Catalogue Search | MBRL
Search Results Heading
Explore the vast range of titles available.
MBRLSearchResults
-
DisciplineDiscipline
-
Is Peer ReviewedIs Peer Reviewed
-
Item TypeItem Type
-
SubjectSubject
-
YearFrom:-To:
-
More FiltersMore FiltersSourceLanguage
Done
Filters
Reset
4,123
result(s) for
"probiotics in cancer"
Sort by:
Probiotics and Cancer: Mechanistic Insights and Organ-Specific Impact
by
Ahmad, Md Faruque
,
Kambal, Nahla
,
Ahmad, Fakhruddin Ali
in
Animals
,
antioxidant
,
Antioxidants
2025
Probiotics have been revealed in various studies to modulate the gut microbiome and have a substantial impact on cancers, comprising oesophageal, lung, liver, and colorectal cancer. These properties are endorsed by a diverse mechanism, including the modulation of the gut microbiome; preventing the metabolism of carcinogenic substances; exertion of anti-inflammatory action, immunopotentiator properties, and antioxidant activities; prevention of tumour growth; and decreasing the adverse effects of chemotherapy. There are promising perspectives regarding the new and developing field of probiotic research in relation to cancer treatment. This review demonstrates the recent findings of probiotics efficacy in cancer prevention and treatment and organ-specific impact along with protection from chemotherapy-induced side effects. The present evidence specifies that strategic probiotics application may be an effective complementary approach for the management of numerous kinds of cancer; still, additional studies and clinical trials are required to comprehend the relationships between cancer and probiotics.
Journal Article
Faecalibacterium prausnitzii Ameliorates Colorectal Tumorigenesis and Suppresses Proliferation of HCT116 Colorectal Cancer Cells
by
Hsien-Tai Chiu
,
Ifeoma Julieth Dikeocha
,
Abdelkodose Mohammed Al-Kabsi
in
Azoxymethane
,
Bacteria
,
Biology (General)
2022
Faecalibacterium prausnitzii is one of the most abundant commensals of gut microbiota that is not commonly administered as a probiotic supplement. Being one of the gut’s major butyrate-producing bacteria, its clinical significance and uses are on the rise and it has been shown to have anti-inflammatory and gut microbiota-modulating properties in the treatment of inflammatory bowel illness, Crohn’s disease, and colorectal cancer. Colorectal cancer (CRC) is a silent killer disease that has become one of the leading causes of cancer-related death worldwide. This study aimed to evaluate the anti-tumorigenic and antiproliferative role of F. prausnitzii as well as to study its effects on the diversity of gut microbiota in rats. Findings showed that F. prausnitzii probiotic significantly reduced the colonic aberrant crypt foci frequency and formation in Azoxymethane (AOM)-induced CRC in rats. In addition, the administration of F. prausnitzii lowered the lipid peroxidation levels in the colon tissues. For in vitro 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT) assay, the cell-free supernatant of F. prausnitzii suppressed the growth of HCT116 colorectal cancer cells in a time/dose-dependent manner. 16S rRNA gene sequencing using rat stool samples showed that the administration of F. prausnitzii modulated the gut microbiota of the rats and enhanced its diversity. Hence, these findings suggest that F. prausnitzii as a probiotic supplement can be used in CRC prevention and management; however, more studies are warranted to understand its cellular and molecular mechanisms of action.
Journal Article
Investigation into the Potential Role of Propionibacterium freudenreichii in Prevention of Colorectal Cancer and Its Effects on the Diversity of Gut Microbiota in Rats
by
Michael Mathai
,
Ifeoma Julieth Dikeocha
,
Abdelkodose Mohammed Al-Kabsi
in
colorectal cancer; probiotics; Propionibacterium freudenreichii; azoxymethane; microbiome
2023
Journal Article
Toxicological Evaluation of a Probiotic-Based Delivery System for P8 Protein as an Anti-Colorectal Cancer Drug
by
Kim, Tai Yeub
,
Kwon, Daebeom
,
An, Byung Chull
in
Administration, Oral
,
Animal models
,
Animals
2021
This study aimed to toxicological evaluate a probiotics-based delivery system for p8 protein as an anti-colorectal cancer drug.
