1
|
Stringer AM, Hargreaves BM, Mendes RA, Blijlevens NMA, Bruno JS, Joyce P, Kamath S, Laheij AMGA, Ottaviani G, Secombe KR, Tonkaboni A, Zadik Y, Bossi P, Wardill HR. Updated perspectives on the contribution of the microbiome to the pathogenesis of mucositis using the MASCC/ISOO framework. Support Care Cancer 2024; 32:558. [PMID: 39080025 PMCID: PMC11289053 DOI: 10.1007/s00520-024-08752-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 07/19/2024] [Indexed: 08/02/2024]
Abstract
Advances in the treatment of cancer have significantly improved mortality rates; however, this has come at a cost, with many treatments still limited by their toxic side effects. Mucositis in both the mouth and gastrointestinal tract is common following many anti-cancer agents, manifesting as ulcerative lesions and associated symptoms throughout the alimentary tract. The pathogenesis of mucositis was first defined in 2004 by Sonis, and almost 20 years on, the model continues to be updated reflecting ongoing research initiatives and more sophisticated analytical techniques. The most recent update, published by the Multinational Association for Supportive Care in Cancer and the International Society for Oral Oncology (MASCC/ISOO), highlights the numerous co-occurring events that underpin mucositis development. Most notably, a role for the ecosystem of microorganisms that reside throughout the alimentary tract (the oral and gut microbiota) was explored, building on initial concepts proposed by Sonis. However, many questions remain regarding the true causal contribution of the microbiota and associated metabolome. This review aims to provide an overview of this rapidly evolving area, synthesizing current evidence on the microbiota's contribution to mucositis development and progression, highlighting (i) components of the 5-phase model where the microbiome may be involved, (ii) methodological challenges that have hindered advances in this area, and (iii) opportunities for intervention.
Collapse
Affiliation(s)
- Andrea M Stringer
- Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Benjamin M Hargreaves
- Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Rui Amaral Mendes
- Faculty of Medicine, University of Porto/CINTESIS@RISE, Porto, Portugal
- Department of Oral and Maxillofacial Medicine and Diagnostic Sciences, Case Western Reserve University, Cleveland, OH, 44106-7401, USA
| | - Nicole M A Blijlevens
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Julia S Bruno
- Molecular Oncology Center, Hospital Sírio-Libanês, São Paulo, Brazil
| | - Paul Joyce
- Centre for Pharmaceutical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Srinivas Kamath
- Centre for Pharmaceutical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Alexa M G A Laheij
- Department of Oral Medicine, Academic Centre for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, The Netherlands
- Department of Oral and Maxillofacial Surgery, UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Giulia Ottaviani
- Department of Surgical, Medical and Health Sciences, University of Trieste, Trieste, Italy
| | - Kate R Secombe
- The School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
| | - Arghavan Tonkaboni
- Department of Oral Medicine, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Yehuda Zadik
- Department of Military Medicine and "Tzameret", Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Oral Medicine, Sedation and Imaging, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Paolo Bossi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Hannah R Wardill
- The School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia.
- Supportive Oncology Research Group, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Level 5S, Adelaide, 5000, Australia.
