Mucosal Injury during Anti-Cancer Treatment: From Pathobiology to Bedside

Debora Basile, Paola Di Nardo, Carla Corvaja, Silvio Ken Garattini, Giacomo Pelizzari, Camilla Lisanti, Lucia Bortot, Lucia Da Ros, Michele Bartoletti, Matteo Borghi, Lorenzo Gerratana, Davide Lombardi, Fabio Puglisi, Debora Basile, Paola Di Nardo, Carla Corvaja, Silvio Ken Garattini, Giacomo Pelizzari, Camilla Lisanti, Lucia Bortot, Lucia Da Ros, Michele Bartoletti, Matteo Borghi, Lorenzo Gerratana, Davide Lombardi, Fabio Puglisi

Abstract

Mucositis is one of the most common debilitating side effects related to chemotherapy (CT), radiation therapy (RT), targeted agents and immunotherapy. It is a complex process potentially involving any portion of the gastrointestinal tract and injuring the mucosa, leading to inflammatory or ulcerative lesions. Mechanisms and clinical presentation can differ according both to the anatomic site involved (oral or gastrointestinal) and the treatment received. Understanding the pathophysiology and management of mucosal injury as a secondary effect of anti-cancer treatment is an important area of clinical research. Prophylaxis, early diagnosis, and adequate management of complications are essential to increase therapeutic success and, thus, improve the survival outcomes of cancer patients. This review focuses on the pathobiology and management guidelines for mucositis, a secondary effect of old and new anti-cancer treatments, highlighting recent advances in prevention and discussing future research options.

Keywords: anti-cancer treatments; mucosal impairment; mucosal injury; mucositis.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Mucositis pathobiology. This model has proven relevant for oral mucositis and gastrointestinal mucositis induced by chemotherapy and radiotherapy [7,13]. Conversely, there is still little information about the pathogenesis of mucositis associated with the newer anticancer treatment, but they likely differ from “classical mucositis”.
Figure 2
Figure 2
Dose modifications and management of mTOR inhibitors.
Figure 3
Figure 3
Dose modifications and management of CDK4/6 inhibitors.
Figure 4
Figure 4
Dose modifications and management of immunotherapy.

References

    1. Pico J.-L., Avila-Garavito A., Naccache P. Mucositis: Its occurrence, consequences, and treatment in the oncology setting. Oncologist. 1998;3:446–451.
    1. Bensinger W., Schubert M., Ang K.K., Brizel D., Brown E., Eilers J.G., Elting L., Mittal B.B., Schattner M.A., Speilberg R., et al. NCCN Task Force Report—Prevention and management of mucositis in cancer care. J. Natl. Compr. Cancer Netw. 2008;6:S1–S21.
    1. Sonis S.T., Elting L.S., Keefe D., Peterson D.E., Schubert M., Hauer-Jensen M., Bekele B.N., Raber-Durlacher J., Donnelly J.P., Rubenstein E.B., et al. Perspectives on cancer therapy-induced mucosal injury: Pathogenesis, measurement, epidemiology, and consequences for patients. Cancer. 2004;100:1995–2025. doi: 10.1002/cncr.20162.
    1. Mortensen H.R., Overgaard J., Specht L., Overgaard M., Johansen J., Evensen J.F., Andersen L.J., Andersen E., Grau C. Prevalence and peak incidence of acute and late normal tissue morbidity in the DAHANCA 6&7 randomised trial with accelerated radiotherapy for head and neck cancer. Radiother. Oncol. J. Eur. Soc. Ther. Radiol. Oncol. 2012;103:69–75.
    1. Blijlevens N., Schwenkglenks M., Bacon P., D’Addio A., Einsele H., Maertens J., Niederwieser D., Rabitsch W., Roosaar A., Ruutu T., et al. Prospective oral mucositis audit: Oral mucositis in patients receiving high-dose melphalan or BEAM conditioning chemotherapy--European Blood and Marrow Transplantation Mucositis Advisory Group. J. Clin. Oncol. 2008;26:1519–1525. doi: 10.1200/JCO.2007.13.6028.
    1. Chaveli-López B. Oral toxicity produced by chemotherapy: A systematic review. J. Clin. Exp. Dent. 2014;6:81–90. doi: 10.4317/jced.51337.
    1. Sonis S.T. The pathobiology of mucositis. Nat. Rev. Cancer. 2004;4:277–284. doi: 10.1038/nrc1318.
    1. Villa A., Sonis S.T. Mucositis: Pathobiology and management. Curr. Opin. Oncol. 2015;27:159–164. doi: 10.1097/CCO.0000000000000180.
    1. Vitale M.C., Modaffari C., Decembrino N., Zhou F.X., Zecca M., Defabianis P. Preliminary study in a new protocol for the treatment of oral mucositis in pediatric patients undergoing hematopoietic stem cell transplantation (HSCT) and chemotherapy (CT) Lasers Med. Sci. 2017;32:1423–1428. doi: 10.1007/s10103-017-2266-y.
    1. Lino M.D.M.da.C., de Carvalho F.B., de Oliveira L.R., Magalhães E.B., Pinheiro A.L.B., Ramalho L.M.P. Laser phototherapy as a treatment for radiotherapy-induced oral mucositis. Braz. Dent. J. 2011;22:162–165. doi: 10.1590/S0103-64402011000200013.
    1. McCarthy G.M., Awde J.D., Ghandi H., Vincent M., Kocha W.I. Risk factors associated with mucositis in cancer patients receiving 5-fluorouracil. Oral Oncol. 1998;34:484–490. doi: 10.1016/S1368-8375(98)00068-2.
    1. Vokurka S., Bystrická E., Koza V., Scudlová J., Pavlicová V., Valentová D., Visokaiová M., Misaniová L. Higher incidence of chemotherapy induced oral mucositis in females: A supplement of multivariate analysis to a randomized multicentre study. Support. Care Cancer. 2006;14:974–976. doi: 10.1007/s00520-006-0031-z.
    1. Wong H.M. Oral complications and management strategies for patients undergoing cancer therapy. ScientificWorldJournal. 2014;2014:581795. doi: 10.1155/2014/581795.
