Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Optimization of nutrition support practices early after hematopoietic cell transplantation

Abstract

Nutrition support is often required during hematopoietic cell transplant (HCT) given the gastrointestinal toxicity that frequently precludes adequate protein-calorie intake. This article reviews the latest evidence for enteral versus parenteral nutrition in the adult and pediatric HCT population and addresses key considerations as well as barriers to implement this in practice. Registered Dietitian Nutritionists are key members of the interdisciplinary team to proactively manage enteral nutrition support to provide timely, adequate protein and calories to help prevent malnutrition, loss of lean body mass, and functional decline as well as provide evidence-based diet recommendations. This article also reviews emerging research supporting the role of luminal nutrients to maintain microbiotal diversity.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Similar content being viewed by others

References

  1. Iestra JA, Fibbe WE, Zwinderman AH, Romijn JA, Kromhout D. Parenteral nutrition following intensive cytotoxic therapy: an exploratory study on the need for parenteral nutrition after various treatment approaches for haematological malignancies. Bone Marrow Transpl. 1999;23:933–9. https://doi.org/10.1038/sj.bmt.1701747.

    Article  CAS  Google Scholar 

  2. Arends J, Bachmann P, Baracos V, Barthelemy N, Bertz H, Bozzetti F, et al. ESPEN guidelines on nutrition in cancer patients. Clin Nutr. 2017;36:11–48. https://doi.org/10.1016/j.clnu.2016.07.015.

    Article  PubMed  Google Scholar 

  3. Beckerson J, Szydlo RM, Hickson M, Mactier CE, Innes AJ, Gabriel IH, et al. Impact of route and adequacy of nutritional intake on outcomes of allogeneic haematopoietic cell transplantation for haematologic malignancies. Clin Nutr. 2019;38:738–44. https://doi.org/10.1016/j.clnu.2018.03.008.

    Article  PubMed  Google Scholar 

  4. Clemmons AB, Orr J, Andrick B, Gandhi A, Sportes C, DeRemer D. Randomized, placebo-controlled, phase III trial of Fosaprepitant, Ondansetron, Dexamethasone (FOND) versus FOND Plus Olanzapine (FOND-O) for the prevention of chemotherapy-induced nausea and vomiting in patients with hematologic malignancies receiving highly emetogenic chemotherapy and hematopoietic cell transplantation regimens: the FOND-O trial. Biol Blood Marrow Transpl. 2018;24:2065–71. https://doi.org/10.1016/j.bbmt.2018.06.005.

    Article  CAS  Google Scholar 

  5. Pastore D, Bruno B, Carluccio P, Stella De Candia M, Mammoliti S, Borghero C, et al. Antiemetic prophylaxis in patients undergoing hematopoietic stem cell transplantation: a multicenter survey of the Gruppo Italiano Trapianto Midollo Osseo (GITMO) transplant programs. Ann Hematol. 2020;99:867–75. https://doi.org/10.1007/s00277-020-03945-3.

    Article  CAS  PubMed  Google Scholar 

  6. Walrath M, Bacon C, Foley S, Fung HC. Gastrointestinal side effects and adequacy of enteral intake in hematopoietic stem cell transplant patients. Nutr Clin Pract. 2015;30:305–10. https://doi.org/10.1177/0884533614547084.

    Article  PubMed  Google Scholar 

  7. Diaconescu R, Flowers CR, Storer B, Sorror ML, Maris MB, Maloney DG, et al. Morbidity and mortality with nonmyeloablative compared with myeloablative conditioning before hematopoietic cell transplantation from HLA-matched related donors. Blood. 2004;104:1550–8. https://doi.org/10.1182/blood-2004-03-0804.

    Article  CAS  PubMed  Google Scholar 

  8. Topcuoglu P, Arat M, Ozcan M, Arslan O, Ilhan O, Beksac M, et al. Case-matched comparison with standard versus reduced intensity conditioning regimen in chronic myeloid leukemia patients. Ann Hematol. 2012;91:577–86. https://doi.org/10.1007/s00277-011-1349-2.

    Article  PubMed  Google Scholar 

  9. Andersen S, Brown T, Kennedy G, Banks M. Implementation of an evidenced based nutrition support pathway for haematopoietic progenitor cell transplant patients. Clin Nutr. 2015;34:536–40. https://doi.org/10.1016/j.clnu.2014.06.006.