| |
Collapse
|
2
|
Wardill HR, de Mooij CEM, Da Silva Ferreira AR, Havinga H, Harmsen HJM, van der Velden WJFM, van Groningen LFJ, Tissing WJE, Blijlevens NMA. Supporting the gastrointestinal microenvironment during high-dose chemotherapy and stem cell transplantation by inhibiting IL-1 signaling with anakinra. Sci Rep 2022; 12:6803. [PMID: 35546555 PMCID: PMC9095632 DOI: 10.1038/s41598-022-10700-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 04/07/2022] [Indexed: 11/20/2022] Open
Abstract
High-dose chemotherapy causes intestinal inflammation and subsequent breakdown of the mucosal barrier, permitting translocation of enteric pathogens, clinically manifesting as fever. Antibiotics are mainstay for controlling these complications, however, they are increasingly recognized for their detrimental effects, including antimicrobial resistance and dysbiosis. Here, we show that mucosal barrier injury induced by the mucotoxic chemotherapeutic agent, high-dose melphalan (HDM), is characterized by hyper-active IL-1b/CXCL1/neutrophil signaling. Inhibition of this pathway with IL-1RA, anakinra, minimized the duration and intensity of mucosal barrier injury and accompanying clinical symptoms, including diarrhea, weight loss and fever in rats. 16S analysis of fecal microbiome demonstrated a more stable composition in rats receiving anakinra, with reduced pathogen expansion. In parallel, we report through Phase IIA investigation that anakinra is safe in stem cell transplant patients with multiple myeloma after HDM. Ramping-up anakinra (100–300 mg administered intravenously for 15 days) did not cause any adverse events or dose limiting toxicities, nor did it change time to neutrophil recovery. Our results reinforce that strengthening the mucosal barrier may be an effective supportive care strategy to mitigate local and systemic clinical consequences of HDM. We are now conducting a Phase IIB multicenter, placebo-controlled, double-blinded trial to assess clinical efficacy of anakinra (AFFECT-2). Trial registration: ClinicalTrials.gov identifier: NCT03233776.
Collapse
Affiliation(s)
- H R Wardill
- School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia. .,The Supportive Oncology Research Group, Precision Medicine Theme (Cancer), The South Australian Health and Medical Research Institute, Adelaide, SA, Australia. .,Department of Pediatrics, The University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - C E M de Mooij
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - A R Da Silva Ferreira
- Department of Medical Microbiology, The University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - H Havinga
- Department of Pediatrics, The University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - H J M Harmsen
- Department of Medical Microbiology, The University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - L F J van Groningen
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - W J E Tissing
- Department of Pediatrics, The University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Princes Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - N M A Blijlevens
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
3
|
Kaliannan K, Donnell SO, Murphy K, Stanton C, Kang C, Wang B, Li XY, Bhan AK, Kang JX. Decreased Tissue Omega-6/Omega-3 Fatty Acid Ratio Prevents Chemotherapy-Induced Gastrointestinal Toxicity Associated with Alterations of Gut Microbiome. Int J Mol Sci 2022; 23:5332. [PMID: 35628140 PMCID: PMC9140600 DOI: 10.3390/ijms23105332] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/02/2022] [Accepted: 05/06/2022] [Indexed: 02/04/2023] Open
Abstract
Gastrointestinal toxicity (GIT) is a debilitating side effect of Irinotecan (CPT-11) and limits its clinical utility. Gut dysbiosis has been shown to mediate this side effect of CPT-11 by increasing gut bacterial β-glucuronidase (GUSB) activity and impairing the intestinal mucosal barrier (IMB). We have recently shown the opposing effects of omega-6 (n-6) and omega-3 (n-3) polyunsaturated fatty acids (PUFA) on the gut microbiome. We hypothesized that elevated levels of tissue n-3 PUFA with a decreased n-6/n-3 PUFA ratio would reduce CPT-11-induced GIT and associated changes in the gut microbiome. Using a unique transgenic mouse (FAT-1) model combined with dietary supplementation experiments, we demonstrate that an elevated tissue n-3 PUFA status with a decreased n-6/n-3 PUFA ratio significantly reduces CPT-11-induced weight loss, bloody diarrhea, gut pathological changes, and mortality. Gut microbiome analysis by 16S rRNA gene sequencing and QIIME2 revealed that improvements in GIT were associated with the reduction in the CPT-11-induced increase in both GUSB-producing bacteria (e.g., Enterobacteriaceae) and GUSB enzyme activity, decrease in IMB-maintaining bacteria (e.g., Bifidobacterium), IMB dysfunction and systemic endotoxemia. These results uncover a host-microbiome interaction approach to the management of drug-induced gut toxicity. The prevention of CPT-11-induced gut microbiome changes by decreasing the tissue n-6/n-3 PUFA ratio could be a novel strategy to prevent chemotherapy-induced GIT.
Collapse
Affiliation(s)
- Kanakaraju Kaliannan
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; (K.K.); (B.W.); (X.-Y.L.)
| | - Shane O. Donnell
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland; (S.O.D.); (C.S.)