    1. Elting L.S., Cooksley C.D., Chambers M.S., Garden A.S. Risk, outcomes, and costs of radiation-induced oral mucositis among patients with head-and-neck malignancies. Int. J. Radiat. Oncol. Biol. Phys. 2007;68:1110–1120. doi: 10.1016/j.ijrobp.2007.01.053.
    1. Blijlevens N., Sonis S. Palifermin (recombinant keratinocyte growth factor-1): A pleiotropic growth factor with multiple biological activities in preventing chemotherapy- and radiotherapy-induced mucositis. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2007;18:817–826. doi: 10.1093/annonc/mdl332.
    1. Logan R.M., Gibson R.J., Bowen J.M., Stringer A.M., Sonis S.T., Keefe D.M.K. Characterisation of mucosal changes in the alimentary tract following administration of irinotecan: Implications for the pathobiology of mucositis. Cancer Chemother. Pharmacol. 2008;62:33–41. doi: 10.1007/s00280-007-0570-0.
    1. Boers-Doets C.B., Epstein J.B., Raber-Durlacher J.E., Ouwerkerk J., Logan R.M., Brakenhoff J.A., Lacouture M.E., Gelderblom H. Oral adverse events associated with tyrosine kinase and mammalian target of rapamycin inhibitors in renal cell carcinoma: A structured literature review. Oncologist. 2012;17:135–144. doi: 10.1634/theoncologist.2011-0111.
    1. Sonis S.T. The biologic role for nuclear factor-kappaB in disease and its potential involvement in mucosal injury associated with anti-neoplastic therapy. Crit. Rev. Oral Biol. Med. Off. Publ. Am. Assoc. Oral Biol. 2002;13:380–389. doi: 10.1177/154411130201300502.
    1. Logan R.M., Gibson R.J., Sonis S.T., Keefe D.M.K. Nuclear factor-kappaB (NF-κB) and cyclooxygenase-2 (COX-2) expression in the oral mucosa following cancer chemotherapy. Oral Oncol. 2007;43:395–401. doi: 10.1016/j.oraloncology.2006.04.011.
    1. Logan R.M., Stringer A.M., Bowen J.M., Gibson R.J., Sonis S.T., Keefe D.M.K. Serum levels of NFκB and pro-inflammatory cytokines following administration of mucotoxic drugs. Cancer Biol. Ther. 2008;7:1139–1145. doi: 10.4161/cbt.7.7.6207.
    1. Boers-Doets C.B., Raber-Durlacher J.E., Treister N.S., Epstein J.B., Arends A.B.P., Wiersma D.R., Lalla R.V., Logan R.M., van Erp N.P., Gelderblom H. Mammalian target of rapamycin inhibitor-associated stomatitis. Future Oncol. Lond. Engl. 2013;9:1883–1892. doi: 10.2217/fon.13.141.
    1. Castellani P., Balza E., Rubartelli A. Inflammation, DAMPs, tumor development, and progression: A vicious circle orchestrated by redox signaling. Antioxid. Redox Signal. 2014;20:1086–1097. doi: 10.1089/ars.2012.5164.
    1. Logan R.M., Stringer A.M., Bowen J.M., Yeoh A.S.-J., Gibson R.J., Sonis S.T., Keefe D.M.K. The role of pro-inflammatory cytokines in cancer treatment-induced alimentary tract mucositis: Pathobiology, animal models and cytotoxic drugs. Cancer Treat. Rev. 2007;33:448–460. doi: 10.1016/j.ctrv.2007.03.001.
    1. Al-Dasooqi N., Gibson R.J., Bowen J.M., Logan R.M., Stringer A.M., Keefe D.M. Matrix metalloproteinases are possible mediators for the development of alimentary tract mucositis in the dark agouti rat. Exp. Biol. Med. Maywood NJ. 2010;235:1244–1256. doi: 10.1258/ebm.2010.010082.
    1. Al-Ansari S., Zecha J.A.E.M., Barasch A., de Lange J., Rozema F.R., Raber-Durlacher J.E. Oral Mucositis Induced by Anticancer Therapies. Curr. Oral Health Rep. 2015;2:202–211. doi: 10.1007/s40496-015-0069-4.
    1. Sloan J.A., Goldberg R.M., Sargent D.J., Vargas-Chanes D., Nair S., Cha S.S., Novotny P.J., Poon M.A., O’Connell M.J., Loprinzi C.L. Women experience greater toxicity with fluorouracil-based chemotherapy for colorectal cancer. J. Clin. Oncol. 2002;20:1491–1498. doi: 10.1200/JCO.2002.20.6.1491.
    1. Chansky K., Benedetti J., Macdonald J.S. Differences in toxicity between men and women treated with 5-fluorouracil therapy for colorectal carcinoma. Cancer. 2005;103:1165–1171. doi: 10.1002/cncr.20878.
    1. Schwab M., Zanger U.M., Marx C., Schaeffeler E., Klein K., Dippon J., Kerb R., Blievernicht J., Fischer J., Hofmann U., et al. Role of genetic and nongenetic factors for fluorouracil treatment-related severe toxicity: A prospective clinical trial by the German 5-FU Toxicity Study Group. J. Clin. Oncol. 2008;26:2131–2138. doi: 10.1200/JCO.2006.10.4182.
    1. Sonis S.T., Sonis A.L., Lieberman A. Oral complications in patients receiving treatment for malignancies other than of the head and neck. J. Am. Dent. Assoc. 1939. 1978;97:468–472. doi: 10.14219/jada.archive.1978.0304.
    1. Zalcberg J., Kerr D., Seymour L., Palmer M. Haematological and non-haematological toxicity after 5-fluorouracil and leucovorin in patients with advanced colorectal cancer is significantly associated with gender, increasing age and cycle number. Tomudex International Study Group. Eur. J. Cancer Oxf. Engl. 1998;34:1871–1875. doi: 10.1016/S0959-8049(98)00259-7.
    1. Prado C.M.M., Baracos V.E., McCargar L.J., Mourtzakis M., Mulder K.E., Reiman T., Butts C.A., Scarfe A.G., Sawyer M.B. Body composition as an independent determinant of 5-fluorouracil-based chemotherapy toxicity. Clin. Cancer Res. 2007;13:3264–3268. doi: 10.1158/1078-0432.CCR-06-3067.
