone CP, Aidala E, Garazzino S, Scolfaro C. Are probiotics safe? Bifidobacterium bacteremia in a child with severe heart failure. Infez Med 2019; 27: 175-178.).

Bacteremia was also found in an immunocompromised individual diagnosed with Acquired Immune Deficiency Syndrome and Hodgkin's disease. The patient, who had completed the first cycle of chemotherapy, was using a probiotic based on L. acidophilus, and cultures obtained from the peripheral blood and the previous catheter site were positive for Lactobacillus spp. (5454. LeDoux D, LaBombardi VJ, Karter D. Lactobacillus acidophilus bacteraemia after use of a probiotic in a patient with AIDS and Hodgkin's disease. Int J STD AIDS 2006; 17: 280-282, doi: 10.1258/095646206776253507.
https://doi.org/10.1258/0956462067762535...
).

These cases support the hypothesis that viable bacteria, although safe for healthy individuals, can be dangerous in immunocompromised subjects, including cancer patients undergoing myelosuppressive chemotherapy. Careful individual assessment is highly demanded since these microorganisms can potentially cause life-threatening infections. Thus, paraprobiotics, composed of inactivated microorganisms, could be considered for immunosuppressed patients.

Biological activities of paraprobiotics

The term paraprobiotic was first used by Taverniti and Guglielmetti (5555. Taverniti V, Guglielmetti S. The immunomodulatory properties of probiotic microorganisms beyond their viability (ghost probiotics: proposal of paraprobiotic concept). Genes Nutr 2011; 6: 261-274, doi: 10.1007/s12263-011-0218-x.
https://doi.org/10.1007/s12263-011-0218-...
) to define non-viable microbial cells or cell fractions that confer human or animal benefits when administered in adequate amounts. Paraprobiotics can be produced by inactivation with heat, high pressure, radiation, and sonication (5656. de Almada CN, Almada CN, Martinez RCR, Sant'Ana AS. Paraprobiotics: Evidences on their ability to modify biological responses, inactivation methods and perspectives on their application in foods. Trends Food Sci Technol 2016; 58: 96-114, doi: 10.1016/j.tifs.2016.09.011.
https://doi.org/10.1016/j.tifs.2016.09.0...
). It contrasts with soluble factors originating from bacterial lysis or secreted by live bacteria, called postbiotics (5757. Aguilar-Toalá JE, Garcia-Varela R, Garcia HS, Mata-Haro V, González-Córdova AF, Vallejo-Cordoba B, et al. Postbiotics: an evolving term within the functional foods field. Trends Food Sci Technol 2018; 75: 105-114, doi: 10.1016/j.tifs.2018.03.009.
https://doi.org/10.1016/j.tifs.2018.03.0...
). These compounds have proven immunomodulatory, antioxidant, and anti-inflammatory activity in in vitro and in vivo assays, but the mechanisms are not fully elucidated (5858. Vallejo-Cordoba B, Castro-López C, García HS, González-Córdova AF, Hernández-Mendoza A. Postbiotics and paraprobiotics: A review of current evidence and emerging trends. Adv Food Nutr Res 2020; 94: 1-34, doi: 10.1016/bs.afnr.2020.06.001.
https://doi.org/10.1016/bs.afnr.2020.06....
).

Although the benefits of using products containing bacteria are independent of cell viability, the mechanisms by which inactivated strains promote their positive effects are still a matter of debate. In a murine model of toxoplasmosis, Lactobacillus casei paraprobiotic increases monocyte chemoattractant protein (MCP-1) production, reduces the percentage of Tregs cells, and parasite load (5959. Salas-Lais AG, Robles-Contreras A, Balderas-López JA, Bautista-de Lucio VM. Immunobiotic and paraprobiotic potential effect of Lactobacillus casei in a systemic toxoplasmosis murine model. Microorganisms 2020; 8: 113, doi: 10.3390/microorganisms8010113.
https://doi.org/10.3390/microorganisms80...
). Lactobacillus paracasei paraprobiotic induces IgA production and the expression of IL-10, IL-21, STAT4, and Bcl-6, inducing follicular Th cells differentiation (6060. Arai S, Iwabuchi N, Takahashi S, Xiao JZ, Abe F, Hachimura S. Orally administered heat-killed Lactobacillus paracasei MCC1849 enhances antigen-specific IgA secretion and induces follicular helper T cells in mice. PLoS One 2018; 13: e0199018, doi: 10.1371/journal.pone.0199018.
https://doi.org/10.1371/journal.pone.019...
). Using Lacticaseibacillus casei in rats promotes the modulation of the microbiota by increasing the proportion of beneficial bacteria compared to harmful ones, prevents the increase in total and low-density lipoprotein cholesterol, and controls insulin resistance in rats (6161. Brandão LR, Alves JLB, da Costa WKA, Ferreira GDAH, de Oliveira MP, da Cruz AG, et al. Live and ultrasound-inactivated: Lacticaseibacillus casei modulate the intestinal microbiota and improve biochemical and cardiovascular parameters in male rats fed a high-fat diet. Food Funct 2021; 12: 5287-5300, doi: 10.1039/D1FO01064F.
https://doi.org/10.1039/D1FO01064F...
). The immunomodulatory effect of Lactobacillus casei Zhan paraprobiotic was described on macrophages, with increased iNOS and IL-6 expression. Unlike the L. casei probiotic, there was increased expression of TLR2, TLR3, TLR4, and TLR9 after six hours of exposure to the non-viable bacteria (6262. Wang Y, Xie J, Wang N, Li Y, Sun X, Zhang Y, et al. Lactobacillus casei Zhang modulate cytokine and Toll-like receptor expression and beneficially regulate poly I: C-induced immune responses in RAW264.7 macrophages. Microbiol Immunol 2013; 57: 54-62, doi: 10.1111/j.1348-0421.516.x.
https://doi.org/10.1111/j.1348-0421.516....
).