Lactic acid bacteria (LAB) have been widely ingested for many years and are regarded as very safe. Recently, a
SL4 (PP) strain that secretes the probiotic-derived anti-cancer protein P8 (PP-P8) has been developed as an anti-colorectal cancer (CRC) biologic by Cell Biotech. We initially identified a
(LR)-derived anti-cancer protein, P8, that suppresses CRC growth. We also showed that P8 penetrates specifically into CRC cells (DLD-1 cells) through endocytosis. We then confirmed the efficacy of PP-P8, showing that oral administration of this agent significantly decreased tumor mass (~42%) relative to controls in a mouse CRC xenograft model. In terms of molecular mechanism, PP-P8 induces cell-cycle arrest in G
phase through down-regulation of Cyclin B1 and Cdk1. In this study, we performed in vivo toxicology profiling to obtain evidence that PP-P8 is safe, with the goal of receiving approval for an investigational new drug application (IND).
Based on gene therapy guidelines of the Ministry of Food and Drug Safety (MFDS) of Korea, the potential undesirable effects of PP-P8 had to be investigated in intact small rodent or marmoset models prior to first-in-human (FIH) administration. The estimated doses of PP-P8 for FIH are 1.0×10
- 1.0×10
CFU/person (60 kg). Therefore, to perform toxicological investigations in non-clinical animal models, we orally administered PP-P8 at doses of 3.375 × 10
, 6.75 × 10
, and 13.5×10
CFU/kg/day; thus the maximum dose was 800-8000-fold higher than the estimated dose for FIH.
In our animal models, we observed no adverse effects of PP-P8 on clinicopathologic findings, relative organ weight, or tissue pathology. In addition, we observed no inflammation or ulceration during pathological necropsy.
These non-clinical toxicology studies could be used to furnish valuable data for the safety certification of PP-P8.
Journal Article
Effects of Lactobacillus plantarum A7 with probiotic potential on colon cancer and normal cells proliferation in comparison with a commercial strain
by
Fazeli, Hossain
,
Sadeghi-Aliabadi, Hojjat
,
Mirlohi, Maryam
in
Colon cancer L. plantarum A7 Lacticacid bacteria MTT assay Probiotics
,
Original
2014
Several beneficial effects have been attributed to the probiotic lactic acid bacteria. It was determined that lactobacilli can exert antiproliferative effects on the various cancer cell lines including colon cancer. Effects of lactic acid bacteria on colon cancer may vary from strain to strain and there is a need to find the new probiotic strains with tumor suppressing properties through in vitro studies.
Anti-proliferative activities of heat-killed cells and cell-free supernatants of a native strain of Lactobacillus plantarum A7 and a commercial strain of lactobacillus rhamnosus GG were assessed on human colon cancer cell lines (Caco-2 and HT-29) and normal cells (L-929), using MTT assay. Cells were seeded at 2×10(4) cells/mlin 96 well plates and incubated for 24 hr. Then heat-killed cells (OD620: 0.025, 0.0.05, 0.1) and cell-free supernatants of bacteria were added at concentration of 2.5, 5 and 10 mg/ml. After 48 hr incubation MTT (5 mg/ml) was added and the absorbance was measured at 540 nm using ELISA plate reader.
Results showed that heat-killed cells and cell-free supernatants of both probiotic strains reduced the growth rate of cancer and normal cells. These results suggested that anti-proliferative effect may not be an exclusive characteris ticwhich is dedicated to officially approved probiotics.
L. plantarum A7 could be considered as colon cancer biological product, most likely due to its advantages in significant organic acid production.