    1. Amstutz U., Henricks L.M., Offer S.M., Barbarino J., Schellens J.H.M., Swen J.J., Klein T.E., McLeod H.L., Caudle K.E., Diasio R.B., et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for Dihydropyrimidine Dehydrogenase Genotype and Fluoropyrimidine Dosing: 2017 Update. Clin. Pharmacol. Ther. 2018;103:210–216. doi: 10.1002/cpt.911.
    1. Cho H.-J., Park Y.S., Kang W.K., Kim J.-W., Lee S.-Y. Thymidylate synthase (TYMS) and dihydropyrimidine dehydrogenase (DPYD) polymorphisms in the Korean population for prediction of 5-fluorouracil-associated toxicity. Ther. Drug Monit. 2007;29:190–196. doi: 10.1097/FTD.0b013e318040b1fe.
    1. Robien K., Schubert M.M., Bruemmer B., Lloid M.E., Potter J.D., Ulrich C.M. Predictors of oral mucositis in patients receiving hematopoietic cell transplants for chronic myelogenous leukemia. J. Clin. Oncol. 2004;22:1268–1275. doi: 10.1200/JCO.2004.05.147.
    1. Sakamoto K., Oka M., Yoshino S., Hazama S., Abe T., Okayama N., Hinoda Y. Relation between cytokine promoter gene polymorphism and toxicity of 5-fluorouracil plus cisplatin chemotherapy. Oncol. Rep. 2006;16:381–387. doi: 10.3892/or.16.2.381.
    1. Sonis S.T. Regimen-related gastrointestinal toxicities in cancer patients. Curr. Opin. Support. Palliat. Care. 2010;4:26–30. doi: 10.1097/SPC.0b013e328335fb76.
    1. Bachour P.C., Sonis S.T. Predicting mucositis risk associated with cytotoxic cancer treatment regimens: Rationale, complexity, and challenges. Curr. Opin. Support. Palliat. Care. 2018;12:198–210. doi: 10.1097/SPC.0000000000000339.
    1. Stringer A.M., Logan R.M. The role of oral flora in the development of chemotherapy-induced oral mucositis. J. Oral Pathol. Med. 2015;44:81–87. doi: 10.1111/jop.12152.
    1. Ciorba M.A. A gastroenterologist’s guide to probiotics. Clin. Gastroenterol. Hepatol. 2012;10:960–968. doi: 10.1016/j.cgh.2012.03.024.
    1. Zelia M.G., Raquel D., Caio T.F., Danielle G.S. Mechanisms Underlying Chemotherapy-Associated Mucositis: The Role of Inflammatory Mediators and Potential Therapeutic Targets. Eur. Med. J. 2018;7:82–91.
    1. 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. doi: 10.1111/apt.12878.
    1. Srikrishna G., Freeze H.H. Endogenous damage-associated molecular pattern molecules at the crossroads of inflammation and cancer. Neoplasia. 2009;11:615–628. doi: 10.1593/neo.09284.
    1. Sonis S.T. Oral mucositis. Anticancer Drugs. 2011;22:607–612. doi: 10.1097/CAD.0b013e3283462086.
    1. Newton K., Dixit V.M. Signaling in innate immunity and inflammation. Cold Spring Harb. Perspect. Biol. 2012;4:a006049. doi: 10.1101/cshperspect.a006049.
    1. Wong D.V.T., Lima-Júnior R.C.P., Carvalho C.B.M., Borges V.F., Wanderley C.W.S., Bem A.X.C., Leite C.A.V.G., Teixeira M.A., Batista G.L.P., Silva R.L., et al. The Adaptor Protein Myd88 Is a Key Signaling Molecule in the Pathogenesis of Irinotecan-Induced Intestinal Mucositis. PLoS ONE. 2015;10:e0139985. doi: 10.1371/journal.pone.0139985.
    1. Ribeiro R.A., Wanderley C.W.S., Wong D.V.T., Mota J.M.S.C., Leite C.A.V.G., Souza M.H.L.P., Cunha F.Q., Lima-Júnior R.C.P. 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.
    1. Lima-Júnior R.C.P., Freitas H.C., Wong D.V.T., Wanderley C.W.S., Nunes L.G., Leite L.L., Miranda S.P., Souza M.H.L.P., Brito G.A.C., Magalhães P.J.C., et al. Targeted inhibition of IL-18 attenuates irinotecan-induced intestinal mucositis in mice. Br. J. Pharmacol. 2014;171:2335–2350. doi: 10.1111/bph.12584.
    1. Fernandes C., Wanderley C.W.S., Silva C.M.S., Muniz H.A., Teixeira M.A., Souza N.R.P., Cândido A.G.F., Falcão R.B., Souza M.H.L.P., Almeida P.R.C., et al. Role of regulatory T cells in irinotecan-induced intestinal mucositis. Eur. J. Pharm. Sci. Eur. Fed. Pharm. Sci. 2018;115:158–166. doi: 10.1016/j.ejps.2018.01.006.
    1. Guabiraba R., Besnard A.G., Menezes G.B., Secher T., Jabir M.S., Amaral S.S., Braun H., Lima-Junior R.C.P., Ribeiro R.A., Cunha F.Q., et al. IL-33 targeting attenuates intestinal mucositis and enhances effective tumor chemotherapy in mice. Mucosal Immunol. 2014;7:1079–1093. doi: 10.1038/mi.2013.124.
    1. Lalla R.V., Bowen J., Barasch A., Elting L., Epstein J., Keefe D.M., McGuire D.B., Migliorati C., Nicolatou-Galitis O., Peterson D.E., et al. MASCC/ISOO clinical practice guidelines for the management of mucositis secondary to cancer therapy. Cancer. 2014;120:1453–1461. doi: 10.1002/cncr.28592.
    1. Peterson D.E., Boers-Doets C.B., Bensadoun R.J., Herrstedt J. ESMO Guidelines Committee Management of oral and gastrointestinal mucosal injury: ESMO Clinical Practice Guidelines for diagnosis, treatment, and follow-up. Ann. Oncol. 2015;26:139–151. doi: 10.1093/annonc/mdv202.