In humans, the effects of paraprobiotic use are also noted, especially on the gastrointestinal tract. One hundred and eighteen healthy adults consuming Lactobacillus gasseri CP230 paraprobiotic daily for three weeks underwent daily questionnaires about their quality of life and stool characteristics. The subjects showed modulation of the intestinal microbiota and improvement in the number and odor of stools (6363. Sugawara T, Sawada D, Ishida Y, Aihara K, Aoki Y, Takehara I, et al. Regulatory effect of paraprobiotic Lactobacillus gasseri CP2305 on gut environment and function . Microb Ecol Heal Dis 2016; 27: 30259, doi: 10.3402/mehd.v27.30259.
https://doi.org/10.3402/mehd.v27.30259...
).

Are paraprobiotics suitable for treating chemotherapy-associated diarrhea?

Despite the few reports about paraprobiotics, they are believed to produce similar effects to probiotics. L. paracasei paraprobiotic, when orally administered in mice, induces antigen-specific IgA production in the small intestine, serum, and lungs and increases the proportion of follicular helper T cells in Peyer's patches (6060. Arai S, Iwabuchi N, Takahashi S, Xiao JZ, Abe F, Hachimura S. Orally administered heat-killed Lactobacillus paracasei MCC1849 enhances antigen-specific IgA secretion and induces follicular helper T cells in mice. PLoS One 2018; 13: e0199018, doi: 10.1371/journal.pone.0199018.
https://doi.org/10.1371/journal.pone.019...
). In a model of intestinal obstruction in mice, S. boulardii paraprobiotic promoted immunomodulation with increased production of the anti-inflammatory cytokine IL-10 and reduced intestinal lesions (6060. Arai S, Iwabuchi N, Takahashi S, Xiao JZ, Abe F, Hachimura S. Orally administered heat-killed Lactobacillus paracasei MCC1849 enhances antigen-specific IgA secretion and induces follicular helper T cells in mice. PLoS One 2018; 13: e0199018, doi: 10.1371/journal.pone.0199018.
https://doi.org/10.1371/journal.pone.019...
,6464. Generoso SV, Viana ML, Santos RG, Arantes RME, Martins FS, Nicoli JR, et al. Protection against increased intestinal permeability and bacterial translocation induced by intestinal obstruction in mice treated with viable and heat-killed Saccharomyces boulardii. Eur J Nutr. 2011; 50: 261-269, doi: 10.1007/s00394-010-0134-7.
https://doi.org/10.1007/s00394-010-0134-...
). Brandão et al. (6161. Brandão LR, Alves JLB, da Costa WKA, Ferreira GDAH, de Oliveira MP, da Cruz AG, et al. Live and ultrasound-inactivated: Lacticaseibacillus casei modulate the intestinal microbiota and improve biochemical and cardiovascular parameters in male rats fed a high-fat diet. Food Funct 2021; 12: 5287-5300, doi: 10.1039/D1FO01064F.
https://doi.org/10.1039/D1FO01064F...
) showed that inactivated Lacticaseibacillus casei bacteria modulate the intestinal microbiota of mice, increasing the proportion of beneficial bacteria (Lachnospiraceae and Ruminoccocaceae) and reducing the number of harmful species, such as Clostridiaceae, Enterobacteriaceae, and Helicobacteriacea. Enterococcus faecalis paraprobiotic administration for one week before chemotherapy reduced the intestinal damage in the ileum of irinotecan-injected mice. It also reduced the presence of neutrophils and macrophages and abolished bacteremia. The mechanism involved the maintenance of zonula occludens protein integrity (6565. Nobre LMS, Lopes MHS, Geraix J, Cajado AG, Silva JMR, Ribeiro LR, et al. Paraprobiotic Enterococcus faecalis EC-12 prevents the development of irinotecan-induced intestinal mucositis in mice. Life Sci 2022; 296: 120445, doi: 10.1016/j.lfs.2022.120445.
https://doi.org/10.1016/j.lfs.2022.12044...
).

Dead bacteria and non-viable fungi from Saccharomyces boulardii were shown to prevent bacterial translocation and increase intestinal permeability in a murine model of intestinal obstruction. Besides increasing cytokine IL-10 and IgA levels, S. boulardii paraprobiotic reduces intestinal mucosal lesions in these animals (6464. Generoso SV, Viana ML, Santos RG, Arantes RME, Martins FS, Nicoli JR, et al. Protection against increased intestinal permeability and bacterial translocation induced by intestinal obstruction in mice treated with viable and heat-killed Saccharomyces boulardii. Eur J Nutr. 2011; 50: 261-269, doi: 10.1007/s00394-010-0134-7.
https://doi.org/10.1007/s00394-010-0134-...
). One study further reported that the daily use of Lactobacillus gasseri CP230 paraprobiotic improves the quality of life of healthy individuals by modulating the intestinal microbiota, the number of bowel movements, and stool odor (6363. Sugawara T, Sawada D, Ishida Y, Aihara K, Aoki Y, Takehara I, et al. Regulatory effect of paraprobiotic Lactobacillus gasseri CP2305 on gut environment and function . Microb Ecol Heal Dis 2016; 27: 30259, doi: 10.3402/mehd.v27.30259.
https://doi.org/10.3402/mehd.v27.30259...
). These studies provide evidence for using paraprobiotics to manage chemo- and radiotherapy-induced diarrhea. They may be as effective as probiotics and have a better safety profile.

