Does Chemotherapy-Induced Gastrointestinal Mucositis Affect the Bioavailability and Efficacy of Anti-Infective Drugs?
Abstract
:1. Introduction
2. Physiological Factors Contributing to Impaired Intestinal Absorption during Chemotherapy
2.1. Gastrointestinal Mucositis and Barrier Function
2.2. Intestinal Motility, Luminal pH and Diet
2.3. The Gut Microbiota
3. The Effects of Gastrointestinal Mucositis on Drug Absorption
4. Where Do We Go from Here?
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Sonis, S.T. The pathobiology of mucositis. Nat. Rev. Cancer. 2004, 4, 277–284. [Google Scholar] [CrossRef]
- Wardill, H.R.; Van Sebille, Y.Z.A.; Ciorba, M.A.; Bowen, J.M. Prophylactic probiotics for cancer therapy-induced diarrhoea: A meta-analysis. Curr. Opin. Support. Palliat. Care 2018, 12, 187–197. [Google Scholar] [CrossRef]
- Welch, H.G.; Schwartz, L.M.; Woloshin, S. Are increasing 5-year survival rates evidence of success against cancer? J. Am. Med. Assoc. 2000, 283, 2975–2978. [Google Scholar] [CrossRef] [PubMed]
- Arnold, M.; Rutherford, M.J.; Bardot, A.; Ferlay, J.; Andersson, T.M.L.; Myklebust, T.Å.; Tervonen, H.; Thursfield, V.; Ransom, D.; Shack, L.; et al. Progress in cancer survival, mortality, and incidence in seven high-income countries 1995–2014 (ICBP SURVMARK-2): A population-based study. Lancet Oncol. 2019, 20, 1493–1505. [Google Scholar] [CrossRef] [Green Version]
- Piñana, J.L.; Montesinos, P.; Martino, R.; Vazquez, L.; Rovira, M.; López, J.; Batlle, M.; Figuera, Á.; Barba, P.; Lahuerta, J.J.; et al. Incidence, risk factors, and outcome of bacteremia following autologous hematopoietic stem cell transplantation in 720 adult patients. Ann. Hematol. 2014, 93, 299–307. [Google Scholar] [CrossRef] [PubMed]
- Taur, Y.; Xavier, J.B.; Lipuma, L.; Ubeda, C.; Goldberg, J.; Gobourne, A.; Lee, Y.J.; Dubin, K.A.; Socci, N.D.; Viale, A.; et al. Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation. Clin. Infect. Dis. 2012, 55, 905–914. [Google Scholar] [CrossRef] [PubMed]
- Sonis, S.T. Pathobiology of mucositis. Semin. Oncol. Nurs. 2004, 20, 11–15. [Google Scholar] [CrossRef] [PubMed]
- Cario, E. Toll-like receptors in the pathogenesis of chemotherapy-induced gastrointestinal toxicity. Curr. Opin. Support. Palliat. Care 2016, 10, 157–164. [Google Scholar] [CrossRef]
- Gafter-Gvili, A.; Fraser, A.; Paul, M.; Leibovici, L. Meta-analysis: Antibiotic prophylaxis reduces mortality in neutropenic patients. Ann. Intern. Med. 2005, 142, 979–995. [Google Scholar] [CrossRef] [PubMed]
- Dahlgren, D.; Lennernäs, H. Intestinal permeability and drug absorption: Predictive experimental, computational and in vivo approaches. Pharmaceutics 2019, 11, 411. [Google Scholar] [CrossRef] [Green Version]
- Basile, D.; Di Nardo, P.; Corvaja, C.; Garattini, S.K.; Pelizzari, G.; Lisanti, C.; Bortot, L.; Da Ros, L.; Bartoletti, M.; Borghi, M.; et al. Mucosal injury during anti-cancer treatment: From pathobiology to bedside. Cancers 2019, 11, 857. [Google Scholar] [CrossRef] [Green Version]
- Elting, L.S.; Cooksley, C.; Chambers, M.; Cantor, S.B.; Manzullo, E.; Rubenstein, E.B. The burdens of cancer therapy: Clinical and economic outcomes of chemotherapy-induced mucositis. Cancer 2003, 98, 1531–1539. [Google Scholar] [CrossRef]
- Sougiannis, A.T.; VanderVeen, B.N.; Davis, J.M.; Fan, D.; Murphy, E.A. Understanding chemotherapy-induced intestinal mucositis and strategies to improve gut resilience. Am. J. Physiol. Liver Physiol. 2021, 320, G712–G719. [Google Scholar] [CrossRef]