    Article  CAS  PubMed  Google Scholar 

  10. Doney K, McMillen K, Buono L, Deeg HJ, Gooley T. Impact of body mass index on outcomes of hematopoietic stem cell transplantation in adults. Biol Blood Marrow Transpl. 2019;25:613–20. https://doi.org/10.1016/j.bbmt.2018.10.006.

    Article  Google Scholar 

  11. Deeg HJ, Seidel K, Bruemmer B, Pepe MS, Appelbaum FR. Impact of patient weight on non-relapse mortality after marrow transplantation. Bone Marrow Transpl. 1995;15:461–8.

    CAS  Google Scholar 

  12. Andersen S, Weber N, Kennedy G, Brown T, Banks M, Bauer J. Tolerability of proactive enteral nutrition post allogeneic haematopoietic progenitor cell transplant: a randomised comparison to standard care. Clin Nutr. 2020;39:1364–70. https://doi.org/10.1016/j.clnu.2019.06.012.

    Article  PubMed  Google Scholar 

  13. Gyurkocza B, Sandmaier BM. Conditioning regimens for hematopoietic cell transplantation: one size does not fit all. Blood. 2014;124:344–53. https://doi.org/10.1182/blood-2014-02-514778.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Baumgartner A, Bargetzi A, Zueger N, Bargetzi M, Medinger M, Bounoure L, et al. Revisiting nutritional support for allogeneic hematologic stem cell transplantation-a systematic review. Bone Marrow Transpl. 2017;52:506–13. https://doi.org/10.1038/bmt.2016.310.

    Article  CAS  Google Scholar 

  15. Uderzo C, Rovelli A, Bonomi M, Fomia L, Pirovano L, Masera G. Total parenteral nutrition and nutritional assessment and leukaemic children undergoing bone marrow transplantation. Eur J Cancer. 1991;27:758–62. https://doi.org/10.1016/0277-5379(91)90183-e.

    Article  CAS  PubMed  Google Scholar 

  16. Yokoyama S, Fujimoto T, Mitomi T, Yabe M, Yabe H, Kato S. Use of total parenteral nutrition in pediatric bone marrow transplantation. Nutrition. 1989;5:27–30.

    CAS  PubMed  Google Scholar 

  17. Peric Z, Botti S, Stringer J, Krawczyk J, et al. Variability of nutritional practices in peritransplant period after allogeneic hematopoietic stem cell transplantation: a survey by the Complications and Quality of Life Working Party of the EBMT. Bone Marrow Transpl. 2018;53:1030–7. https://doi.org/10.1038/s41409-018-0137-1.

    Article  CAS  Google Scholar 

  18. Fuji S, Einsele H, Savani BN, Kapp M. Systematic nutritional support in allogeneic hematopoietic stem cell transplant recipients. Biol Blood Marrow Transpl. 2015;21:1707–13. https://doi.org/10.1016/j.bbmt.2015.07.003.

    Article  Google Scholar 

  19. Andersen S, Banks M, Brown T, Weber N, Kennedy G, Bauer J. Nutrition support during allogeneic stem cell transplantation: evidence versus practice. Support Care Cancer. Published online March 10, 2020. https://doi.org/10.1007/s00520-020-05397-x.

  20. Staffas A, Burgos da Silva M, van den Brink MRM. The intestinal microbiota in allogeneic hematopoietic cell transplant and graft-versus-host disease. Blood. 2017;129:927–33. https://doi.org/10.1182/blood-2016-09-691394.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Jenq RR, Taur Y, Devlin SM, Ponce DM, Goldberg JD, Ahr KF, et al. Intestinal blautia is associated with reduced death from graft-versus-host disease. Biol Blood Marrow Transpl. 2015;21:1373–83. https://doi.org/10.1016/j.bbmt.2015.04.016.

    Article  Google Scholar 

  22. D’Amico F, Biagi E, Rampelli S, Fiori J, Zama D, Soverini M, et al. Enteral nutrition in pediatric patients undergoing hematopoietic SCT promotes the recovery of gut microbiome homeostasis. Nutrients. 2019;11. https://doi.org/10.3390/nu11122958.

  23. White M, Murphy AJ, Hallahan A, Ware RS, Fraser C, Davies PSW. Survival in overweight and underweight children undergoing hematopoietic stem cell transplantation. Eur J Clin Nutr. 2012;66:1120–3. https://doi.org/10.1038/ejcn.2012.109.