Conclusions

Despite probiotics' efficacy in modulating intestinal microbiota, there is a growing concern regarding their use in immunocompromised subjects. The literature provides novel evidence supporting the indication of paraprobiotics with no loss of effectiveness compared with probiotics to control chemotherapy-associated dysbiosis. Conversely, the benefits of other bacterial strains to compose paraprobiotic-based formulations need to be validated. Producing paraprobiotics formulations is feasible and facilitates the applicability of such compounds in the clinical setting. Additionally, the potential advantages of paraprobiotics include the safety profile and reduction in the risk of systemic infection and transference of antimicrobial resistance mechanisms. Clinical trials are warranted to delineate rational therapeutic protocols based on paraprobiotics to manage the gastrointestinal manifestations of cancer treatment toxicities.

Acknowledgments

This work was supported by EBSERH (Empresa Brasileira de Serviços Hospitalares) and CNPq (Conselho Nacional de Desenvolvimento Científico e Tecnológico), CNPq/MCTI/FNDCT, grant number: 407381/2021-0; CAPES (Fundação Coordenação de Aperfeiçoamento de Pessoal de Nível Superior, project number 0756/2020 PROEX/CAPES, grant number 23038.006689/2020-14); and FUNCAP (Fundação Cearense de Apoio ao Desenvolvimento Científico, grant number: PR2-0101-00054.01.00/15 [PRONEX/FUNCAP/CNPq], and P20-0171-00121.01.00/20 [FUNCAP-SESA-Decit/SCTIE/MS-CNPq]). R.C.P. Lima-Júnior is a CNPq Research Productivity Fellow (grant number: 314904/2020-4).