    1. Rugo H.S., Seneviratne L., Beck J.T., Glaspy J.A., Peguero J.A., Pluard T.J., Dhillon N., Hwang L.C., Nangia C., Mayer I.A., et al. Prevention of everolimus-related stomatitis in women with hormone receptor-positive, HER2-negative metastatic breast cancer using dexamethasone mouthwash (SWISH): A single-arm, phase 2 trial. Lancet Oncol. 2017;18:654–662. doi: 10.1016/S1470-2045(17)30109-2.
    1. Culy C.R., Spencer C.M. Amifostine: An update on its clinical status as a cytoprotectant in patients with cancer receiving chemotherapy or radiotherapy and its potential therapeutic application in myelodysplastic syndrome. Drugs. 2001;61:641–684. doi: 10.2165/00003495-200161050-00012.
    1. Mallick S., Benson R., Rath G.K. Radiation induced oral mucositis: A review of current literature on prevention and management. Eur. Arch. Oto-Rhino-Laryngol. 2016;273:2285–2293. doi: 10.1007/s00405-015-3694-6.
    1. Epstein J.B., Silverman S., Paggiarino D.A., Crockett S., Schubert M.M., Senzer N.N., Lockhart P.B., Gallagher M.J., Peterson D.E., Leveque F.G. Benzydamine HCl for prophylaxis of radiation-induced oral mucositis: Results from a multicenter, randomized, double-blind, placebo-controlled clinical trial. Cancer. 2001;92:875–885. doi: 10.1002/1097-0142(20010815)92:4<875::AID-CNCR1396>;2-1.
    1. Mahood D.J., Dose A.M., Loprinzi C.L., Veeder M.H., Athmann L.M., Therneau T.M., Sorensen J.M., Gainey D.K., Mailliard J.A., Gusa N.L. Inhibition of fluorouracil-induced stomatitis by oral cryotherapy. J. Clin. Oncol. 1991;9:449–452. doi: 10.1200/JCO.1991.9.3.449.
    1. Cascinu S., Fedeli A., Fedeli S.L., Catalano G. Oral cooling (cryotherapy), an effective treatment for the prevention of 5-fluorouracil-induced stomatitis. Eur. J. Cancer. B. Oral Oncol. 1994;30:234–236. doi: 10.1016/0964-1955(94)90003-5.
    1. Sorensen J.B., Skovsgaard T., Bork E., Damstrup L., Ingeberg S. Double-blind, placebo-controlled, randomized study of chlorhexidine prophylaxis for 5-fluorouracil-based chemotherapy-induced oral mucositis with nonblinded randomized comparison to oral cooling (cryotherapy) in gastrointestinal malignancies. Cancer. 2008;112:1600–1606. doi: 10.1002/cncr.23328.
    1. Riley P., Glenny A.-M., Worthington H.V., Littlewood A., Clarkson J.E., McCabe M.G. Interventions for preventing oral mucositis in patients with cancer receiving treatment: Oral cryotherapy. Cochrane Database Syst. Rev. 2015;23:CD011552.
    1. Barasch A., Peterson D.E., Tanzer J.M., D’Ambrosio J.A., Nuki K., Schubert M.M., Franquin J.C., Clive J., Tutschka P. Helium-neon laser effects on conditioning-induced oral mucositis in bone marrow transplantation patients. Cancer. 1995;76:2550–2556. doi: 10.1002/1097-0142(19951215)76:12<2550::AID-CNCR2820761222>;2-X.
    1. Cowen D., Tardieu C., Schubert M., Peterson D., Resbeut M., Faucher C., Franquin J.C. Low energy Helium-Neon laser in the prevention of oral mucositis in patients undergoing bone marrow transplant: Results of a double blind randomized trial. Int. J. Radiat. Oncol. Biol. Phys. 1997;38:697–703. doi: 10.1016/S0360-3016(97)00076-X.
    1. Schubert M.M., Eduardo F.P., Guthrie K.A., Franquin J.-C., Bensadoun R.-J.J., Migliorati C.A., Lloid C.M.E., Eduardo C.P., Walter N.-F., Marques M.M., et al. A phase III randomized double-blind placebo-controlled clinical trial to determine the efficacy of low level laser therapy for the prevention of oral mucositis in patients undergoing hematopoietic cell transplantation. Support. Care Cancer. 2007;15:1145–1154. doi: 10.1007/s00520-007-0238-7.
    1. Ferreira B., da Motta Silveira F.M., de Orange F.A. Low-level laser therapy prevents severe oral mucositis in patients submitted to hematopoietic stem cell transplantation: A randomized clinical trial. Support. Care Cancer. 2016;24:1035–1042. doi: 10.1007/s00520-015-2881-8.
    1. Sonis S.T., Hashemi S., Epstein J.B., Nair R.G., Raber-Durlacher J.E. Could the biological robustness of low level laser therapy (Photobiomodulation) impact its use in the management of mucositis in head and neck cancer patients. Oral Oncol. 2016;54:7–14. doi: 10.1016/j.oraloncology.2016.01.005.
    1. Van Sebille Y.Z.A., Stansborough R., Wardill H.R., Bateman E., Gibson R.J., Keefe D.M. Management of Mucositis During Chemotherapy: From Pathophysiology to Pragmatic Therapeutics. Curr. Oncol. Rep. 2015;17:50. doi: 10.1007/s11912-015-0474-9.
    1. Arbabi-kalati F., Arbabi-kalati F., Deghatipour M., Ansari Moghadam A. Evaluation of the efficacy of zinc sulfate in the prevention of chemotherapy-induced mucositis: A double-blind randomized clinical trial. Arch. Iran. Med. 2012;15:413–417.
    1. Koukourakis M.I., Maltezos E. Amifostine administration during radiotherapy for cancer patients with genetic, autoimmune, metabolic and other diseases. Anticancer. Drugs. 2006;17:133–138. doi: 10.1097/00001813-200602000-00003.
    1. Nicolatou-Galitis O., Sarri T., Bowen J., Di Palma M., Kouloulias V.E., Niscola P., Riesenbeck D., Stokman M., Tissing W., Yeoh E., et al. Systematic review of amifostine for the management of oral mucositis in cancer patients. Support. Care Cancer. 2013;21:357–364. doi: 10.1007/s00520-012-1613-6.