- Teagasc Moorepark Food Research Centre, Fermoy, P61 C996 Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland;
| | - Kiera Murphy
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland;
| | - Catherine Stanton
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland; (S.O.D.); (C.S.)
- Teagasc Moorepark Food Research Centre, Fermoy, P61 C996 Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland;
| | - Chao Kang
- Department of Nutrition, The General Hospital of Western Theater Command, Chengdu 610000, China;
| | - Bin Wang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; (K.K.); (B.W.); (X.-Y.L.)
| | - Xiang-Yong Li
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; (K.K.); (B.W.); (X.-Y.L.)
| | - Atul K. Bhan
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA;
| | - Jing X. Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; (K.K.); (B.W.); (X.-Y.L.)
| |
Collapse
|
4
|
A Perspective on the Role of Microbiome for Colorectal Cancer Treatment. Cancers (Basel) 2021; 13:cancers13184623. [PMID: 34572850 PMCID: PMC8468110 DOI: 10.3390/cancers13184623] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/13/2021] [Accepted: 09/13/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Colorectal cancer is the third most diagnosed cancer worldwide and contributes significantly to global mortality and morbidity. The gut microbiome, composed of the trillions of microbes endemic to the human gastrointestinal tract, has been shown to be implicated in colorectal cancer oncogenesis; however, the roles of microbiota and dysbiosis in CRC treatment remain poorly understood. This review sought to characterize this relationship and in doing so, identify how these interactions may inform future treatments in the form of synbiotics designed to alter the host microbiota to achieve optimized treatment outcomes. Abstract In healthy hosts, trillions of microbes colonise the gut and oral cavity in a well-balanced state, maintaining a mutually beneficial relationship. Loss of this balance, termed dysbiosis, is strongly implicated in the pathogenesis of colorectal cancer (CRC). However, the roles of microbiota and dysbiosis in CRC treatment remain poorly understood. Recent studies suggest that the gut microbiota has the ability to affect the host response to chemotherapeutic agents by enhancing drug efficacy, promoting chemoresistance and mediating chemotherapy-induced toxicity and side effects via a variety of mechanisms. Several other studies have also proposed manipulation of the microbiota to optimise CRC treatment. In this review, we summarise the current advancement of knowledge on how microbiota and CRC treatments interact with each other and how this interaction may shed some light on the development of personalised microbiota manipulations that improve CRC treatment outcomes.
Collapse
|
5
|
Wei L, Wen XS, Xian CJ. Chemotherapy-Induced Intestinal Microbiota Dysbiosis Impairs Mucosal Homeostasis by Modulating Toll-like Receptor Signaling Pathways. Int J Mol Sci 2021; 22:ijms22179474. [PMID: 34502383 PMCID: PMC8431669 DOI: 10.3390/ijms22179474] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 08/26/2021] [Accepted: 08/28/2021] [Indexed: 02/06/2023] Open
Abstract
Chemotherapy-induced intestinal mucositis, a painful debilitating condition affecting up to 40–100% of patients undergoing chemotherapy, can reduce the patients’ quality of life, add health care costs and even postpone cancer treatment. In recent years, the relationships between intestinal microbiota dysbiosis and mucositis have drawn much attention in mucositis research. Chemotherapy can shape intestinal microbiota, which, in turn, can aggravate the mucositis through toll-like receptor (TLR) signaling pathways, leading to an increased expression of inflammatory mediators and elevated epithelial cell apoptosis but decreased epithelial cell differentiation and mucosal regeneration. This review summarizes relevant studies related to the relationships of mucositis with chemotherapy regimens, microbiota, TLRs, inflammatory mediators, and intestinal homeostasis, aiming to explore how gut microbiota affects the pathogenesis of mucositis and provides potential new strategies for mucositis alleviation and treatment and development of new therapies.