References

  • 1
    Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin 2016; 66: 7-30, doi: 10.3322/caac.21332.
    » https://doi.org/10.3322/caac.21332
  • 2
    Lee YT, Tan YJ, Oon CE. Molecular targeted therapy: treating cancer with specificity. Eur J Pharmacol 2018; 834: 188-196, doi: 10.1016/j.ejphar.2018.07.034.
    » https://doi.org/10.1016/j.ejphar.2018.07.034
  • 3
    WHO (World Health Organization). Cancer. Geneva: World Health Organization; 2021.
  • 4
    Wallington M, Saxon EB, Bomb M, Smittenaar R, Wickenden M, McPhail S, et al. 30-day mortality after systemic anticancer treatment for breast and lung cancer in England: a population-based, observational study. Lancet Oncol 2016; 17: 1203-1216, doi: 10.1016/S1470-2045(16)30383-7.
    » https://doi.org/10.1016/S1470-2045(16)30383-7
  • 5
    Van Sebille YZA, Stansborough R, Wardill HR, Bateman E, Gibson RJ, Keefe DM. Management of mucositis during chemotherapy: from pathophysiology to pragmatic therapeutics. Curr Oncol Rep 2015; 17: 50, doi: 10.1007/s11912-015-0474-9.
    » https://doi.org/10.1007/s11912-015-0474-9
  • 6
    Stringer AM. Interaction between host cells and microbes in chemotherapy-induced mucositis. Nutrients 2013; 5: 1488-1499, doi: 10.3390/nu5051488.
    » https://doi.org/10.3390/nu5051488
  • 7
    Touchefeu Y, Montassier E, Nieman K, Gastinne T, Potel G, Bruley Des Varannes S, et al. 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, doi: 10.1111/apt.12878.
    » https://doi.org/10.1111/apt.12878
  • 8
    Ulluwishewa D, Anderson RC, McNabb WC, Moughan PJ, Wells JM, Roy NC. Regulation of tight junction permeability by intestinal bacteria and dietary components. J Nutr 2011; 141: 769-776, doi: 10.3945/jn.110.135657.
    » https://doi.org/10.3945/jn.110.135657
  • 9
    Lu D, Yan J, Liu F, Ding P, Chen B, Lu Y, et al. Probiotics in preventing and treating chemotherapy-induced diarrhea: a meta-analysis. Asia Pac J Clin Nutr 2019; 28: 701-710, doi: 10.6133/apjcn.201912_28(4).0005.
    » https://doi.org/10.6133/apjcn.201912_28(4).0005
  • 10
    Molska M, Reguła J. Potential mechanisms of probiotics action in the prevention and treatment of colorectal cancer. Nutrients 2019; 11: 2453, doi: 10.3390/nu11102453.
    » https://doi.org/10.3390/nu11102453
  • 11
    Vesterlund S, Vankerckhoven V, Saxelin M, Goossens H, Salminen S, Ouwehand AC. Safety assessment of Lactobacillus strains: presence of putative risk factors in faecal, blood and probiotic isolates. Int J Food Microbiol 2007; 116: 325-331, doi: 10.1016/j.ijfoodmicro.2007.02.002.
    » https://doi.org/10.1016/j.ijfoodmicro.2007.02.002
  • 12
    Aprile G, Rihawi K, De Carlo E, Sonis ST. Treatment-related gastrointestinal toxicities and advanced colorectal or pancreatic cancer: a critical update. World J Gastroenterol 2015; 21: 11793-11803, doi: 10.3748/wjg.v21.i41.11793.
    » https://doi.org/10.3748/wjg.v21.i41.11793
  • 13
    Stein A, Voigt W, Jordan K. Chemotherapy-induced diarrhea: pathophysiology, frequency and guideline-based management. Ther Adv Med Oncol 2010; 2: 51-63, doi: 10.1177/1758834009355164.
    » https://doi.org/10.1177/1758834009355164
  • 14
    Ghoreishi Z, Shidfar F, Iravani M, Esfahani A, Ghavamzadeh A. Effect of vitamin E on chemotherapy-induced mucositis and neutropenia in leukemic patients undergoing bone marrow transplantation. Asia Pac J Clin Oncol 2007; 3: 113-118, doi: 10.1111/j.1743-7563.2007.00111.x.
    » https://doi.org/10.1111/j.1743-7563.2007.00111.x
  • 15
    Viele CS. Overview of chemotherapy-induced diarrhea. Semin Oncol Nurs 2003; 19: 2-5, doi: 10.1053/j.soncn.2003.09.007.
    » https://doi.org/10.1053/j.soncn.2003.09.007
  • 16
    Saltz LB. Understanding and managing chemotherapy-induced diarrhea. J Support Oncol 2003; 1: 35-46; discussion 38-41, 45-46.
  • 17
    Ribeiro RA, Wanderley CWS, Wong DVT, Mota JMSC, Leite CAVG, Souza MHLP, et al. Irinotecan- and 5-fluorouracil-induced intestinal mucositis: insights into pathogenesis and therapeutic perspectives. Cancer Chemother Pharmacol 2016; 78: 881-893, doi: 10.1007/s00280-016-3139-y.
    » https://doi.org/10.1007/s00280-016-3139-y
  • 18
    Gibson RJ, Keefe DMK. Cancer chemotherapy-induced diarrhoea and constipation: mechanisms of damage and prevention strategies. Support Care Cancer 2006; 14: 890-900, doi: 10.1007/s00520-006-0040-y.
    » https://doi.org/10.1007/s00520-006-0040-y
  • 19
    Ikuno N, Soda H, Watanabe M, Oka M. Irinotecan (CPT-11) and characteristic mucosal changes in the mouse ileum and cecum. J Natl Cancer Inst 1995; 87: 1876-1883, doi: 10.1093/jnci/87.24.1876.
    » https://doi.org/10.1093/jnci/87.24.1876
  • 20
    Gibson RJ, Bowen JM, Inglis MRB, Cummins AG, Keefe DMK. Irinotecan causes severe small intestinal damage, as well as colonic damage, in the rat with implanted breast cancer. J Gastroenterol Hepatol 2003; 18: 1095-1100, doi: 10.1046/j.1440-1746.2003.03136.x.
    » https://doi.org/10.1046/j.1440-1746.2003.03136.x
  • 21
    Deleemans JM, Chleilat F, Reimer RA, Baydoun M, Piedalue KA, Lowry DE, et al. The chemo-gut pilot study: associations between gut microbiota, gastrointestinal symptoms, and psychosocial health outcomes in a cross-sectional sample of young adult cancer survivors. Curr Oncol 2022; 29: 2973-2994, doi: 10.3390/curroncol29050243.