    Article  CAS  PubMed  Google Scholar 

  24. Hoffmeister PA, Storer BE, Macris PC, Carpenter PA, Baker KS. Relationship of body mass index and arm anthropometry to outcomes after pediatric allogeneic hematopoietic cell transplantation for hematologic malignancies. Biol Blood Marrow Transpl. 2013;19:1081–6. https://doi.org/10.1016/j.bbmt.2013.04.017.

    Article  Google Scholar 

  25. Lipkin AC, Lenssen P, Dickson BJ. Nutrition issues in hematopoietic stem cell transplantation: state of the art. Nutr Clin Pract. 2005;20:423–39. https://doi.org/10.1177/0115426505020004423.

    Article  PubMed  Google Scholar 

  26. Baumgartner A, Zueger N, Bargetzi A, Medinger M, Passweg JR, Stanga Z, et al. Association of Nutritional Parameters with Clinical Outcomes in patients with acute myeloid leukemia undergoing haematopoietic stem cell transplantation. Ann Nutr Metab. 2016;69:89–98. https://doi.org/10.1159/000449451.

    Article  CAS  PubMed  Google Scholar 

  27. Horsley P, Bauer J, Gallagher B. Poor nutritional status prior to peripheral blood stem cell transplantation is associated with increased length of hospital stay. Bone Marrow Transpl. 2005;35:1113–6. https://doi.org/10.1038/sj.bmt.1704963.

    Article  CAS  Google Scholar 

  28. August DA, Huhmann MB, American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) Board of Directors. A.S.P.E.N. clinical guidelines: nutrition support therapy during adult anticancer treatment and in hematopoietic cell transplantation. J Parenter Enter Nutr. 2009;33:472–500. https://doi.org/10.1177/0148607109341804.

    Article  Google Scholar 

  29. Thompson JL, Duffy J. Nutrition support challenges in hematopoietic stem cell transplant patients. Nutr Clin Pract. 2008;23:533–46. https://doi.org/10.1177/0884533608323423.

    Article  PubMed  Google Scholar 

  30. Shono Y, van den Brink MRM. Gut microbiota injury in allogeneic haematopoietic stem cell transplantation. Nat Rev Cancer. 2018;18:283–95. https://doi.org/10.1038/nrc.2018.10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Halpern-Silveira D, Susin LRO, Borges LR, Paiva SI, Assunção MCF, Gonzalez MC. Body weight and fat-free mass changes in a cohort of patients receiving chemotherapy. Support Care Cancer. 2010;18:617–25. https://doi.org/10.1007/s00520-009-0703-6.

    Article  PubMed  Google Scholar 

  32. Ferguson ML, Bauer J, Gallagher B, Capra S, Christie DR, Mason BR. Validation of a malnutrition screening tool for patients receiving radiotherapy. Australas Radio. 1999;43:325–7. https://doi.org/10.1046/j.1440-1673.1999.433665.x.

    Article  CAS  Google Scholar 

  33. Ferguson M, Capra S, Bauer J, Banks M. Development of a valid and reliable malnutrition screening tool for adult acute hospital patients. Nutrition. 1999;15:458–64. https://doi.org/10.1016/s0899-9007(99)00084-2.

    Article  CAS  PubMed  Google Scholar 

  34. Isenring E, Bauer J, Capra S. The scored Patient-generated Subjective Global Assessment (PG-SGA) and its association with quality of life in ambulatory patients receiving radiotherapy. Eur J Clin Nutr. 2003;57:305–9. https://doi.org/10.1038/sj.ejcn.1601552.

    Article  CAS  PubMed  Google Scholar 

  35. Bauer J, Capra S, Ferguson M. Use of the scored Patient-Generated Subjective Global Assessment (PG-SGA) as a nutrition assessment tool in patients with cancer. Eur J Clin Nutr. 2002;56:779–85. https://doi.org/10.1038/sj.ejcn.1601412.

    Article  CAS  PubMed  Google Scholar 

  36. Isenring E, Cross G, Daniels L, Kellett E, Koczwara B. Validity of the malnutrition screening tool as an effective predictor of nutritional risk in oncology outpatients receiving chemotherapy. Support Care Cancer. 2006;14:1152–6. https://doi.org/10.1007/s00520-006-0070-5.