Collapse
Affiliation(s)
- Ling Wei
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China;
| | - Xue-Sen Wen
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China;
- Correspondence: (X.-S.W.); (C.J.X.); Tel.: +86-531-88382028 (X.-S.W.); +61-88302-1944 (C.J.X.)
| | - Cory J. Xian
- UniSA Clinical & Health Science, City West Campus, University of South Australia, Adelaide, SA 5001, Australia
- Correspondence: (X.-S.W.); (C.J.X.); Tel.: +86-531-88382028 (X.-S.W.); +61-88302-1944 (C.J.X.)
| |
Collapse
|
6
|
Tsunoda SM, Gonzales C, Jarmusch AK, Momper JD, Ma JD. Contribution of the Gut Microbiome to Drug Disposition, Pharmacokinetic and Pharmacodynamic Variability. Clin Pharmacokinet 2021; 60:971-984. [PMID: 33959897 PMCID: PMC8332605 DOI: 10.1007/s40262-021-01032-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2021] [Indexed: 12/20/2022]
Abstract
The trillions of microbes that make up the gut microbiome are an important contributor to health and disease. With respect to xenobiotics, particularly orally administered compounds, the gut microbiome interacts directly with drugs to break them down into metabolic products. In addition, microbial products such as bile acids interact with nuclear receptors on host drug-metabolizing enzyme machinery, thus indirectly influencing drug disposition and pharmacokinetics. Gut microbes also influence drugs that undergo enterohepatic recycling by reversing host enzyme metabolic processes and increasing exposure to toxic metabolites as exemplified by the chemotherapy agent irinotecan and non-steroidal anti-inflammatory drugs. Recent data with immune checkpoint inhibitors demonstrate the impact of the gut microbiome on drug pharmacodynamics. We summarize the clinical importance of gut microbe interaction with digoxin, irinotecan, immune checkpoint inhibitors, levodopa, and non-steroidal anti-inflammatory drugs. Understanding the complex interactions of the gut microbiome with xenobiotics is challenging; and highly sensitive methods such as untargeted metabolomics with molecular networking along with other in silico methods and animal and human in vivo studies will uncover mechanisms and pathways. Incorporating the contribution of the gut microbiome to drug disposition, pharmacokinetics, and pharmacodynamics is vital in this era of precision medicine.
Collapse
Affiliation(s)
- Shirley M Tsunoda
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, MC 0657, La Jolla, San Diego, CA, 90293-0657, USA.
| | - Christopher Gonzales
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, MC 0657, La Jolla, San Diego, CA, 90293-0657, USA
| | - Alan K Jarmusch
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, MC 0657, La Jolla, San Diego, CA, 90293-0657, USA.,Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Jeremiah D Momper
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, MC 0657, La Jolla, San Diego, CA, 90293-0657, USA
| | - Joseph D Ma
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, MC 0657, La Jolla, San Diego, CA, 90293-0657, USA
| |
Collapse
|
7
|
Gastrointestinal cancers: the role of microbiota in carcinogenesis and the role of probiotics and microbiota in anti-cancer therapy efficacy. Cent Eur J Immunol 2021; 45:476-487. [PMID: 33658894 PMCID: PMC7882408 DOI: 10.5114/ceji.2020.103353] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 09/24/2019] [Indexed: 02/06/2023] Open
Abstract
The gut epithelium is a habitat of a variety of microorganisms, including bacteria, fungi, viruses and Archaea. With the advent of sophisticated molecular techniques and bioinformatics tools, more information on the composition and thus function of gut microbiota was revealed. The gut microbiota as an integral part of the intestinal barrier has been shown to be involved in shaping the mucosal innate and adaptive immune response and to provide protection against pathogens. Consequently, a set of biochemical signals exchanged within microbes and communication between the microbiota and the host have opened a new way of thinking about cancer biology. Probiotics are living organisms which administered in adequate amounts may bring health benefits and have the potential to be an integral part of the prevention/treatment strategies in clinical approaches. Here we provide a comprehensive review of data linking gut microbiota to cancer pathogenesis and its clinical course. We focus on gastrointestinal cancers, such as gastric, colorectal, pancreatic and liver cancer.