    » https://doi.org/10.3390/curroncol29050243
  • 22
    Akbarali HI, Muchhala KH, Jessup DK, Cheatham S. Chemotherapy induced gastrointestinal toxicities. Adv Cancer Res 2022; 155: 131-166, doi: 10.1016/bs.acr.2022.02.007.
    » https://doi.org/10.1016/bs.acr.2022.02.007
  • 23
    Stringer AM, Gibson RJ, Logan RM, Bowen JM, Yeoh AS, Burns J, et al. Chemotherapy-induced diarrhea is associated with changes in the luminal environment in the DA rat. Exp Biol Med (Maywood) 2007; 232: 96-106.
  • 24
    Secombe KR, Crame EE, Tam JSY, Wardill HR, Gibson RJ, Coller JK, et al. Intestinal toll-like receptor 4 knockout alters the functional capacity of the gut microbiome following irinotecan treatment. Cancer Chemother Pharmacol 2022; 89: 275-281, doi: 10.1007/s00280-021-04382-3.
    » https://doi.org/10.1007/s00280-021-04382-3
  • 25
    Von Bültzingslöwen I, Adlerberth I, Wold AE, Dahlén G, Jontell M. Oral and intestinal microflora in 5-fluorouracil treated rats, translocation to cervical and mesenteric lymph nodes and effects of probiotic bacteria. Oral Microbiol Immunol 2003; 18: 278-284, doi: 10.1034/j.1399-302X.2003.00075.x.
    » https://doi.org/10.1034/j.1399-302X.2003.00075.x
  • 26
    Montassier E, Gastinne T, Vangay P, Al-Ghalith GA, Bruley des Varannes S, Massart S, et al. Chemotherapy-driven dysbiosis in the intestinal microbiome. Aliment Pharmacol Ther 2015; 42: 515-528, doi: 10.1111/apt.13302.
    » https://doi.org/10.1111/apt.13302
  • 27
    Loman BR, Jordan KR, Haynes B, Bailey MT, Pyter LM. Chemotherapy-induced neuroinflammation is associated with disrupted colonic and bacterial homeostasis in female mice. Sci Rep 2019; 9: 16490, doi: 10.1038/s41598-019-52893-0.
    » https://doi.org/10.1038/s41598-019-52893-0
  • 28
    Thomsen M, Vitetta L. Adjunctive treatments for the prevention of chemotherapy- and radiotherapy-induced mucositis. Integr Cancer Ther 2018; 17: 1027-1047, doi: 10.1177/1534735418794885.
    » https://doi.org/10.1177/1534735418794885
  • 29
    Mego M, Chovanec J, Vochyanova-Andrezalova I, Konkolovsky P, Mikulova M, Reckova M, et al. Prevention of irinotecan induced diarrhea by probiotics: A randomized double blind, placebo controlled pilot study. Complement Ther Med 2015; 23: 356-362, doi: 10.1016/j.ctim.2015.03.008.
    » https://doi.org/10.1016/j.ctim.2015.03.008
  • 30
    Reyna-Figueroa J, Barrón-Calvillo E, García-Parra C, Galindo-Delgado P, Contreras-Ochoa C, Lagunas-Martínez A, et al. Probiotic supplementation decreases chemotherapy-induced gastrointestinal side effects in patients with acute leukemia. J Pediatr Hematol Oncol 2019; 41: 468-472, doi: 10.1097/MPH.0000000000001497.
    » https://doi.org/10.1097/MPH.0000000000001497
  • 31
    Zaharuddin L, Mokhtar NM, Muhammad Nawawi KN, Raja Ali RA. A randomized double-blind placebo-controlled trial of probiotics in post-surgical colorectal cancer. BMC Gastroenterol 2019; 19: 131, doi: 10.1186/s12876-019-1047-4.
    » https://doi.org/10.1186/s12876-019-1047-4
  • 32
    Osterlund P, Ruotsalainen T, Korpela R, Saxelin M, Ollus A, Valta P, et al. Lactobacillus supplementation for diarrhoea related to chemotherapy of colorectal cancer: a randomised study. Br J Cancer 2007; 97: 1028-1034, doi: 10.1038/sj.bjc.6603990.
    » https://doi.org/10.1038/sj.bjc.6603990
  • 33
    Justino PFC, Melo LFM, Nogueira AF, Costa JVG, Silva LMN, Santos CM, et al. Treatment with Saccharomyces boulardii reduces the inflammation and dysfunction of the gastrointestinal tract in 5-fluorouracil-induced intestinal mucositis in mice. Br J Nutr 2014; 111: 1611-1621, doi: 10.1017/S0007114513004248.
    » https://doi.org/10.1017/S0007114513004248
  • 34
    Justino PFC, Franco AX, Pontier-Bres R, Monteiro CES, Barbosa ALR, Souza MHLP, et al. Modulation of 5-fluorouracil activation of toll-like/MyD88/NF-κB/MAPK pathway by Saccharomyces boulardii CNCM I-745 probiotic. Cytokine 2020; 125: 154791, doi: 10.1016/j.cyto.2019.154791.
    » https://doi.org/10.1016/j.cyto.2019.154791
  • 35
    Justino PFC, Melo LFM, Nogueira AF, Morais CM, Mendes WO, Franco AX, et al. Regulatory role of Lactobacillus acidophilus on inflammation and gastric dysmotility in intestinal mucositis induced by 5-fluorouracil in mice. Cancer Chemother Pharmacol 2015; 75: 559-567, doi: 10.1007/s00280-014-2663-x.
    » https://doi.org/10.1007/s00280-014-2663-x
  • 36
    Bowen JM, Stringer AM, Gibson RJ, Yeoh ASJ, Hannam S, Keefe DMK. VSL#3 probiotic treatment reduces chemotherapy-induced diarrhea and weight loss. Cancer Biol Ther 2007; 6: 1449-1454, doi: 10.4161/cbt.6.9.4622.
    » https://doi.org/10.4161/cbt.6.9.4622
  • 37
    Linn YH, Thu KK, Win NHH. Effect of probiotics for the prevention of acute radiation-induced diarrhoea among cervical cancer patients: a randomized double-blind placebo-controlled study. Probiotics Antimicrob Proteins 2019; 11: 638-647, doi: 10.1007/s12602-018-9408-9.
    » https://doi.org/10.1007/s12602-018-9408-9
  • 38