    Article  PubMed  Google Scholar 

  37. Becker P, Carney LN, Corkins MR, Monczka J, Smith E, Smith SE, et al. Consensus statement of the Academy of Nutrition and Dietetics/American Society for Parenteral and Enteral Nutrition: indicators recommended for the identification and documentation of pediatric malnutrition (undernutrition). Nutr Clin Pract. 2015;30:147–61. https://doi.org/10.1177/0884533614557642.

    Article  PubMed  Google Scholar 

  38. White M, Murphy AJ, Hastings Y, Shergold J, Young J, Montgomery C, et al. Nutritional status and energy expenditure in children pre-bone-marrow-transplant. Bone Marrow Transpl. 2005;35:775–9. https://doi.org/10.1038/sj.bmt.1704891.

    Article  CAS  Google Scholar 

  39. Steele C, Salazar A, Rypkema L. Utilization of a nutrition support algorithm reduces unnecessary parenteral nutrition use in pediatric oncology inpatients. J Acad Nutr Diet. 2016;116:1235–8. https://doi.org/10.1016/j.jand.2015.12.007.

    Article  PubMed  Google Scholar 

  40. Murphy AJ, White M, Viani K, Mosby TT. Evaluation of the nutrition screening tool for childhood cancer (SCAN). Clin Nutr. 2016;35:219–24. https://doi.org/10.1016/j.clnu.2015.02.009.

    Article  PubMed  Google Scholar 

  41. Co-Reyes E, Li R, Huh W, Chandra J. Malnutrition and obesity in pediatric oncology patients: causes, consequences, and interventions. Pediatr Blood Cancer. 2012;59:1160–7. https://doi.org/10.1002/pbc.24272.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Elia M, Stratton RJ. Considerations for screening tool selection and role of predictive and concurrent validity. Curr Opin Clin Nutr Metab Care. 2011;14:425–33. https://doi.org/10.1097/MCO.0b013e328348ef51.

    Article  PubMed  Google Scholar 

  43. Secker DJ, Jeejeebhoy KN. How to perform Subjective Global Nutritional assessment in children. J Acad Nutr Diet. 2012;112:424–.e6. https://doi.org/10.1016/j.jada.2011.08.039.

    Article  PubMed  Google Scholar 

  44. Bowman LC, Williams R, Sanders M, Ringwald-Smith K, Baker D, Gajjar A. Algorithm for nutritional support: experience of the Metabolic and Infusion Support Service of St. Jude Children’s Research Hospital. Int J Cancer Suppl. 1998;11:76–80.

    Article  CAS  Google Scholar 

  45. Fuji S, Rovó A, Ohashi K, Griffith M, Einsele H, Kapp M, et al. How do I manage hyperglycemia/post-transplant diabetes mellitus after allogeneic HSCT. Bone Marrow Transpl. 2016;51:1041–9. https://doi.org/10.1038/bmt.2016.81.

    Article  CAS  Google Scholar 

  46. Sopfe J, Pyle L, Keating AK, Campbell K, Liu AK, Wadwa RP, et al. Malglycemia is associated with poor outcomes in pediatric and adolescent hematopoietic stem cell transplant patients. Blood Adv. 2019;3:350–9. https://doi.org/10.1182/bloodadvances.2018021014.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Fuji S, Kim S-W, Mori S, Fukuda T, Kamiya S, Yamasaki S, et al. Hyperglycemia during the neutropenic period is associated with a poor outcome in patients undergoing myeloablative allogeneic hematopoietic stem cell transplantation. Transplantation. 2007;84:814–20. https://doi.org/10.1097/01.tp.0000296482.50994.1c.

    Article  PubMed  Google Scholar 

  48. Sheean PM, Freels SA, Helton WS, Braunschweig CA. Adverse clinical consequences of hyperglycemia from total parenteral nutrition exposure during hematopoietic stem cell transplantation. Biol Blood Marrow Transpl. 2006;12:656–64. https://doi.org/10.1016/j.bbmt.2006.01.010.

    Article  Google Scholar 

  49. Verdi Schumacher M, Moreira, Faulhaber GA. Nutritional status and hyperglycemia in the peritransplant period: a review of associations with parenteral nutrition and clinical outcomes. Rev Bras Hematol E Hemoter. 2017;39:155–62. https://doi.org/10.1016/j.bjhh.2016.09.016.