- Naidu, M.U.R.; Ramana, G.V.; Rani, P.U.; Mohan, I.K.; Suman, A.; Roy, P. Chemotherapy-induced and/or radiation therapy-induced oral mucositis--complicating the treatment of cancer. Neoplasia 2004, 6, 423–431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wadleigh, R.G.; Redman, R.S.; Graham, M.L.; Krasnow, S.H.; Anderson, A.; Cohen, M.H. Vitamin E in the treatment of chemotherapy-induced mucositis. Am. J. Med. 1992, 92, 481–484. [Google Scholar] [CrossRef]
- Touchefeu, Y.; Montassier, E.; Nieman, K.; Gastinne, T.; Potel, G.; Bruley Des Varannes, S.; Le Vacon, F.; De La Cochetière, M.F. Systematic review: The role of the gut microbiota in chemotherapy- or radiation-induced gastrointestinal mucositis—Current evidence and potential clinical applications. Aliment. Pharmacol. Ther. 2014, 40, 409–421. [Google Scholar] [CrossRef] [PubMed]
- Sonis, S.; Elting, L.; Keefe, D.; Peterson, D.; Schubert, M.; Hauer-Jensen, M.; Bekele, B.; Raber-Durlacher, J.; Donnelly, J.; Rubenstein, E. Perspectives on cancer therapy-induced mucosal injury. Cancer 2004, 100, 1995–2025. [Google Scholar] [CrossRef] [PubMed]
- Carlotto, A.; Hogsett, V.L.; Maiorini, E.M.; Razulis, J.G.; Sonis, S.T. The economic burden of toxicities associated with cancer treatment: Review of the literature and analysis of nausea and vomiting, diarrhoea, oral mucositis and fatigue. Pharmacoeconomics 2013, 31, 753–766. [Google Scholar] [CrossRef] [PubMed]
- Bowen, J.; Al-Dasooqi, N.; Bossi, P.; Wardill, H.; Van Sebille, Y.; Al-Azri, A.; Bateman, E.; Correa, M.E.; Raber-Durlacher, J.; Kandwal, A.; et al. The pathogenesis of mucositis: Updated perspectives and emerging targets. Support. Care Cancer 2019, 27, 4023–4033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cinausero, M.; Aprile, G.; Ermacora, P.; Basile, D.; Vitale, M.G.; Fanotto, V.; Parisi, G.; Calvetti, L.; Sonis, S.T. New frontiers in the pathobiology and treatment of cancer regimen-related mucosal injury. Front. Pharmacol. 2017, 8, 354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Galaup, A.; Paci, A. Pharmacology of dimethanesulfonate alkylating agents: Busulfan and treosulfan. Expert Opin. Drug Metab. Toxicol. 2013, 9, 333–347. [Google Scholar] [CrossRef] [PubMed]
- Voelcker, G. The Mechanism of Action of Cyclophosphamide and Its Consequences for the Development of a New Generation of Oxazaphosphorine Cytostatics. Sci. Pharm. 2020, 88, 42. [Google Scholar] [CrossRef]
- Dasari, S.; Tchounwou, P.B. Cisplatin in cancer therapy: Molecular mechanisms of action. Eur. J. Pharmacol. 2014, 740, 364–378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ehrsson, H.; Eksborg, S.; Österborg, A.; Mellstedt, H.; Lindfors, A. Oral melphalan pharmacokinetics—Relation to dose in patients with multiple myeloma. Med. Oncol. Tumor Pharmacother. 1989, 6, 151–154. [Google Scholar] [CrossRef] [PubMed]
- Ecknauer, R.; Löhrs, U. The effect of a single dose of cyclophosphamide on the jejunum of specified pathogenfree and germfree rats. Digestion 1976, 14, 269–280. [Google Scholar] [CrossRef] [PubMed]
- Wardill, H.R.; de Mooij, C.E.M.; da Silva Ferreira, A.R.; van de Peppel, I.P.; Havinga, R.; Harmsen, H.J.M.; Tissing, W.J.E.; Blijlevens, N.M.A. Translational model of melphalan-induced gut toxicity reveals drug-host-microbe interactions that drive tissue injury and fever. Cancer Chemother. Pharmacol. 2021, 88, 173–188. [Google Scholar] [CrossRef] [PubMed]
- Longley, D.B.; Harkin, D.P.; Johnston, P.G. 5-fluorouracil: Mechanisms of action and clinical strategies. Nat. Rev. Cancer 2003, 3, 330–338. [Google Scholar] [CrossRef] [PubMed]