    Kolaček S, Hojsak I, Canani RB, Guarino A, Indrio F, Orel R, et al. Commercial probiotic products: a call for improved quality control. a position paper by the ESPGHAN working group for probiotics and prebiotics. J Pediatr Gastroenterol Nutr 2017; 65: 117-124, doi: 10.1097/MPG.0000000000001603.
    » https://doi.org/10.1097/MPG.0000000000001603
  • 39
    Doron S, Snydman DR. Risk and safety of probiotics. Clin Infect Dis 2015; 60: S129-S134, doi: 10.1093/cid/civ085.
    » https://doi.org/10.1093/cid/civ085
  • 40
    Salminen MK, Rautelin H, Tynkkynen S, Poussa T, Saxelin M, Valtonen V, et al. Lactobacillus bacteremia, clinical significance, and patient outcome, with special focus on probiotic L. rhamnosus GG. Clin Infect Dis 2004; 38: 62-69, doi: 10.1086/380455.
    » https://doi.org/10.1086/380455
  • 41
    Salminen MK, Rautelin H, Tynkkynen S, Poussa T, Saxelin M, Valtonen V, et al. Lactobacillus bacteremia, species identification, and antimicrobial susceptibility of 85 blood isolates. Clin Infect Dis 2006; 42: e35-44, doi: 10.1086/500214.
    » https://doi.org/10.1086/500214
  • 42
    Meini S, Laureano R, Fani L, Tascini C, Galano A, Antonelli A, et al. Breakthrough Lactobacillus rhamnosus GG bacteremia associated with probiotic use in an adult patient with severe active ulcerative colitis: case report and review of the literature. Infection 2015; 43: 777-781, doi: 10.1007/s15010-015-0798-2.
    » https://doi.org/10.1007/s15010-015-0798-2
  • 43
    Vahabnezhad E, Mochon AB, Wozniak LJ, Ziring DA. Lactobacillus bacteremia associated with probiotic use in a pediatric patient with ulcerative colitis. J Clin Gastroenterol 2013; 47: 437-439, doi: 10.1097/MCG.0b013e318279abf0.
    » https://doi.org/10.1097/MCG.0b013e318279abf0
  • 44
    De Groote MA, Frank DN, Dowell E, Glode MP, Pace NR. Lactobacillus rhamnosus GG bacteremia associated with probiotic use in a child with short gut syndrome. Pediatr Infect Dis J 2005; 24: 278-280, doi: 10.1097/01.inf.0000154588.79356.e6.
    » https://doi.org/10.1097/01.inf.0000154588.79356.e6
  • 45
    Conen A, Zimmerer S, Trampuz A, Frei R, Battegay M, Elzi L. A pain in the neck: probiotics for ulcerative colitis. Ann Int Med 2009; 151: 895-897, doi: 10.7326/0003-4819-151-12-200912150-00020.
    » https://doi.org/10.7326/0003-4819-151-12-200912150-00020
  • 46
    Rautio M, Jousimies-Somer H, Kauma H, Pietarinen I, Saxelin M, Tynkkynen S, et al. Liver abscess due to a Lactobacillus rhamnosus strain indistinguishable from L. rhamnosus Strain GG. Clin Infect Dis 1999; 28: 1159-1160, doi: 10.1086/514766.
    » https://doi.org/10.1086/514766
  • 47
    Mackay AD, Taylor MB, Kibbler CC, Hamilton-Miller JMT. Lactobacillus endocarditis caused by a probiotic organism. Clin Microbiol Infect 1999; 5: 290-292, doi: 10.1111/j.1469-0691.1999.tb00144.x.
    » https://doi.org/10.1111/j.1469-0691.1999.tb00144.x
  • 48
    Pasala S, Singer L, Arshad T, Roach K. Lactobacillus endocarditis in a healthy patient with probiotic use. IDCases 2020; 22: e00915, doi: 10.1016/j.idcr.2020.e00915.
    » https://doi.org/10.1016/j.idcr.2020.e00915
  • 49
    Naqvi SSB, Nagendra V, Hofmeyr A. Probiotic related Lactobacillus rhamnosus endocarditis in a patient with liver cirrhosis. IDCases 2018; 13: e00439, doi: 10.1016/j.idcr.2018.e00439.
    » https://doi.org/10.1016/j.idcr.2018.e00439
  • 50
    Franko B, Vaillant M, Recule C, Vautrin E, Brion JP, Pavese P. Lactobacillus paracasei endocarditis in a consumer of probiotics. Med Mal Infect 2013; 43: 171-173, doi: 10.1016/j.medmal.2013.01.007.
    » https://doi.org/10.1016/j.medmal.2013.01.007
  • 51
    Esaiassen E, Cavanagh P, Hjerde E, Simonsen GS, Støen R, Klingenberg C. Bifidobacterium longum subspecies infantis bacteremia in 3 extremely preterm infants receiving probiotics. Emerg Infect Dis 2016; 22: 1664-1666, doi: 10.3201/eid2209.160033.
    » https://doi.org/10.3201/eid2209.160033
  • 52
    Sato S, Uchida T, Kuwana S, Sasaki K, Watanabe T, Saito J, et al. Bacteremia induced by Bifidobacterium breve in a newborn with cloacal exstrophy. Pediatr Int 2016; 58: 1226-1228, doi: 10.1111/ped.13103.
    » https://doi.org/10.1111/ped.13103
  • 53
    Pruccoli G, Silvestro E, Napoleone CP, Aidala E, Garazzino S, Scolfaro C. Are probiotics safe? Bifidobacterium bacteremia in a child with severe heart failure. Infez Med 2019; 27: 175-178.
  • 54
    LeDoux D, LaBombardi VJ, Karter D. Lactobacillus acidophilus bacteraemia after use of a probiotic in a patient with AIDS and Hodgkin's disease. Int J STD AIDS 2006; 17: 280-282, doi: 10.1258/095646206776253507.
    » https://doi.org/10.1258/095646206776253507
  • 55
    Taverniti V, Guglielmetti S. The immunomodulatory properties of probiotic microorganisms beyond their viability (ghost probiotics: proposal of paraprobiotic concept). Genes Nutr 2011; 6: 261-274, doi: 10.1007/s12263-011-0218-x.
    » https://doi.org/10.1007/s12263-011-0218-x
  • 56
    de Almada CN, Almada CN, Martinez RCR, Sant'Ana AS. Paraprobiotics: Evidences on their ability to modify biological responses, inactivation methods and perspectives on their application in foods. Trends Food Sci Technol 2016; 58: 96-114, doi: 10.1016/j.tifs.2016.09.011.
    » https://doi.org/10.1016/j.tifs.2016.09.011
  • 57