    Article  Google Scholar 

  50. Seguy D, Duhamel A, Rejeb MB, Gomez E, Buhl ND, Bruno B, et al. Better outcome of patients undergoing enteral tube feeding after myeloablative conditioning for allogeneic stem cell transplantation. Transplantation. 2012;94:287–94. https://doi.org/10.1097/TP.0b013e3182558f60.

    Article  CAS  PubMed  Google Scholar 

  51. Gonzales F, Bruno B, Alarcón Fuentes M, De Berranger E, Guimber D, Behal H, et al. Better early outcome with enteral rather than parenteral nutrition in children undergoing MAC allo-SCT. Clin Nutr. 2018;37:2113–21. https://doi.org/10.1016/j.clnu.2017.10.005.

    Article  CAS  PubMed  Google Scholar 

  52. Herrmann VM, Petruska PJ. Nutrition support in bone marrow transplant recipients. Nutr Clin Pract. 1993;8:19–27. https://doi.org/10.1177/011542659300800119.

    Article  CAS  PubMed  Google Scholar 

  53. Langdana A, Tully N, Molloy E, Bourke B, O’Meara A. Intensive enteral nutrition support in paediatric bone marrow transplantation. Bone Marrow Transpl. 2001;27:741–6. https://doi.org/10.1038/sj.bmt.1702855.

    Article  CAS  Google Scholar 

  54. Taur Y, Jenq RR, Perales M, Littman ER, Morjaria S, Ling L, et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood. 2014;124:1174–82. https://doi.org/10.1182/blood-2014-02-554725.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Holler E, Butzhammer P, Schmid K, Hundsrucker C, Koestler J, Peter K, et al. Metagenomic analysis of the stool microbiome in patients receiving allogeneic stem cell transplantation: loss of diversity is associated with use of systemic antibiotics and more pronounced in gastrointestinal graft-versus-host disease. Biol Blood Marrow Transpl. 2014;20:640–5. https://doi.org/10.1016/j.bbmt.2014.01.030.

    Article  Google Scholar 

  56. Hills RD, Pontefract BA, Mishcon HR, Black CA, Sutton SC, Theberge CR. Gut microbiome: profound implications for diet and disease. Nutrients. 2019;11. https://doi.org/10.3390/nu11071613.

  57. Andermann TM, Peled JU, Ho C, Reddy P, Riches M, Storb R, et al. The microbiome and hematopoietic cell transplantation: past, present, and future. Biol Blood Marrow Transpl. 2018;24:1322–40. https://doi.org/10.1016/j.bbmt.2018.02.009.

    Article  Google Scholar 

  58. Moody K, Charlson ME, Finlay J. The neutropenic diet: what’s the evidence? J Pediatr Hematol Oncol. 2002;24:717–21. https://doi.org/10.1097/00043426-200212000-00007.

    Article  PubMed  Google Scholar 

  59. USDA Food Safety and Insepection Service. Food safety for transplant recipients. https://www.fsis.usda.gov/shared/PDF/Food_Safety_for_Transplant_Recipients.pdf.

  60. Peled JU, Gomes ALC, Devlin SM, Littmann ER, Taur Y, Sung AD, et al. Microbiota as predictor of mortality in allogeneic hematopoietic-cell transplantation. N Engl J Med. 2020;382:822–34. https://doi.org/10.1056/NEJMoa1900623.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Stein-Thoeringer CK, Nichols KB, Lazrak A, Docampo MD, Slingerland AE, Slingerland JB, et al. Lactose drives Enterococcus expansion to promote graft-versus-host disease. Science. 2019;366:1143–9. https://doi.org/10.1126/science.aax3760.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Bafeta A, Koh M, Riveros C, Ravaud P.Harms reporting in randomized controlled trials of interventions aimed at modifying microbiota: a systematic review.Ann Intern Med. 2018;169:240–7. https://doi.org/10.7326/M18-0343.

    Article  PubMed  Google Scholar 

  63. Hassan H, Rompola M, Glaser AW, Kinsey SE, Phillips RS. Systematic review and meta-analysis investigating the efficacy and safety of probiotics in people with cancer. Support Care Cancer. 2018;26:2503–9. https://doi.org/10.1007/s00520-018-4216-z.

    Article  PubMed  Google Scholar 

  64. Doron S, Snydman DR. Risk and safety of probiotics. Clin Infect Dis. 2015;60:S129–134. https://doi.org/10.1093/cid/civ085.