    1. Prisciandaro L.D., Geier M.S., Butler R.N., Cummins A.G., Howarth G.S. Probiotic factors partially improve parameters of 5-fluorouracil-induced intestinal mucositis in rats. Cancer Biol. Ther. 2011;11:671–677. doi: 10.4161/cbt.11.7.14896.
    1. Abdel-Rahman O., ElHalawani H., Essam-Eldin S. S-1-based regimens and the risk of oral and gastrointestinal mucosal injury: A meta-analysis with comparison to other fluoropyrimidines. Expert Opin. Drug Saf. 2016;15:5–20. doi: 10.1517/14740338.2016.1105959.
    1. 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: Implications for chemotherapy-induced gut toxicity. Cancer Biol. Ther. 2014;15:236–244. doi: 10.4161/cbt.27222.
    1. Chen P., Mancini M., Sonis S.T., Fernandez-Martinez J., Liu J., Cohen E.E.W., Toback F.G. A Novel Peptide for Simultaneously Enhanced Treatment of Head and Neck Cancer and Mitigation of Oral Mucositis. PLoS ONE. 2016;11:e0152995. doi: 10.1371/journal.pone.0152995.
    1. Mayo B.J., Stringer A.M., Bowen J.M., Bateman E.H., Keefe D.M. Irinotecan-induced mucositis: The interactions and potential role of GLP-2 analogues. Cancer Chemother. Pharmacol. 2017;79:233–249. doi: 10.1007/s00280-016-3165-9.
    1. Keefe D.M., Elting L.S., Nguyen H.T., Grunberg S.M., Aprile G., Bonaventura A., Selva-Nayagam S., Barsevick A., Koczwara B., Sonis S.T. Risk and outcomes of chemotherapy-induced diarrhea (CID) among patients with colorectal cancer receiving multi-cycle chemotherapy. Cancer Chemother. Pharmacol. 2014;74:675–680. doi: 10.1007/s00280-014-2526-5.
    1. Hartmann J.T., Lipp H.-P. Toxicity of platinum compounds. Expert Opin. Pharmacother. 2003;4:889–901. doi: 10.1517/14656566.4.6.889.
    1. Aprile G., Rihawi K., De Carlo E., Sonis S.T. 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.
    1. 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. doi: 10.3389/fphar.2017.00354.
    1. Worthington H.V., Clarkson J.E., Bryan G., Furness S., Glenny A.-M., Littlewood A., McCabe M.G., Meyer S., Khalid T. Interventions for preventing oral mucositis for patients with cancer receiving treatment. Cochrane Database Syst. Rev. 2011;13:CD000978. doi: 10.1002/14651858.CD000978.pub5.
    1. Dodd M.J., Dibble S.L., Miaskowski C., MacPhail L., Greenspan D., Paul S.M., Shiba G., Larson P. Randomized clinical trial of the effectiveness of 3 commonly used mouthwashes to treat chemotherapy-induced mucositis. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 2000;90:39–47. doi: 10.1067/moe.2000.105713.
    1. Ala S., Saeedi M., Janbabai G., Ganji R., Azhdari E., Shiva A. Efficacy of Sucralfate Mouth Wash in Prevention of 5-fluorouracil Induced Oral Mucositis: A Prospective, Randomized, Double-Blind, Controlled Trial. Nutr. Cancer. 2016;68:456–463. doi: 10.1080/01635581.2016.1153666.
    1. Arash A., Somayeh A., Pardis P., Ahmad R.M. Efficacy of Topical and Systemic Vitamin E in Preventing Chemotherapy-Induced Oral Mucositis. Rep. Radiother. Oncol. 2015;2:e796.
    1. Cascinu S., Fedeli A., Fedeli S.L., Catalano G. Octreotide versus loperamide in the treatment of fluorouracil-induced diarrhea: A randomized trial. J. Clin. Oncol. 1993;11:148–151. doi: 10.1200/JCO.1993.11.1.148.
    1. Goumas P., Naxakis S., Christopoulou A., Chrysanthopoulos C., Nikolopoulou V., Kalofonos H.P. Octreotide acetate in the treatment of fluorouracil-induced diarrhea. Oncologist. 1998;3:50–53.
    1. Cascinu S., Fedeli A., Fedeli S.L., Catalano G. Control of chemotherapy-induced diarrhea with octreotide. A randomized trial with placebo in patients receiving cisplatin. Oncology. 1994;51:70–73. doi: 10.1159/000227313.
    1. Zidan J., Haim N., Beny A., Stein M., Gez E., Kuten A. Octreotide in the treatment of severe chemotherapy-induced diarrhea. Ann. Oncol. 2001;12:227–229. doi: 10.1023/A:1008372228462.
    1. Barbounis V., Koumakis G., Vassilomanolakis M., Demiri M., Efremidis A.P. Control of irinotecan-induced diarrhea by octreotide after loperamide failure. Support. Care Cancer. 2001;9:258–260. doi: 10.1007/s005200000220.
    1. Bonner J.A., Giralt J., Harari P.M., Baselga J., Spencer S., Bell D., Raben D., Liu J., Schulten J., Ang K.K., et al. Association of human papillomavirus and p16 status with mucositis and dysphagia for head and neck cancer patients treated with radiotherapy with or without cetuximab: Assessment from a phase 3 registration trial. Eur. J. Cancer Oxf. Engl. 1990. 2016;64:1–11. doi: 10.1016/j.ejca.2016.05.008.
    1. Bossi P., Bergamini C., Miceli R., Cova A., Orlandi E., Resteghini C., Locati L., Alfieri S., Imbimbo M., Granata R., et al. Salivary Cytokine Levels and Oral Mucositis in Head and Neck Cancer Patients Treated with Chemotherapy and Radiation Therapy. Int. J. Radiat. Oncol. Biol. Phys. 2016;96:959–966. doi: 10.1016/j.ijrobp.2016.08.047.
    1. Vera-Llonch M., Oster G., Hagiwara M., Sonis S. Oral mucositis in patients undergoing radiation treatment for head and neck carcinoma. Cancer. 2006;106:329–336. doi: 10.1002/cncr.21622.