    Aguilar-Toalá JE, Garcia-Varela R, Garcia HS, Mata-Haro V, González-Córdova AF, Vallejo-Cordoba B, et al. Postbiotics: an evolving term within the functional foods field. Trends Food Sci Technol 2018; 75: 105-114, doi: 10.1016/j.tifs.2018.03.009.
    » https://doi.org/10.1016/j.tifs.2018.03.009
  • 58
    Vallejo-Cordoba B, Castro-López C, García HS, González-Córdova AF, Hernández-Mendoza A. Postbiotics and paraprobiotics: A review of current evidence and emerging trends. Adv Food Nutr Res 2020; 94: 1-34, doi: 10.1016/bs.afnr.2020.06.001.
    » https://doi.org/10.1016/bs.afnr.2020.06.001
  • 59
    Salas-Lais AG, Robles-Contreras A, Balderas-López JA, Bautista-de Lucio VM. Immunobiotic and paraprobiotic potential effect of Lactobacillus casei in a systemic toxoplasmosis murine model. Microorganisms 2020; 8: 113, doi: 10.3390/microorganisms8010113.
    » https://doi.org/10.3390/microorganisms8010113
  • 60
    Arai S, Iwabuchi N, Takahashi S, Xiao JZ, Abe F, Hachimura S. Orally administered heat-killed Lactobacillus paracasei MCC1849 enhances antigen-specific IgA secretion and induces follicular helper T cells in mice. PLoS One 2018; 13: e0199018, doi: 10.1371/journal.pone.0199018.
    » https://doi.org/10.1371/journal.pone.0199018
  • 61
    Brandão LR, Alves JLB, da Costa WKA, Ferreira GDAH, de Oliveira MP, da Cruz AG, et al. Live and ultrasound-inactivated: Lacticaseibacillus casei modulate the intestinal microbiota and improve biochemical and cardiovascular parameters in male rats fed a high-fat diet. Food Funct 2021; 12: 5287-5300, doi: 10.1039/D1FO01064F.
    » https://doi.org/10.1039/D1FO01064F
  • 62
    Wang Y, Xie J, Wang N, Li Y, Sun X, Zhang Y, et al. Lactobacillus casei Zhang modulate cytokine and Toll-like receptor expression and beneficially regulate poly I: C-induced immune responses in RAW264.7 macrophages. Microbiol Immunol 2013; 57: 54-62, doi: 10.1111/j.1348-0421.516.x.
    » https://doi.org/10.1111/j.1348-0421.516.x
  • 63
    Sugawara T, Sawada D, Ishida Y, Aihara K, Aoki Y, Takehara I, et al. Regulatory effect of paraprobiotic Lactobacillus gasseri CP2305 on gut environment and function . Microb Ecol Heal Dis 2016; 27: 30259, doi: 10.3402/mehd.v27.30259.
    » https://doi.org/10.3402/mehd.v27.30259
  • 64
    Generoso SV, Viana ML, Santos RG, Arantes RME, Martins FS, Nicoli JR, et al. Protection against increased intestinal permeability and bacterial translocation induced by intestinal obstruction in mice treated with viable and heat-killed Saccharomyces boulardii Eur J Nutr. 2011; 50: 261-269, doi: 10.1007/s00394-010-0134-7.
    » https://doi.org/10.1007/s00394-010-0134-7
  • 65
    Nobre LMS, Lopes MHS, Geraix J, Cajado AG, Silva JMR, Ribeiro LR, et al. Paraprobiotic Enterococcus faecalis EC-12 prevents the development of irinotecan-induced intestinal mucositis in mice. Life Sci 2022; 296: 120445, doi: 10.1016/j.lfs.2022.120445.
    » https://doi.org/10.1016/j.lfs.2022.120445
  • 66
    Vallianou N, Stratigou T, Christodoulatos GS, Tsigalou C, Dalamaga M. Probiotics, prebiotics, synbiotics, postbiotics, and obesity: current evidence, controversies, and perspectives. Curr Obes Rep 2020; 9: 179-192, doi: 10.1007/s13679-020-00379-w.
    » https://doi.org/10.1007/s13679-020-00379-w
  • 67
    Walter J. Ecological role of lactobacilli in the gastrointestinal tract: Implications for fundamental and biomedical research. Appl Environ Microbiol 2008; 74: 4985-4996, doi: 10.1128/AEM.00753-08.
    » https://doi.org/10.1128/AEM.00753-08
  • 68
    Fijan S. Microorganisms with claimed probiotic properties: an overview of recent literature. Int J Environ Res Public Health 2014; 11: 4745-4767, doi: 10.3390/ijerph110504745.
    » https://doi.org/10.3390/ijerph110504745
  • 69
    Choi CH, Jo SY, Park HJ, Chang SK, Byeon JS, Myung SJ. A randomized, double-blind, placebo-controlled multicenter trial of Saccharomyces boulardii in irritable bowel syndrome: effect on quality of life. J Clin Gastroenterol 2011; 45: 679-683, doi: 10.1097/MCG.0b013e318204593e.
    » https://doi.org/10.1097/MCG.0b013e318204593e
  • 70
    Guslandi M, Giollo P, Testoni PA. A pilot trial of Saccharomyces boulardii in ulcerative colitis. Eur J Gastroenterol Hepatol 2003; 15: 697-698, doi: 10.1097/00042737-200306000-00017.
    » https://doi.org/10.1097/00042737-200306000-00017
  • 71
    Lai H, Chiu CH, Kong MS, Chang CJ, Chen CC. Nutrients probiotic Lactobacillus casei: effective for managing childhood diarrhea by altering gut microbiota and attenuating fecal inflammatory markers. Nutrients 2019; 11: 1150, doi: 10.3390/nu11051150.
    » https://doi.org/10.3390/nu11051150
  • 72
    Zhao R, Wang Y, Huang Y, Cui Y, Xia L, Rao Z, et al. Effects of fiber and probiotics on diarrhea associated with enteral nutrition in gastric cancer patients: a prospective randomized and controlled trial. Medicine (Baltimore) 2017; 96: e8418, doi: 10.1097/MD.0000000000008418.
    » https://doi.org/10.1097/MD.0000000000008418
  • 73
    Ishaque SM, Khosruzzaman SM, Ahmed DS, Sah MP. A randomized placebo-controlled clinical trial of a multi-strain probiotic formulation (Bio-Kult®) in the management of diarrhea-predominant irritable bowel syndrome. BMC Gastroenterol 2018; 18: 71, doi: 10.1186/s12876-018-0788-9.