    Article  PubMed  PubMed Central  Google Scholar 

  65. White JV, Guenter P, Jensen G, Malone A, Schofield M.Academy Malnutrition Work Group et al. Consensus statement of the Academy of Nutrition and Dietetics/American Society for Parenteral and Enteral Nutrition: characteristics recommended for the identification and documentation of adult malnutrition (undernutrition). J Acad Nutr Diet. 2012;112:730–8. https://doi.org/10.1016/j.jand.2012.03.012.

    Article  PubMed  Google Scholar 

  66. McGrath KH, Evans V, Yap J. Indications and patterns of use for parenteral nutrition in pediatric oncology. JPEN J Parenter Enter Nutr. 2020;44:632–8. https://doi.org/10.1002/jpen.1685.

    Article  Google Scholar 

  67. Deswarte-Wallace J, Firouzbakhsh S, Finklestein JZ. Using research to change practice: enteral feedings for pediatric oncology patients. J Pediatr Oncol Nurs. 2001;18:217–23. https://doi.org/10.1053/jpon.2001.26875.

    Article  CAS  PubMed  Google Scholar 

  68. Guièze R, Lemal R, Cabrespine A, Hermet E, Tournilhac O, Combal C, et al. Enteral versus parenteral nutritional support in allogeneic haematopoietic stem-cell transplantation. Clin Nutr. 2014;33:533–8. https://doi.org/10.1016/j.clnu.2013.07.012.

    Article  PubMed  Google Scholar 

  69. Mueller C. The ASPEN Adult Nutrition Support Core Curriculum; 2017. Accessed 5 Sep 2020. http://search.ebscohost.com/login.aspx?direct=true&scope=site&db=nlebk&db=nlabk&AN=1831579.

  70. Juvé-Udina M-E, Valls-Miró C, Carreño-Granero A, Martinez-Estalella G, Monterde-Prat D, Domingo-Felici C, et al. To return or to discard? Randomised trial on gastric residual volume management. Intensive Crit Care Nurs. 2009;25:258–67. https://doi.org/10.1016/j.iccn.2009.06.004.

    Article  PubMed  Google Scholar 

  71. Taylor SJ, Manara AR, Brown J. Treating delayed gastric emptying in critical illness: metoclopramide, erythromycin, and bedside (cortrak) nasointestinal tube placement. J Parenter Enter Nutr. 2010;34:289–94. https://doi.org/10.1177/0148607110362533.

    Article  Google Scholar 

  72. Patel RP, Canada TW, Nates JL. Bleeding associated with feeding tube placement in critically Ill oncology patients with thrombocytopenia. Nutr Clin Pract. 2016;31:111–5. https://doi.org/10.1177/0884533615598964.

    Article  CAS  PubMed  Google Scholar 

  73. da Silva JSV, Seres DS, Sabino K, Adams SC, Berdahl GJ, Citty SW, et al. ASPEN consensus recommendations for refeeding syndrome. Nutr Clin Pract. 2020;35:178–95. https://doi.org/10.1002/ncp.10474.

    Article  PubMed  Google Scholar 

  74. Charuhas Macris P, Schilling K, Palko R. Academy of Nutrition and Dietetics: revised 2017 standards of practice and standards of professional performance for registered dietitian nutritionists (competent, proficient, and expert) in oncology nutrition. J Acad Nutr Diet. 2018;118:736–.e42. https://doi.org/10.1016/j.jand.2018.01.012.

    Article  PubMed  Google Scholar 

  75. Academy Quality Management Committee. Academy of Nutrition and Dietetics: revised 2017 scope of practice for the registered dietitian nutritionist. J Acad Nutr Diet. 2018;118:141–65. https://doi.org/10.1016/j.jand.2017.10.002.

    Article  Google Scholar 

  76. Thompson KL, Elliott L, Fuchs-Tarlovsky V, Levin RM, Voss AC, Piemonte T. Oncology evidence-based nutrition practice guideline for adults. J Acad Nutr Diet. 2017;117:297–310.e47. https://doi.org/10.1016/j.jand.2016.05.010.

    Article  PubMed  Google Scholar 

  77. Andrassy RJ, Chwals WJ. Nutritional support of the pediatric oncology patient. Nutrition. 1998;14:124–9. https://doi.org/10.1016/s0899-9007(97)00225-6.