- Chang, C.T.; Ho, T.Y.; Lin, H.; Liang, J.A.; Huang, H.C.; Li, C.C.; Lo, H.Y.; Wu, S.L.; Huang, Y.F.; Hsiang, C.Y. 5-fluorouracil induced intestinal mucositis via nuclear factor-??B activation by transcriptomic analysis and in vivo bioluminescence imaging. PLoS ONE 2012, 7, e31808. [Google Scholar] [CrossRef] [Green Version]
- Cronstein, B.N.; Aune, T.M. Methotrexate and its mechanisms of action in inflammatory arthritis. Nat. Rev. Rheumatol. 2020, 16, 145–154. [Google Scholar] [CrossRef] [PubMed]
- De Sousa Cavalcante, L.; Monteiro, G. Gemcitabine: Metabolism and molecular mechanisms of action, sensitivity and chemoresistance in pancreatic cancer. Eur. J. Pharmacol. 2014, 741, 8–16. [Google Scholar] [CrossRef]
- Logan, R.M.; Stringer, A.M.; Bowen, J.M.; Gibson, R.J.; Sonis, S.T.; Keefe, D.M.K. Is the pathobiology of chemotherapy-induced alimentary tract mucositis influenced by the type of mucotoxic drug administered? Cancer Chemother. Pharmacol. 2009, 63, 239–251. [Google Scholar] [CrossRef]
- Chang, C.-W.; Lee, H.-C.; Li, L.-H.; Chiang Chiau, J.-S.; Wang, T.-E.; Chuang, W.-H.; Chen, M.-J.; Wang, H.-Y.; Shih, S.-C.; Liu, C.-Y.; et al. Fecal Microbiota Transplantation Prevents Intestinal Injury, Upregulation of Toll-Like Receptors, and 5-Fluorouracil/Oxaliplatin-Induced Toxicity in Colorectal Cancer. Int. J. Mol. Sci. 2020, 21, 386. [Google Scholar] [CrossRef] [Green Version]
- Vanhoecke, B.; Bateman, E.; Mayo, B.; Vanlancker, E.; Stringer, A.; Thorpe, D.; Keefe, D. Dark Agouti rat model of chemotherapy-induced mucositis: Establishment and current state of the art. Exp. Biol. Med. 2015, 240, 725–741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Daniele, B.; Secondulfo, M.; De Vivo, R.; Pignata, S.; De Magistris, L.; Delrio, P.; Palaia, R.; Barletta, E.; Tambaro, R.; Carratù, R. Effect of Chemotherapy with 5-Fluorouracil on Intestinal Permeability and Absorption in Patients with Advanced Colorectal Cancer. J. Clin. Gastroenterol. 2001, 32, 228–230. [Google Scholar] [CrossRef] [PubMed]
- Fijlstra, M.; Ferdous, M.; Koning, A.M.; Rings, E.H.H.M.; Harmsen, H.J.M.; Tissing, W.J.E. Substantial decreases in the number and diversity of microbiota during chemotherapy-induced gastrointestinal mucositis in a rat model. Support. Care Cancer 2015, 23, 1513–1522. [Google Scholar] [CrossRef] [PubMed]
- Hamada, K.; Shitara, Y.; Sekine, S.; Horie, T. Zonula Occludens-1 alterations and enhanced intestinal permeability in methotrexate-treated rats. Cancer Chemother. Pharmacol. 2010, 66, 1031–1038. [Google Scholar] [CrossRef] [PubMed]
- Fujita, K.; Kubota, Y.; Ishida, H.; Sasaki, Y. Irinotecan, a key chemotherapeutic drug for metastatic colorectal cancer. World J. Gastroenterol. 2015, 21, 12234–12248. [Google Scholar] [CrossRef] [PubMed]
- Wardill, H.R.; Bowen, J.M.; Van Sebille, Y.Z.A.; Secombe, K.R.; Coller, J.K.; Ball, I.A.; Logan, R.M.; Gibson, R.J. TLR4-dependent claudin-1 internalization and secretagogue-mediated chloride secretion regulate irinotecan-induced diarrhea. Mol. Cancer Ther. 2016, 15, 2767–2779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stringer, A.M.; Gibson, R.J.; Bowen, J.M.; Logan, R.M.; Ashton, K.; Yeoh, A.S.J.; Al-Dasooqi, N.; Keefe, D.M.K. Irinotecan-induced mucositis manifesting as diarrhoea corresponds with an amended intestinal flora and mucin profile. Int. J. Exp. Pathol. 2009, 90, 489–499. [Google Scholar] [CrossRef]