    1. Trotti A., Bellm L.A., Epstein J.B., Frame D., Fuchs H.J., Gwede C.K., Komaroff E., Nalysnyk L., Zilberberg M.D. Mucositis incidence, severity and associated outcomes in patients with head and neck cancer receiving radiotherapy with or without chemotherapy: A systematic literature review. Radiother. Oncol. 2003;66:253–262. doi: 10.1016/S0167-8140(02)00404-8.
    1. McGuire D.B., Fulton J.S., Park J., Brown C.G., Correa M.E.P., Eilers J., Elad S., Gibson F., Oberle-Edwards L.K., Bowen J., et al. Systematic review of basic oral care for the management of oral mucositis in cancer patients. Support. Care Cancer. 2013;21:3165–3177. doi: 10.1007/s00520-013-1942-0.
    1. Brizel D.M., Wasserman T.H., Henke M., Strnad V., Rudat V., Monnier A., Eschwege F., Zhang J., Russell L., Oster W., et al. Phase III randomized trial of amifostine as a radioprotector in head and neck cancer. J. Clin. Oncol. 2000;18:3339–3345. doi: 10.1200/JCO.2000.18.19.3339.
    1. Büntzel J., Küttner K., Fröhlich D., Glatzel M. Selective cytoprotection with amifostine in concurrent radiochemotherapy for head and neck cancer. Ann. Oncol. 1998;9:505–509. doi: 10.1023/A:1008282412670.
    1. Yuan A., Sonis S. Emerging therapies for the prevention and treatment of oral mucositis. Expert Opin. Emerg. Drugs. 2014;19:343–351. doi: 10.1517/14728214.2014.946403.
    1. Tsujimoto T., Yamamoto Y., Wasa M., Takenaka Y., Nakahara S., Takagi T., Tsugane M., Hayashi N., Maeda K., Inohara H., et al. L-glutamine decreases the severity of mucositis induced by chemoradiotherapy in patients with locally advanced head and neck cancer: A double-blind, randomized, placebo-controlled trial. Oncol. Rep. 2015;33:33–39. doi: 10.3892/or.2014.3564.
    1. Rubenstein E.B., Peterson D.E., Schubert M., Keefe D., McGuire D., Epstein J., Elting L.S., Fox P.C., Cooksley C., Sonis S.T., et al. Clinical practice guidelines for the prevention and treatment of cancer therapy-induced oral and gastrointestinal mucositis. Cancer. 2004;100:2026–2046. doi: 10.1002/cncr.20163.
    1. Kochhar R., Patel F., Dhar A., Sharma S.C., Ayyagari S., Aggarwal R., Goenka M.K., Gupta B.D., Mehta S.K. Radiation-induced proctosigmoiditis. Prospective, randomized, double-blind controlled trial of oral sulfasalazine plus rectal steroids versus rectal sucralfate. Dig. Dis. Sci. 1991;36:103–107. doi: 10.1007/BF01300096.
    1. Martins F., de Oliveira M.A., Wang Q., Sonis S., Gallottini M., George S., Treister N. A review of oral toxicity associated with mTOR inhibitor therapy in cancer patients. Oral Oncol. 2013;49:293–298. doi: 10.1016/j.oraloncology.2012.11.008.
    1. Tian T., Li X., Zhang J. mTOR Signaling in Cancer and mTOR Inhibitors in Solid Tumor Targeting Therapy. Int. J. Mol. Sci. 2019;20:755. doi: 10.3390/ijms20030755.
    1. Bachelot T., Bourgier C., Cropet C., Ray-Coquard I., Ferrero J.-M., Freyer G., Abadie-Lacourtoisie S., Eymard J.-C., Debled M., Spaëth D., et al. Randomized phase II trial of everolimus in combination with tamoxifen in patients with hormone receptor-positive, human epidermal growth factor receptor 2-negative metastatic breast cancer with prior exposure to aromatase inhibitors: A GINECO study. J. Clin. Oncol. 2012;30:2718–2724. doi: 10.1200/JCO.2011.39.0708.
    1. Shameem R., Lacouture M., Wu S. Incidence and risk of high-grade stomatitis with mTOR inhibitors in cancer patients. Cancer Invest. 2015;33:70–77. doi: 10.3109/07357907.2014.1001893.
    1. Vigarios E., Epstein J.B., Sibaud V. Oral mucosal changes induced by anticancer targeted therapies and immune checkpoint inhibitors. Support. Care Cancer. 2017;25:1713–1739. doi: 10.1007/s00520-017-3629-4.
    1. Sonis S., Treister N., Chawla S., Demetri G., Haluska F. Preliminary characterization of oral lesions associated with inhibitors of mammalian target of rapamycin in cancer patients. Cancer. 2010;116:210–215. doi: 10.1002/cncr.24696.
    1. Paplomata E., Zelnak A., O’Regan R. Everolimus: Side effect profile and management of toxicities in breast cancer. Breast Cancer Res. Treat. 2013;140:453–462. doi: 10.1007/s10549-013-2630-y.
    1. Harris D.J., Eilers J., Harriman A., Cashavelly B.J., Maxwell C. Putting evidence into practice: Evidence-based interventions for the management of oral mucositis. Clin. J. Oncol. Nurs. 2008;12:141–152. doi: 10.1188/08.CJON.141-152.
    1. Nicolatou-Galitis O., Nikolaidi A., Athanassiadis I., Papadopoulou E., Sonis S. Oral ulcers in patients with advanced breast cancer receiving everolimus: A case series report on clinical presentation and management. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2013;116:110–116. doi: 10.1016/j.oooo.2013.02.022.
    1. Zecha J.A.E.M., Raber-Durlacher J.E., Nair R.G., Epstein J.B., Elad S., Hamblin M.R., Barasch A., Migliorati C.A., Milstein D.M.J., Genot M.-T., et al. Low-level laser therapy/photobiomodulation in the management of side effects of chemoradiation therapy in head and neck cancer: Part 2: Proposed applications and treatment protocols. Support. Care Cancer. 2016;24:2793–2805. doi: 10.1007/s00520-016-3153-y.