    » https://doi.org/10.1186/s12876-018-0788-9
  • 74
    Kommers MJ, Rodrigues RAS, Miyajima F, Zavala AAZ, Ultramari VRLM, Fett WCR, et al. Effects of probiotic use on quality of life and physical activity in constipated female university students: a randomized, double-blind placebo-controlled study. J Altern Complement Med 2019; 25: 1163-1171, doi: 10.1089/acm.2019.0134.
    » https://doi.org/10.1089/acm.2019.0134
  • 75
    Kijmanawat A, Panburana P, Reutrakul S, Tangshewinsirikul C. Effects of probiotic supplements on insulin resistance in gestational diabetes mellitus: a double-blind randomized controlled trial. J Diabetes Investig 2019; 10: 163-170, doi: 10.1111/jdi.12863.
    » https://doi.org/10.1111/jdi.12863
  • 76
    Kazemi A, Noorbala AA, Azam K, Eskandari MH, Djafarian K. Effect of probiotic and prebiotic vs placebo on psychological outcomes in patients with major depressive disorder: a randomized clinical trial. Clin Nutr 2019; 38: 522-528, doi: 10.1016/j.clnu.2018.04.010.
    » https://doi.org/10.1016/j.clnu.2018.04.010
  • 77
    Teame T, Wang A, Xie M, Zhang Z, Yang Y, Ding Q, et al. Paraprobiotics and postbiotics of probiotic Lactobacilli, their positive effects on the host and action mechanisms: a review. Front Nutr 2020; 7: 570344, doi: 10.3389/fnut.2020.570344.
    » https://doi.org/10.3389/fnut.2020.570344
  • 78
    Orlando A, Refolo MG, Messa C, Amati L, Lavermicocca P, Guerra V, et al. Antiproliferative and proapoptotic effects of viable or heat-killed Lactobacillus paracasei IMPC2.1 and Lactobacillus rhamnosus GG in HGC-27 gastric and DLD-1 colon cell lines. Nutr Cancer 2012; 64: 1103-1111, doi: 10.1080/01635581.2012.717676.
    » https://doi.org/10.1080/01635581.2012.717676
  • 79
    Chung IC, OuYang CN, Yuan SN, Lin HC, Huang KY, Wu PS, et al. Pretreatment with a heat-killed probiotic modulates the NLRP3 inflammasome and attenuates colitis-associated colorectal cancer in mice. Nutrients 2019; 11: 516, doi: 10.3390/nu11030516.
    » https://doi.org/10.3390/nu11030516
  • 80
    Karimi Ardestani S, Tafvizi F, Tajabadi Ebrahimi M. Heat-killed probiotic bacteria induce apoptosis of HT-29 human colon adenocarcinoma cell line via the regulation of Bax/Bcl2 and caspases pathway. Hum Exp Toxicol 2019; 38: 1069-1081, doi: 10.1177/0960327119851255.
    » https://doi.org/10.1177/0960327119851255
  • 81
    Motevaseli E, Khorramizadeh MR, Hadjati J, Bonab SF, Eslami S, Ghafouri-Fard S. Investigation of antitumor effects of Lactobacillus crispatus in experimental model of breast cancer in BALB/c mice. Immunotherapy 2018; 10: 119-129, doi: 10.2217/imt-2017-0088.
    » https://doi.org/10.2217/imt-2017-0088
  • 82
    Seo KH, Jeong J, Kim H. Synergistic effects of heat-killed kefir paraprobiotics and flavonoid-rich prebiotics on western diet-induced obesity. Nutrients 2020; 12: 2465, doi: 10.3390/nu12082465.
    » https://doi.org/10.3390/nu12082465
  • 83
    Sugawara T, Sawada D, Yanagihara S, Aoki Y, Takehara I, Sugahara H, et al. Daily intake of paraprobiotic Lactobacillus amylovorus CP1563 improves pre-obese conditions and affects the gut microbial community in healthy pre-obese subjects: a double-blind, randomized, placebo-controlled study. Microorganisms 2020; 8: 304, doi: 10.3390/microorganisms8020304.
    » https://doi.org/10.3390/microorganisms8020304
  • 84
    Kiencke P, Viehmann K, Rychlik R. Cost-effectiveness analysis, prevention of atopic dermatitis by oral application of bacterial lysate in newborns/small children. Eur J Health Econ 2013; 14: 995-1002, doi: 10.1007/s10198-012-0448-x.
    » https://doi.org/10.1007/s10198-012-0448-x
  • 85
    Heintz B, Schlenter WW, Kirsten R, Nelson K. Clinical efficacy of Broncho-Vaxom in adult patients with chronic purulent sinusitis--a multi-centric, placebo-controlled, double-blind study. Int J Clin Pharmacol Ther Toxicol 1989; 27: 530-534.
  • 86
    Lu Y, Li Y, Xu L, Xia M, Cao L. Bacterial lysate increases the percentage of natural killer T cells in peripheral blood and alleviates asthma in children. Pharmacology 2015; 95: 139-144, doi: 10.1159/000377683.
    » https://doi.org/10.1159/000377683
  • 87
    Zhang M, Luan H, Zhang Q, Wang L, Lv YM, He F, et al. Prevention of infection in immunosuppressive patients with autoimmune nephrosis by using an immunostimulating bacterial lysate Broncho-vaxom. Hum Vaccines Immunother 2012; 8: 1802-1807, doi: 10.4161/hv.21874.
    » https://doi.org/10.4161/hv.21874
  • 88
    Bertelli C, Pillonel T, Torregrossa A, Prod'hom G, Julie Fischer C, Greub G, et al. Bifidobacterium longum bacteremia in preterm infants receiving probiotics. Clin Infect Dis 2015; 60: 924-927, doi: 10.1093/cid/ciu946.
    » https://doi.org/10.1093/cid/ciu946

Publication Dates

  • Publication in this collection
    16 Jan 2023
  • Date of issue
    2022

History

  • Received
    10 Oct 2022
  • Accepted
    24 Nov 2022
Associação Brasileira de Divulgação Científica Av. Bandeirantes, 3900, 14049-900 Ribeirão Preto SP Brazil, Tel. / Fax: +55 16 3315-9120 - Ribeirão Preto - SP - Brazil
E-mail: bjournal@terra.com.br