- Russo, F.; Linsalata, M.; Clemente, C.; D’Attoma, B.; Orlando, A.; Campanella, G.; Giotta, F.; Riezzo, G. The effects of fluorouracil, epirubicin, and cyclophosphamide (FEC60) on the intestinal barrier function and gut peptides in breast cancer patients: An observational study. BMC Cancer 2013, 13, 56. [Google Scholar] [CrossRef] [Green Version]
- Secombe, K.R.; Coller, J.K.; Gibson, R.J.; Wardill, H.R.; Bowen, J.M. The bidirectional interaction of the gut microbiome and the innate immune system: Implications for chemotherapy-induced gastrointestinal toxicity. Int. J. Cancer 2019, 144, 2365–2376. [Google Scholar] [CrossRef] [PubMed]
- Al-Dasooqi, N.; Sonis, S.T.; Bowen, J.M.; Bateman, E.; Blijlevens, N.; Gibson, R.J.; Logan, R.M.; Nair, R.G.; Stringer, A.M.; Yazbeck, R.; et al. Emerging evidence on the pathobiology of mucositis. Support. Care Cancer 2013, 21, 2075–2083. [Google Scholar] [CrossRef] [PubMed]
- Villa, A.; Sonis, S.T. Mucositis: Pathobiology and management. Curr. Opin. Oncol. 2015, 27, 159–164. [Google Scholar] [CrossRef] [PubMed]
- Nugent, S.G.; Kumar, D.; Rampton, D.S.; Evans, D.F. Intestinal luminal pH in inflammatory bowel disease: Possible determinants and implications for therapy with aminosalicylates and other drugs. Gut 2001, 48, 571–577. [Google Scholar] [CrossRef]
- Zhang, X.; Han, Y.; Huang, W.; Jin, M.; Gao, Z. The influence of the gut microbiota on the bioavailability of oral drugs. Acta Pharm. Sin. B 2020, 11, 1789–1812. [Google Scholar] [CrossRef] [PubMed]
- Parsons, R.L. Drug Absorption in Gastrointestinal Disease With Particular Reference to Malabsorption Syndromes. Clin. Pharmacokinet. 1977, 2, 45–60. [Google Scholar] [CrossRef] [PubMed]
- Effinger, A.; O’Driscoll, C.M.; McAllister, M.; Fotaki, N. Impact of gastrointestinal disease states on oral drug absorption—Implications for formulation design—A PEARRL review. J. Pharm. Pharmacol. 2019, 71, 674–698. [Google Scholar] [CrossRef] [Green Version]
- Castoldi, A.; Favero de Aguiar, C.; Moraes-Vieira, P.; Olsen Saraiva Câmara, N. They Must Hold Tight: Junction Proteins, Microbiota And Immunity In Intestinal Mucosa. Curr. Protein Pept. Sci. 2015, 16, 655–671. [Google Scholar] [CrossRef]
- Groschwitz, K.R.; Hogan, S.P. Intestinal barrier function: Molecular regulation and disease pathogenesis. J. Allergy Clin. Immunol. 2009, 124, 2–3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, J.; Zhang, J.; Wang, R. Gut microbiota modulates drug pharmacokinetics. Drug Metab. Rev. 2018, 50, 357–368. [Google Scholar] [CrossRef] [PubMed]
- van Vliet, M.J.; Harmsen, H.J.M.; de Bont, E.S.J.M.; Tissing, W.J.E. The role of intestinal microbiota in the development and severity of chemotherapy-induced mucositis. PLoS Pathog. 2010, 6, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- da Silva Ferreira, A.R.; Wardill, H.R.; Tissing, W.J.E.; Harmsen, H.J.M. Pitfalls and novel experimental approaches to optimize microbial interventions for chemotherapy-induced gastrointestinal mucositis. Curr. Opin. Support. Palliat. Care 2020, 14, 127. [Google Scholar] [CrossRef] [PubMed]
- Gibson, R.J.; Bowen, J.M. Biomarkers of regimen-related mucosal injury. Cancer Treat. Rev. 2011, 37, 487–493. [Google Scholar] [CrossRef] [PubMed]
- Crenn, P.; Messing, B.; Cynober, L. Citrulline as a biomarker of intestinal failure due to enterocyte mass reduction. Clin. Nutr. 2008, 27, 328–339. [Google Scholar] [CrossRef] [PubMed]
- Fragkos, K.C.; Forbes, A. Citrulline as a marker of intestinal function and absorption in clinical settings: A systematic review and meta-analysis. United Eur. Gastroenterol. J. 2018, 6, 181–191. [Google Scholar] [CrossRef] [PubMed]