Collapse
|
8
|
Drug Disposition in the Lower Gastrointestinal Tract: Targeting and Monitoring. Pharmaceutics 2021; 13:pharmaceutics13020161. [PMID: 33530468 PMCID: PMC7912393 DOI: 10.3390/pharmaceutics13020161] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/20/2022] Open
Abstract
The increasing prevalence of colonic diseases calls for a better understanding of the various colonic drug absorption barriers of colon-targeted formulations, and for reliable in vitro tools that accurately predict local drug disposition. In vivo relevant incubation conditions have been shown to better capture the composition of the limited colonic fluid and have resulted in relevant degradation and dissolution kinetics of drugs and formulations. Furthermore, drug hurdles such as efflux transporters and metabolising enzymes, and the presence of mucus and microbiome are slowly integrated into drug stability- and permeation assays. Traditionally, the well characterized Caco-2 cell line and the Ussing chamber technique are used to assess the absorption characteristics of small drug molecules. Recently, various stem cell-derived intestinal systems have emerged, closely mimicking epithelial physiology. Models that can assess microbiome-mediated drug metabolism or enable coculturing of gut microbiome with epithelial cells are also increasingly explored. Here we provide a comprehensive overview of the colonic physiology in relation to drug absorption, and review colon-targeting formulation strategies and in vitro tools to characterize colonic drug disposition.
Collapse
|
9
|
Horniblow RD, Mistry P, Quraishi MN, Beggs AD, Van de Wiele T, Iqbal TH, Tselepis C. The Safety and Tolerability of a Potential Alginate-Based Iron Chelator; Results of A Healthy Participant Study. Nutrients 2019; 11:E674. [PMID: 30901846 PMCID: PMC6471009 DOI: 10.3390/nu11030674] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/16/2022] Open
Abstract
Evidence supporting the ferro-toxic nature of iron in the progression of inflammatory bowel disease (IBD) is becoming well established. A microbial dysbiosis is observed in IBD patients, and intra-luminal colonic-iron is able to support a more pathogenic community of bacteria; whether this is attributed to the development of IBD and how iron could be mediating these microbial changes is still unknown. Dietary fibres are commonly used in pre-biotic supplements to beneficially affect the host by improving the viability of bacterial communities within the colon. Alginates are a class of biopolymers considered as prebiotics due to their fibre-like composition and are able to bind metal cations, in particular, iron. Considering that iron excess is able to negatively alter the microbiome, the use of alginate as a food supplement could be useful in colonic-iron chelation. As such, this first-in-man study aimed to assess whether the use of alginate as a dietary iron chelator was both safe and well tolerated. In addition, the impact of alginate on the microbiome and iron levels was assessed by using an intestinal model SHIME (Simulation of the Human Intestinal Microbial Ecosystem). Alginate was supplemented into the diets (3 g/day) of healthy volunteers (n = 17) for 28 days. Results from this study suggest that daily ingestion of 3 g alginate was well tolerated with very minor side effects. There were no detrimental changes in a variety of haematological parameters or the intestinal microbiome. The bacterial communities within the SHIME model were also not influenced by iron and or alginate; it is possible that alginate may be susceptible to bacterial or enzymatic degradation within the gastro-intestinal tract.
Collapse
Affiliation(s)
- Richard D Horniblow
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Pritesh Mistry
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
- Queen Elizabeth Hospital Birmingham, Edgbaston, Birmingham B15 2TH, UK.
| | - Mohammed N Quraishi
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
- The University of Birmingham Microbiome Treatment Centre, University of Birmingham, Birmingham B15 2TT, UK.
| | - Andrew D Beggs
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
- Queen Elizabeth Hospital Birmingham, Edgbaston, Birmingham B15 2TH, UK.
| | - Tom Van de Wiele
- CMET, Center for Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Gent, Belgium.
| | - Tariq H Iqbal
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
- Queen Elizabeth Hospital Birmingham, Edgbaston, Birmingham B15 2TH, UK.
- The University of Birmingham Microbiome Treatment Centre, University of Birmingham, Birmingham B15 2TT, UK.
| | - Chris Tselepis
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| |
Collapse
|