    1. Price T.J., Peeters M., Kim T.W., Li J., Cascinu S., Ruff P., Suresh A.S., Thomas A., Tjulandin S., Zhang K., et al. Panitumumab versus cetuximab in patients with chemotherapy-refractory wild-type KRAS exon 2 metastatic colorectal cancer (ASPECCT): A randomised, multicentre, open-label, non-inferiority phase 3 study. Lancet Oncol. 2014;15:569–579. doi: 10.1016/S1470-2045(14)70118-4.
    1. Cunningham D., Humblet Y., Siena S., Khayat D., Bleiberg H., Santoro A., Bets D., Mueser M., Harstrick A., Verslype C., et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N. Engl. J. Med. 2004;351:337–345. doi: 10.1056/NEJMoa033025.
    1. Sibaud V., Niec R.E., Schindler K., Busam K.J., Roché H., Modi S., Delord J.P., Lacouture M.E. Ado-trastuzumab emtansine-associated telangiectasias in metastatic breast cancer: A case series. Breast Cancer Res. Treat. 2014;146:451–456. doi: 10.1007/s10549-014-3001-z.
    1. Sibaud V., Vigarios E., Combemale P., Lamant L., Lacouture M.E., Lacaze J.L., Dalenc F., Delord J.P. T-DM1-related telangiectasias: A potential role in secondary bleeding events. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2015;26:436–437. doi: 10.1093/annonc/mdu533.
    1. Arena C., Troiano G., De Lillo A., Testa N.F., Lo Muzio L. Stomatitis and VEGFR-Tyrosine Kinase Inhibitors (VR-TKIs): A Review of Current Literature in 4369 Patients. BioMed Res. Int. 2018;2018:5035217. doi: 10.1155/2018/5035217.
    1. Yuan A., Kurtz S.L., Barysauskas C.M., Pilotte A.P., Wagner A.J., Treister N.S. Oral adverse events in cancer patients treated with VEGFR-directed multitargeted tyrosine kinase inhibitors. Oral Oncol. 2015;51:1026–1033. doi: 10.1016/j.oraloncology.2015.09.003.
    1. Kollmannsberger C., Bjarnason G., Burnett P., Creel P., Davis M., Dawson N., Feldman D., George S., Hershman J., Lechner T., et al. Sunitinib in metastatic renal cell carcinoma: Recommendations for management of noncardiovascular toxicities. Oncologist. 2011;16:543–553. doi: 10.1634/theoncologist.2010-0263.
    1. Califano R., Tariq N., Compton S., Fitzgerald D.A., Harwood C.A., Lal R., Lester J., McPhelim J., Mulatero C., Subramanian S., et al. Expert Consensus on the Management of Adverse Events from EGFR Tyrosine Kinase Inhibitors in the UK. Drugs. 2015;75:1335–1348. doi: 10.1007/s40265-015-0434-6.
    1. Lacouture M., Sibaud V. Toxic Side Effects of Targeted Therapies and Immunotherapies Affecting the Skin, Oral Mucosa, Hair, and Nails. Am. J. Clin. Dermatol. 2018;19:31–39. doi: 10.1007/s40257-018-0384-3.
    1. Krausova M., Korinek V. Wnt signaling in adult intestinal stem cells and cancer. Cell. Signal. 2014;26:570–579. doi: 10.1016/j.cellsig.2013.11.032.
    1. Xu H., Yu S., Liu Q., Yuan X., Mani S., Pestell R.G., Wu K. Recent advances of highly selective CDK4/6 inhibitors in breast cancer. J. Hematol. Oncol. 2017;10:97. doi: 10.1186/s13045-017-0467-2.
    1. Sledge G.W., Toi M., Neven P., Sohn J., Inoue K., Pivot X., Burdaeva O., Okera M., Masuda N., Kaufman P.A., et al. MONARCH 2: Abemaciclib in Combination With Fulvestrant in Women With HR+/HER2- Advanced Breast Cancer Who Had Progressed While Receiving Endocrine Therapy. J. Clin. Oncol. 2017;35:2875–2884. doi: 10.1200/JCO.2017.73.7585.
    1. Dickler M.N., Tolaney S.M., Rugo H.S., Cortés J., Diéras V., Patt D., Wildiers H., Hudis C.A., O’Shaughnessy J., Zamora E., et al. MONARCH 1, A Phase II Study of Abemaciclib, a CDK4 and CDK6 Inhibitor, as a Single Agent, in Patients with Refractory HR+/HER2- Metastatic Breast Cancer. Clin. Cancer Res. 2017;23:5218–5224. doi: 10.1158/1078-0432.CCR-17-0754.
    1. Lasheen S., Shohdy K.S., Kassem L., Abdel-Rahman O. Fatigue, alopecia and stomatitis among patients with breast cancer receiving cyclin-dependent kinase 4 and 6 inhibitors: A systematic review and meta-analysis. Expert Rev. Anticancer Ther. 2017;17:851–856. doi: 10.1080/14737140.2017.1355242.
    1. Thill M., Schmidt M. Management of adverse events during cyclin-dependent kinase 4/6 (CDK4/6) inhibitor-based treatment in breast cancer. Ther. Adv. Med. Oncol. 2018;10:1758835918793326. doi: 10.1177/1758835918793326.
    1. Gupta A., Felice K.M.D., Loftus E.V., Khanna S. Systematic review: Colitis associated with anti-CTLA-4 therapy. Aliment. Pharmacol. Ther. 2015;42:406–417. doi: 10.1111/apt.13281.
    1. Horvat T.Z., Adel N.G., Dang T.-O., Momtaz P., Postow M.A., Callahan M.K., Carvajal R.D., Dickson M.A., D’Angelo S.P., Woo K.M., et al. Immune-Related Adverse Events, Need for Systemic Immunosuppression, and Effects on Survival and Time to Treatment Failure in Patients With Melanoma Treated With Ipilimumab at Memorial Sloan Kettering Cancer Center. J. Clin. Oncol. 2015;33:3193–3198. doi: 10.1200/JCO.2015.60.8448.
    1. Robert C., Ribas A., Wolchok J.D., Hodi F.S., Hamid O., Kefford R., Weber J.S., Joshua A.M., Hwu W.-J., Gangadhar T.C., et al. Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: A randomised dose-comparison cohort of a phase 1 trial. Lancet. 2014;384:1109–1117. doi: 10.1016/S0140-6736(14)60958-2.