- Kuchay, R.A.H.; Bhatia, A.; Mahmood, A.; Anwar, M.; Mahmood, S. Brush border enzyme activity and expression of apoptotic marker genes in lycopene fed rats with 5-Fu induced gastrointestinal mucositis. Nutr. Aging 2015, 3, 1–8. [Google Scholar] [CrossRef] [Green Version]
- Vermette, D.; Hu, P.; Canarie, M.F.; Funaro, M.; Glover, J.; Pierce, R.W. Tight junction structure, function, and assessment in the critically ill: A systematic review. Intensive Care Med. Exp. 2018, 6, 1–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wardill, H.R.; Bowen, J.M.; Al-Dasooqi, N.; Sultani, M.; Bateman, E.; Stansborough, R.; Shirren, J.; Gibson, R.J. Irinotecan disrupts tight junction proteins within the gut. Cancer Biol. Ther. 2014, 15, 236–244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chelakkot, C.; Ghim, J.; Ryu, S.H. Mechanisms regulating intestinal barrier integrity and its pathological implications. Exp. Mol. Med. 2018, 50, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Keefe, D.M.K.; Brealey, J.; Goland, G.J.; Cummins, A.G. Chemotherapy for cancer causes apoptosis that precedes hypoplasia in crypts of the small intestine in humans. Gut 2000, 47, 632–637. [Google Scholar] [CrossRef] [Green Version]
- Ulluwishewa, D.; Anderson, R.C.; McNabb, W.C.; Moughan, P.J.; Wells, J.M.; Roy, N.C. Regulation of tight junction permeability by intestinal bacteria and dietary components. J. Nutr. 2011, 141, 769–776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fallingborg, J. Intraluminal pH of the human gastrointestinal tract. Dan. Med. Bull. 1999, 46, 183–196. [Google Scholar] [PubMed]
- Sasaki, Y.; Hada, R.; Nakajima, H.; Fukuda, S.; Munakata, A. Improved localizing method of radiopill in measurement of entire gastrointestinal pH profiles: Colonic luminal pH in normal subjects and patients with Crohn’s disease. Am. J. Gastroenterol. 1997, 92, 114–118. [Google Scholar] [PubMed]
- Gibson, R.J.; Keefe, D.M.K. Cancer chemotherapy-induced diarrhoea and constipation: Mechanisms of damage and prevention strategies. Support. Care Cancer Off. J. Multinatl. Assoc. Support. Care Cancer 2006, 14, 890–900. [Google Scholar] [CrossRef]
- McQuade, R.M.; Carbone, S.E.; Stojanovska, V.; Rahman, A.; Gwynne, R.M.; Robinson, A.M.; Goodman, C.A.; Bornstein, J.C.; Nurgali, K. Role of oxidative stress in oxaliplatin-induced enteric neuropathy and colonic dysmotility in mice. Br. J. Pharmacol. 2016, 173, 3502–3521. [Google Scholar] [CrossRef] [Green Version]
- Tamai, I. Molecular characterization of intestinal absorption of drugs by carrier- mediated transport mechanisms. Yakugaku Zasshi 1997, 117, 415–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wardill, H.R.; Gibson, R.J.; Van Sebille, Y.Z.A.; Secombe, K.R.; Coller, J.K.; White, I.A.; Manavis, J.; Hutchinson, M.R.; Staikopoulos, V.; Logan, R.M.; et al. Irinotecan-Induced gastrointestinal dysfunction and pain are mediated by common TLR4-dependent mechanisms. Mol. Cancer Ther. 2016, 15, 1376–1386. [Google Scholar] [CrossRef] [Green Version]
- McQuade, R.M.; Stojanovska, V.; Abalo, R.; Bornstein, J.C.; Nurgali, K. Chemotherapy-Induced Constipation and Diarrhea: Pathophysiology, Current and Emerging Treatments. Front. Pharmacol. 2016, 7, 414. [Google Scholar] [CrossRef] [Green Version]
- Deng, J.; Zhu, X.; Chen, Z.; Fan, C.H.; Kwan, H.S.; Wong, C.H.; Shek, K.Y.; Zuo, Z.; Lam, T.N. A Review of Food–Drug Interactions on Oral Drug Absorption. Drugs 2017, 77, 1833–1855. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Krekels, E.H.J.; Verweij, P.E.; Buil, J.B.; Knibbe, C.A.J.; Brüggemann, R.J.M. Pharmacokinetics and Pharmacodynamics of Posaconazole. Drugs 2020, 80, 671–695. [Google Scholar] [CrossRef] [Green Version]