Journal Article
The pros, cons, and many unknowns of probiotics
2019
Consumption of over-the-counter probiotics for promotion of health and well-being has increased worldwide in recent years. However, although probiotic use has been greatly popularized among the general public, there are conflicting clinical results for many probiotic strains and formulations. Emerging insights from microbiome research enable an assessment of gut colonization by probiotics, strain-level activity, interactions with the indigenous microbiome, safety and impacts on the host, and allow the association of probiotics with physiological effects and potentially useful medical indications. In this Perspective, we highlight key advances, challenges and limitations in striving toward an unbiased interpretation of the large amount of data regarding over-the-counter probiotics, and propose avenues to improve the quality of evidence, transparency, public awareness and regulation of their use.The use and promotion of probiotics is widespread, but debatable in many cases. Prospective large-scale randomized studies that assess their effectiveness in promoting health and curing disease and take into account personalized responses of discrete human subpopulations will help clarify specific indications in which probiotics may be safe and beneficial.
Journal Article
Probiotic neoantigen delivery vectors for precision cancer immunotherapy
2024
Microbial systems have been synthetically engineered to deploy therapeutic payloads in vivo
1
,
2
. With emerging evidence that bacteria naturally home in on tumours
3
,
4
and modulate antitumour immunity
5
,
6
, one promising application is the development of bacterial vectors as precision cancer vaccines
2
,
7
. Here we engineered probiotic
Escherichia coli
Nissle 1917 as an antitumour vaccination platform optimized for enhanced production and cytosolic delivery of neoepitope-containing peptide arrays, with increased susceptibility to blood clearance and phagocytosis. These features enhance both safety and immunogenicity, achieving a system that drives potent and specific T cell-mediated anticancer immunity that effectively controls or eliminates tumour growth and extends survival in advanced murine primary and metastatic solid tumours. We demonstrate that the elicited antitumour immune response involves recruitment and activation of dendritic cells, extensive priming and activation of neoantigen-specific CD4
+
and CD8
+
T cells, broader activation of both T and natural killer cells, and a reduction of tumour-infiltrating immunosuppressive myeloid and regulatory T and B cell populations. Taken together, this work leverages the advantages of living medicines to deliver arrays of tumour-specific neoantigen-derived epitopes within the optimal context to induce specific, effective and durable systemic antitumour immunity.
Probiotic
Escherichia
coli
Nissle 1917 is engineered as an antitumour vaccination platform optimized for enhanced production and cytosolic delivery of neoepitope-containing peptide arrays to safely induce specific, effective and durable systemic antitumour immunity.
Journal Article
Postbiotics—A Step Beyond Pre- and Probiotics
by
Żółkiewicz, Jakub
,
Ruszczyński, Marek
,
Feleszko, Wojciech
in
Animals
,
Antioxidants
,
Antitubercular Agents
2020
As an imbalance in the intestinal microbiota can lead to the development of several diseases (e.g., type 1 diabetes, cancer, among others), the use of prebiotics, probiotics, and postbiotics to alter the gut microbiome has attracted recent interest. Postbiotics include any substance released by or produced through the metabolic activity of the microorganism, which exerts a beneficial effect on the host, directly or indirectly. As postbiotics do not contain live microorganisms, the risks associated with their intake are minimized. Here, we provided a critical review of postbiotics described in the literature, including their mechanisms of action, clinical characteristics, and potential therapeutic applications. We detailed the pleiotropic effects of postbiotics, including their immunomodulatory, anti-inflammatory, antioxidant, and anti-cancer properties. Although the use of postbiotics is an attractive strategy for altering the microbiome, further study into its efficacy and safety is warranted.
Journal Article
Probiotics ameliorate H. pylori-associated gastric β-catenin and COX-2 carcinogenesis signaling by regulating miR-185
by
Wu, Chung-Tai
,
Tseng, Joseph T.
,
Chen, Wei-Ying
in
Antibodies
,
Bacteria
,
Bacterial infections
2025
Background
This study aimed to investigate whether probiotics can ameliorate the
H. pylori
-induced Wnt/β-catenin-related COX-2 carcinogenesis signaling pathway by regulating the expression of microRNAs (miRNAs).