    1. Larkin J., Hodi F.S., Wolchok J.D. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N. Engl. J. Med. 2015;373:1270–1271. doi: 10.1056/NEJMoa1504030.
    1. Sibaud V., Meyer N., Lamant L., Vigarios E., Mazieres J., Delord J.P. Dermatologic complications of anti-PD-1/PD-L1 immune checkpoint antibodies. Curr. Opin. Oncol. 2016;28:254–263. doi: 10.1097/CCO.0000000000000290.
    1. Beck K.E., Blansfield J.A., Tran K.Q., Feldman A.L., Hughes M.S., Royal R.E., Kammula U.S., Topalian S.L., Sherry R.M., Kleiner D., et al. Enterocolitis in patients with cancer after antibody blockade of cytotoxic T-lymphocyte-associated antigen 4. J. Clin. Oncol. 2006;24:2283–2289. doi: 10.1200/JCO.2005.04.5716.
    1. Haanen J.B.A.G., Carbonnel F., Robert C., Kerr K.M., Peters S., Larkin J., Jordan K. On behalf of the ESMO Guidelines Committee Management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2017;28:119–142. doi: 10.1093/annonc/mdx225.
    1. Hofmann L., Forschner A., Loquai C., Goldinger S.M., Zimmer L., Ugurel S., Schmidgen M.I., Gutzmer R., Utikal J.S., Göppner D., et al. Cutaneous, gastrointestinal, hepatic, endocrine, and renal side-effects of anti-PD-1 therapy. Eur. J. Cancer Oxf. Engl. 1990. 2016;60:190–209. doi: 10.1016/j.ejca.2016.02.025.
    1. Collins L.K., Chapman M.S., Carter J.B., Samie F.H. Cutaneous adverse effects of the immune checkpoint inhibitors. Curr. Probl. Cancer. 2017;41:125–128. doi: 10.1016/j.currproblcancer.2016.12.001.
    1. Marthey L., Mateus C., Mussini C., Nachury M., Nancey S., Grange F., Zallot C., Peyrin-Biroulet L., Rahier J.F., Bourdier de Beauregard M., et al. Cancer Immunotherapy with Anti-CTLA-4 Monoclonal Antibodies Induces an Inflammatory Bowel Disease. J. Crohns Colitis. 2016;10:395–401. doi: 10.1093/ecco-jcc/jjv227.
    1. Berman D., Parker S.M., Siegel J., Chasalow S.D., Weber J., Galbraith S., Targan S.R., Wang H.L. Blockade of cytotoxic T-lymphocyte antigen-4 by ipilimumab results in dysregulation of gastrointestinal immunity in patients with advanced melanoma. Cancer Immun. 2010;10:11.
    1. Weber J., Thompson J.A., Hamid O., Minor D., Amin A., Ron I., Ridolfi R., Assi H., Maraveyas A., Berman D., et al. A randomized, double-blind, placebo-controlled, phase II study comparing the tolerability and efficacy of ipilimumab administered with or without prophylactic budesonide in patients with unresectable stage III or IV melanoma. Clin. Cancer Res. 2009;15:5591–5598. doi: 10.1158/1078-0432.CCR-09-1024.
    1. Epstein J.B., Gorsky M., Cabay R.J., Day T., Gonsalves W. Screening for and diagnosis of oral premalignant lesions and oropharyngeal squamous cell carcinoma: Role of primary care physicians. Can. Fam. Physician Med. Fam. Can. 2008;54:870–875.
    1. Leenstra J.L., Miller R.C., Qin R., Martenson J.A., Dornfeld K.J., Bearden J.D., Puri D.R., Stella P.J., Mazurczak M.A., Klish M.D., et al. Doxepin rinse versus placebo in the treatment of acute oral mucositis pain in patients receiving head and neck radiotherapy with or without chemotherapy: A phase III, randomized, double-blind trial (NCCTG-N09C6 [Alliance]) J. Clin. Oncol. 2014;32:1571–1577. doi: 10.1200/JCO.2013.53.2630.
    1. Guo S.-P., Wu S.-G., Zhou J., Feng H.-X., Li F.-Y., Wu Y.-J., Sun J.-Y., He Z.-Y. Transdermal fentanyl for pain due to chemoradiotherapy-induced oral mucositis in nasopharyngeal cancer patients: Evaluating efficacy, safety, and improvement in quality of life. Drug Des. Devel. Ther. 2014;8:497–503.
    1. Orvain C., Moles-Moreau M.P., François S., Mercier M., Moal F., Hamel J.F., Parot-Schinkel E., Ifrah N., Hunault-Berger M., Tanguy-Schmidt A. Miconazole mucoadhesive buccal tablet in high-dose therapy with autologous stem cell transplantation (HDT/ASCT)-induced mucositis. Support. Care Cancer. 2015;23:359–364. doi: 10.1007/s00520-014-2365-2.
    1. Rao N.G., Han G., Greene J.N., Tanvetyanon T., Kish J.A., De Conti R.C., Chuong M.D., Shridhar R., Biagioli M.C., Caudell J.J., et al. Effect of prophylactic fluconazole on oral mucositis and candidiasis during radiation therapy for head-and-neck cancer. Pract. Radiat. Oncol. 2013;3:229–233. doi: 10.1016/j.prro.2012.05.008.
    1. Arends J., Bachmann P., Baracos V., Barthelemy N., Bertz H., Bozzetti F., Fearon K., Hütterer E., Isenring E., Kaasa S., et al. ESPEN guidelines on nutrition in cancer patients. Clin. Nutr. Edinb. Scotl. 2017;36:11–48. doi: 10.1016/j.clnu.2016.07.015.
    1. Caccialanza R., Pedrazzoli P., Cereda E., Gavazzi C., Pinto C., Paccagnella A., Beretta G.D., Nardi M., Laviano A., Zagonel V. Nutritional Support in Cancer Patients: A Position Paper from the Italian Society of Medical Oncology (AIOM) and the Italian Society of Artificial Nutrition and Metabolism (SINPE) J. Cancer. 2016;7:131–135. doi: 10.7150/jca.13818.

Source: PubMed

3
Se inscrever