- Press, A.G.; Hauptmann, I.A.; Hauptmann, L.; Fuchs, B.; Fuchs, M.; Ewe, K.; Ramadori, G. Gastrointestinal pH profiles in patients with inflammatory bowel disease. Aliment. Pharmacol. Ther. 1998, 12, 673–678. [Google Scholar] [CrossRef]
- Vertzoni, M.; Augustijns, P.; Grimm, M.; Koziolek, M.; Lemmens, G.; Parrott, N.; Pentafragka, C.; Reppas, C.; Rubbens, J.; Van Den Abeele, J.; et al. Impact of regional differences along the gastrointestinal tract of healthy adults on oral drug absorption: An UNGAP review. Eur. J. Pharm. Sci. 2019, 134, 153–175. [Google Scholar] [CrossRef]
- Moghimipour, E.; Ameri, A.; Handali, S. Absorption-Enhancing Effects of Bile Salts. Molecules 2015, 20, 14451–14473. [Google Scholar] [CrossRef] [Green Version]
- Sinko, P.J.; Amidon, G.L. Characterization of the Oral Absorption of β-Lactam Antibiotics II: Competitive Absorption and Peptide Carrier Specificity. J. Pharm. Sci. 1989, 78, 723–727. [Google Scholar] [CrossRef] [PubMed]
- Krishna, G.; Ma, L.; Martinho, M.; O’Mara, E. Single-dose phase I study to evaluate the pharmacokinetics of posaconazole in new tablet and capsule formulations relative to oral suspension. Antimicrob. Agents Chemother. 2012, 56, 4196–4201. [Google Scholar] [CrossRef] [Green Version]
- Zama, D.; Gori, D.; Muratore, E.; Leardini, D.; Rallo, F.; Turroni, S.; Prete, A.; Brigidi, P.; Pession, A.; Masetti, R. Enteral versus Parenteral Nutrition as Nutritional Support after Allogeneic Hematopoietic Stem Cell Transplantation: A Systematic Review and Meta-Analysis. Transplant. Cell. Ther. 2021, 27, 180.e1–180.e8. [Google Scholar] [CrossRef] [PubMed]
- Wardill, H.R.; Bowen, J.M. Chemotherapy-induced mucosal barrier dysfunction: An updated review on the role of intestinal tight junctions. Curr. Opin. Support. Palliat. Care 2013, 7, 155–161. [Google Scholar] [CrossRef] [PubMed]
- Alexander, J.L.; Wilson, I.D.; Teare, J.; Marchesi, J.R.; Nicholson, J.K.; Kinross, J.M. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 356–365. [Google Scholar] [CrossRef] [PubMed]
- Sokol, H.; Pigneur, B.; Watterlot, L.; Lakhdari, O.; Bermúdez-Humarán, L.G.; Gratadoux, J.-J.; Blugeon, S.; Bridonneau, C.; Furet, J.-P.; Corthier, G.; et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc. Natl. Acad. Sci. USA 2008, 105, 16731–16736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barcenilla, A.; Pryde, S.E.; Martin, J.C.; Duncan, S.H.; Stewart, C.S.; Henderson, C.; Flint, H.J. Phylogenetic relationships of butyrate-producing bacteria from the human gut. Appl. Environ. Microbiol. 2000, 66, 1654–1661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hamouda, N.; Sano, T.; Oikawa, Y.; Ozaki, T.; Shimakawa, M.; Matsumoto, K.; Amagase, K.; Higuchi, K.; Kato, S. Apoptosis, Dysbiosis and Expression of Inflammatory Cytokines are Sequential Events in the Development of 5-Fluorouracil-Induced Intestinal Mucositis in Mice. Basic Clin. Pharmacol. Toxicol. 2017, 121, 159–168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gu, S.L.; Gong, Y.; Zhang, J.; Chen, Y.; Wu, Z.; Xu, Q.; Fang, Y.; Wang, J.; Tang, L.L. Effect of the short-term use of fluoroquinolone and β-lactam antibiotics on mouse gut microbiota. Infect. Drug Resist. 2020, 13, 4547. [Google Scholar] [CrossRef]
- Schröder, H.; Gustafsson, B.E. Azo Reduction of Salicyl-Azo-Sulphapyridine in Germ-free and Conventional Rats. Xenobiotica 1973, 3, 225–231. [Google Scholar] [CrossRef] [PubMed]