Methods
An
H. pylori
isolate and GES-1 cells were used to establish a COX-2-associated carcinogenesis axis. Western blot analysis was conducted to investigate Wnt/β-catenin and COX-2 signaling. Next-generation sequencing and DIANA Tools identified significant differences in miRNA expressions. The probiotics
Lactobacillus acidophilus
and
Bifidobacterium lactis
were used to study anti-carcinogenesis effects in GES-1 and miRNA-transfected GES-1 cells. The
H. pylori
-infected patients with intestinal metaplasia (IM) were randomly allocated into probiotic treatment or not after successful eradication, the IM regression was assessed by the 2nd esophagogastroduodenoscopy one year after treatment.
Results
Pretreatment with probiotics significantly reduced
H. pylori
-induced nuclear β-catenin phosphorylation and COX-2 levels in GES-1 cells. Among 9 significantly altered miRNAs, miR-185 was the only miRNA targeting the Wnt/β-catenin signaling pathway.
H. pylori
increased miR-185 expression and upregulated COX-2 carcinogenesis through the Wnt/β-catenin pathway, but not the JAK2/STAT3 pathway.
B. lactis
ameliorated
H. pylori
-induced miR-185 expression and nuclear β-catenin/COX-2 signaling in a dose-dependent manner. In the 6-month probiotic-treated patients had a significantly higher IM regression rate than controls (intention-to-treat: 37.5 vs 11.5%, OR: 4.60, 95% CI: 1.134–18.65,
p
= 0.025; per-protocol: 46.2 vs 17.6%, OR: 4.00, 95% CI: 0.923–17.33,
p
= 0.055). Patients without IM regression had significantly higher miR-185 levels in follow-up biopsies (
p
< 0.01).
Conclusions
Pretreatment with
B. lactis
ameliorated the
H. pylori
-induced COX-2 carcinogenesis pathway by reducing miR-185 expression, which targets Wnt/β-catenin signaling. (ClinicalTrials.gov, NCT05544396).
Journal Article
Beyond Analgesia: Psychobiotics as an Adjunctive Approach to Pain Management in Gastrointestinal Oncology—A Post Hoc Analysis from the ProDeCa Study
by
Menni, Alexandra-Eleftheria
,
Stavrou, George
,
Kotzampassi, Katerina
in
Adult
,
Aged
,
Analgesia
2025
Background: Pain is a multifaceted and debilitating symptom in patients with gastrointestinal cancer, especially those undergoing surgical resection followed by chemotherapy. The interplay between inflammatory, neuropathic, and psychosocial components often renders conventional analgesia insufficient. Psychobiotics—probiotic strains with neuroactive properties—have recently emerged as potential modulators of pain perception through neuroimmune and gut–brain axis pathways. Methods: This post hoc analysis is based on the ProDeCa randomized, placebo-controlled trial, which originally aimed to assess the psychotropic effects of a four-strain psychobiotic formulation in postoperative gastrointestinal cancer patients receiving chemotherapy. In the current analysis, we evaluated changes in pain perception among non-depressed and depressed participants, who received either psychobiotics or placebo, along with standard analgesic regimes. Pain was assessed at baseline, after a month of treatment, and at follow-up, 2 months thereafter, using the Short-Form McGill Pain Questionnaire (SF-MPQ), capturing both sensory and affective components, as well as with the Present Pain Intensity and the VAS scores. Results: Psychobiotic-treated participants—particularly the non-depressed ones—exhibited a significant reduction in both quantitative and qualitative pain indices over time compared with placebo-treated ones. Improvements were noted in total pain rating index scores, sensory and affective subscales, and present pain intensity. These effects were sustained up to 2 months after intervention. In contrast, placebo groups demonstrated worsening in pain scores, probably influenced by ongoing chemotherapy and disease progression. The analgesic effect was less pronounced but still observable in the subgroup with symptoms of depression. Conclusions: Adjunctive psychobiotic therapy appears to beneficially modulate pain perception in gastrointestinal oncology patients receiving chemotherapy, with the most pronounced effects being in non-depressed individuals. These findings suggest psychobiotics as a promising non-opioid add-on for comprehensive cancer pain management and support further investigation in larger pain-targeted trials.
Journal Article