    Article  CAS  PubMed  Google Scholar 

  78. Bauer J, Jürgens H, Frühwald MC. Important aspects of nutrition in children with cancer. Adv Nutr. 2011;2:67–77. https://doi.org/10.3945/an.110.000141.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Bendelsmith CR, Linabery AM, Nickel AJ, Laquere RM, Ingram KM, Hansen MB, et al. Effects of proactive and rescue enteral tube feedings on weight change in children undergoing treatment for high-grade CNS tumors. Neuro-Oncol Pract. 2020;7:428–38. https://doi.org/10.1093/nop/npaa003.

    Article  Google Scholar 

  80. Hastings Y, White M, Young J. Enteral nutrition and bone marrow transplantation. J Pediatr Oncol Nurs. 2006;23:103–10. https://doi.org/10.1177/1043454205285866.

    Article  PubMed  Google Scholar 

  81. Ward EJ, Henry LM, Friend AJ, Wilkins S, Phillips RS. Nutritional support in children and young people with cancer undergoing chemotherapy. Cochrane Database Syst Rev. 2015;CD003298. https://doi.org/10.1002/14651858.CD003298.pub3.

  82. Evans J, Needle JJ, Hirani SP. Early outcomes of gastrostomy feeding in paediatric allogenic bone marrow transplantation: a retrospective cohort study. Clin Nutr ESPEN. 2019;31:71–79. https://doi.org/10.1016/j.clnesp.2019.02.014.

    Article  PubMed  Google Scholar 

  83. Hannah E, John RM. Everything the nurse practitioner should know about pediatric feeding tubes. J Am Assoc Nurse Pract. 2013;25:567–77. https://doi.org/10.1002/2327-6924.12075.

    Article  PubMed  Google Scholar 

  84. Azarnoush S, Bruno B, Beghin L, Guimber D, Nelken B, Yakoub-Agha I, et al. Enteral nutrition: a first option for nutritional support of children following allo-SCT? Bone Marrow Transpl. 2012;47:1191–5. https://doi.org/10.1038/bmt.2011.248.

    Article  CAS  Google Scholar 

  85. DeMille D, Deming P, Lupinacci P, Jacobs LA. The effect of the neutropenic diet in the outpatient setting: a pilot study. Oncol Nurs Forum. 2006;33:337–43. https://doi.org/10.1188/ONF.06.337-343.

    Article  PubMed  Google Scholar 

  86. Moody K, Finlay J, Mancuso C, Charlson M. Feasibility and safety of a pilot randomized trial of infection rate: neutropenic diet versus standard food safety guidelines. J Pediatr Hematol Oncol. 2006;28:126–33. https://doi.org/10.1097/01.mph.0000210412.33630.fb.

    Article  PubMed  Google Scholar 

  87. Trifilio S, Helenowski I, Giel M, Gobel B, Pi J, Greenberg D, et al. Questioning the role of a neutropenic diet following hematopoetic stem cell transplantation. Biol Blood Marrow Transpl. 2012;18:1385–90. https://doi.org/10.1016/j.bbmt.2012.02.015.

    Article  Google Scholar 

  88. Boyle NM, Podczervinski S, Jordan K, Stednick Z, Butler-Wu S, McMillen K, et al. Bacterial foodborne infections after hematopoietic cell transplantation. Biol Blood Marrow Transpl. 2014;20:1856–61. https://doi.org/10.1016/j.bbmt.2014.06.034.

    Article  Google Scholar 

  89. Taggart C, Neumann N, Alonso PB, Lane A, Pate A, Stegman A, et al. Comparing a neutropenic diet to a food safety-based diet in pediatric patients undergoing hematopoietic stem cell transplantation. Biol Blood Marrow Transpl. 2019;25:1382–6. https://doi.org/10.1016/j.bbmt.2019.03.017.

    Article  Google Scholar 

  90. Sonbol MB, Jain T, Firwana B, Hilal T, Deleon T, Murad A, et al. Neutropenic diets to prevent cancer infections: updated systematic review and meta-analysis. BMJ Support Palliat Care. 2019;9:425–33. https://doi.org/10.1136/bmjspcare-2018-001742.

    Article  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Kerry K. McMillen.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

McMillen, K.K., Coghlin-Dickson, T. & Adintori, P.A. Optimization of nutrition support practices early after hematopoietic cell transplantation. Bone Marrow Transplant 56, 314–326 (2021). https://doi.org/10.1038/s41409-020-01078-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41409-020-01078-9

This article is cited by

Search

Quick links