- Geller, L.T.; Barzily-Rokni, M.; Danino, T.; Jonas, O.H.; Shental, N.; Nejman, D.; Gavert, N.; Zwang, Y.; Cooper, Z.A.; Shee, K.; et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science 2017, 357, 1156–1160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gao, Y.; Shang, Q.; Li, W.; Guo, W.; Stojadinovic, A.; Mannion, C.; Man, Y.G.; Chen, T. Antibiotics for cancer treatment: A double-edged sword. J. Cancer 2020, 11, 5135. [Google Scholar] [CrossRef]
- Iida, N.; Dzutsev, A.; Stewart, C.A.; Smith, L.; Bouladoux, N.; Weingarten, R.A.; Molina, D.A.; Salcedo, R.; Back, T.; Cramer, S.; et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 2013, 342, 967–970. [Google Scholar] [CrossRef] [PubMed]
- Cheng, W.Y.; Wu, C.Y.; Yu, J. The role of gut microbiota in cancer treatment: Friend or foe? Gut 2020, 69, 1867–1876. [Google Scholar] [CrossRef]
- Romano, M.; Ricci, V.; Zarrilli, R. Mechanisms of disease: Helicobacter pylori-related gastric carcinogenesis--implications for chemoprevention. Nat. Clin. Pract. Gastroenterol. Hepatol. 2006, 3, 622–632. [Google Scholar] [CrossRef] [PubMed]
- Kwon, Y. Mechanism-based management for mucositis: Option for treating side effects without compromising the efficacy of cancer therapy. Onco. Targets. Ther. 2016, 9, 2007–2016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karbelkar, S.A.; Majumdar, A.S. Altered systemic bioavailability and organ distribution of azathioprine in methotrexate-induced intestinal mucositis in rats. Indian J. Pharmacol. 2016, 48, 241. [Google Scholar] [CrossRef]
- Fijlstra, M.; Rings, E.H.H.M.; Verkade, H.J.; van Dijk, T.H.; Kamps, W.A.; Tissing, W.J.E. Lactose maldigestion during methotrexate-induced gastrointestinal mucositis in a rat model. Am. J. Physiol. Gastrointest. Liver Physiol. 2011, 300, G283–G291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fijlstra, M.; Tissing, W.J.E.; Stellaard, F.; Verkade, H.J.; Rings, E.H.H.M. Reduced absorption of long-chain fatty acids during methotrexate-induced gastrointestinal mucositis in the rat. Clin. Nutr. 2013, 32, 452–459. [Google Scholar] [CrossRef] [PubMed]
- Kovanda, L.L.; Marty, F.M.; Maertens, J.; Desai, A.V.; Lademacher, C.; Engelhardt, M.; Lu, Q.; Hope, W.W. Impact of mucositis on absorption and systemic drug exposure of isavuconazole. Antimicrob. Agents Chemother. 2017, 61, e00101-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gattis, W.A.; Petros, W.P.; Pickard, W.W.; Drew, R.H.; May, D.B.; Hathorn, J.W. A prospective, open-label study of single-dose ciprofloxacin absorption after chemotherapy in patients with malignancy. Pharmacotherapy 1997, 17, 836–840. [Google Scholar] [CrossRef] [PubMed]
- Vanstraelen, K.; Prattes, J.; Maertens, J.; Lagrou, K.; Schoemans, H.; Peersman, N.; Vermeersch, P.; Theunissen, K.; Mols, R.; Augustijns, P.; et al. Posaconazole plasma exposure correlated to intestinal mucositis in allogeneic stem cell transplant patients. Eur. J. Clin. Pharmacol. 2016, 72, 953–963. [Google Scholar] [CrossRef] [PubMed]
- Alqahtani, S.; Mohamed, L.A.; Kaddoumi, A. Experimental models for predicting drug absorption and metabolism. Expert Opin. Drug Metab. Toxicol. 2013, 9, 1241–1254. [Google Scholar] [CrossRef] [PubMed]
- Xiang, Y.; Wen, H.; Yu, Y.; Li, M.; Fu, X.; Huang, S. Gut-on-chip: Recreating human intestine in vitro. J. Tissue Eng. 2020, 11, 2041731420965318. [Google Scholar] [CrossRef]
- Pilmis, B.; Le Monnier, A.; Zahar, J.R. Gut microbiota, antibiotic therapy and antimicrobial resistance: A narrative review. Microorganisms 2020, 8, 269. [Google Scholar] [CrossRef] [Green Version]
- Lee, S.H.; Choi, N.; Sung, J.H. Pharmacokinetic and pharmacodynamic insights from microfluidic intestine-on-a-chip models. Expert Opin. Drug Metab. Toxicol. 2019, 15, 1005–1019. [Google Scholar] [CrossRef] [PubMed]
Class of Agent | Chemotherapeutic Agent | Mechanism of Action | Gastrointestinal Toxicity | Intestinal Permeability |
---|---|---|---|---|
Alkylating agents [14] | Busulfan [21] Cyclophosphamide [22] Cisplatin [23] Melphalan [24] | Cross-link between DNA/RNA strands | Mucosal ulceration ↑* Cell loss ↓# Villus height ↑ Infiltrations [25,26] | Barrier disruption Bacterial translocation ↑ Permeability of the intestine (rats) [26] |
Antimetabolites | 5-Fluouracil [27,28] Methotrexate [29] Gemcitabine [30] | 5-FU conversion to fluorouridine monophosphate (FUMP) Competitive inhibition of dihydrofolate reductase via displacement of dihydrofolate Incorporation of pyramidine analog into DNA | ↑Inflammation ↑Crypt apoptosis ↑Villus atrophy ↑Increased risk of infection [31,32,33] | ↑ Ratio of crypt cells to villous enterocytes ↑ Intestinal permeability (associated with reduced Zonula Occludens-1 expression in rats) [34] Bacterial translocation [35,36] |
Topoisomerase I inhibitor | Irinotecan hydrochloride [37] | Inhibition of the DNA enzyme topoisomerase I | ↑ Villus atrophy Crypt ablation Goblet cell metaplasia ↑Inflammation [19] | ↓ Intestinal barrier function ↑ Intestinal permeability [38,39] |
FEC | Fluorouracil, Epirubicin, and Cyclophosphamide | 5-FU conversion to fluorouridine monophosphate (FUMP) + cross-link between DNA/RNA strands | ↑ Paracellular permeability ↓ Intestinal barrier function [40] | ↓ Glucagon-like peptide-2 circulating concentrations Mucosal ulcerations |
FOLFOX | 5-FU, leucovorin, and oxaliplatin | 5-FU conversion to fluorouridine monophosphate (FUMP) + inhibits the synthesis of deoxyribonucleic acid (DNA) | ↑Inflammation ↑Crypt apoptosis ↑Villus atrophy ↑Increased risk of infection [31,32,33] | ↑ Ratio of crypt cells to villous enterocytes ↑ Permeability of the intestine (rats) [34] ↑ Intestinal permeability (associated with reduced Zonula Occludens-1 expression in rats) Bacterial translocation [35,36] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
da Silva Ferreira, A.R.; Märtson, A.-G.; de Boer, A.; Wardill, H.R.; Alffenaar, J.-W.; Harmsen, H.J.M.; Tissing, W.J.E. Does Chemotherapy-Induced Gastrointestinal Mucositis Affect the Bioavailability and Efficacy of Anti-Infective Drugs? Biomedicines 2021, 9, 1389. https://doi.org/10.3390/biomedicines9101389
da Silva Ferreira AR, Märtson A-G, de Boer A, Wardill HR, Alffenaar J-W, Harmsen HJM, Tissing WJE. Does Chemotherapy-Induced Gastrointestinal Mucositis Affect the Bioavailability and Efficacy of Anti-Infective Drugs? Biomedicines. 2021; 9(10):1389. https://doi.org/10.3390/biomedicines9101389
Chicago/Turabian Styleda Silva Ferreira, Ana Rita, Anne-Grete Märtson, Alyse de Boer, Hannah R. Wardill, Jan-Willem Alffenaar, Hermie J. M. Harmsen, and Wim J. E. Tissing. 2021. "Does Chemotherapy-Induced Gastrointestinal Mucositis Affect the Bioavailability and Efficacy of Anti-Infective Drugs?" Biomedicines 9, no. 10: 1389. https://doi.org/10.3390/biomedicines9101389
APA Styleda Silva Ferreira, A. R., Märtson, A. -G., de Boer, A., Wardill, H. R., Alffenaar, J. -W., Harmsen, H. J. M., & Tissing, W. J. E. (2021). Does Chemotherapy-Induced Gastrointestinal Mucositis Affect the Bioavailability and Efficacy of Anti-Infective Drugs? Biomedicines, 9(10), 1389. https://doi.org/10.3390/biomedicines9101389