• No results found

The clinical implications of this work

The CRC-NORDIET study is designed to gain a better understanding of whether diet after diagnosis has impact on long-term outcomes, risk of recurrence and survival in patients with CRC. Due to lack of studies, CRC patients currently have no specific dietary

recommendations, other than the general recommendations for people who are not

diagnosed with cancer. Although observational studies suggest that diet after diagnosis may play a role, RCTs are urgently needed to confirm the potential of diet to improve outcomes in this patient population. Our study will therefore be of great importance for this group of patients.

The majority of the patients included in this work underwent nutritional assessments at a time point where cancer treatment was completed. Existing literature shows that

malnutrition is prevalent in CRC patients at the time of diagnosis, however, the high prevalence of malnutrition, FFM depletion and sarcopenia found in our population post-surgery, suggest that a significant proportion of these patients are still in need for nutritional intervention. Persistent nutritional problems may lead to continued loss of body weight and further deterioration of nutritional status, which may influence the further course of

survivorship. Although the cancer disease is cured and the patients are predicted good prognosis, decline in nutritional status may negatively impact quality of life, functional capacity and survival, as shown by Ravasco and coworkers who demonstrated that CRC patients who did not receive intensive nutritional counseling experienced decline in

nutritional status, quality of life and survival compared to the patients who received dietary counseling [45].

68

Most of the studies demonstrating associations between malnutrition and sarcopenia, and clinical outcome measurements and survival in CRC patients have included high proportions of patients with locally advanced or metastatic disease. The current work provides new knowledge about the high prevalence of malnutrition and sarcopenia in a CRC population without metastatic cancer. The relationship regarding low FFM and sarcopenia and clinical outcomes and survival will be investigated in future analyses in the CRC-NORDIET study.

In the current work, we demonstrated that by using the PG-SGA categories, only half of the patients with FFM depletion and less than half of the patients with sarcopenia were

identified by this tool. Consequently, a great proportion of the patients would incorrectly be evaluated as “well nourished” if PG-SGA was used as the only assessment tool for nutritional assessment. Based on our findings, we recommend that body composition analysis should be performed in addition to the PG-SGA.

PG-SGA was developed to evaluate the patient in terms of anabolic or catabolic, with emphasis on recent changes in body weight. However, in the context of the increasing prevalence of overweight and obesity in cancer patients, and the support from studies demonstrating that lean mass depletion may be masked by weight increase and increased BMI, monitoring only body weight will exclude important information regarding body composition. For example, if the patient continues to gain weight in terms of fat at the expense of lean body mass, it may lead to increased risk of sarcopenic obesity with all the health risks associated. Although the literature is scarce in non-metastatic CRC patients, preliminary data suggest that sarcopenia is associated with reduced survival [57] , and hence, these individuals should be identified and monitored.

In clinical practice, access to sophisticated instruments such as DXA may be limited for this purpose. In the current work, it was demonstrated that use of BIA resulted in estimates of FFM that were comparable to DXA on group level when the most appropriate equations were used. Interestingly, our work demonstrated that use of the various equations resulted in significant different estimates of FFM, and moreover, that the two BIA devices gave significant differences in FFM estimates, when using the same equation. These findings imply that BIA devices should not be used interchangeably in the clinical setting.

69

The results in paper 3 showed that the proportion of patients identified with low FFM according to the cutoff values by ESPEN, varied substantially depending on BIA device and equation used. Thus, some patients will not be correctly identified with low FFM as part of the malnutrition diagnosis if a suboptimal equation is used.

70

5 C ONCLUSIONS

The present thesis concludes with the following:

 The CRC-NORDIET study was developed and established in 2012. At the end of January 2018, 340 patients were enrolled in the study. It is estimated that the calculated sample size of 500 participants will be reached by the end of 2020. Of the 447 patients enrolled in the study so far, only 16 patients (5 %) have voluntary quitted. Based on the high retention rate in our study we suggest that the study design is conceived as feasible by the patients.

 In our population with non-metastatic CRC patients, 69 % were evaluated as well nourished (PG-SGA A) and 31 % patients were categorized as malnourished (PG-SGA B). No patients were evaluated as severely malnourished (PG-SGA C). Based on BIA assessments, low FFM was identified in 29 % of the patients. Twenty-two percent were diagnosed with sarcopenia. Our findings indicate that a significant portion of the patients were in need for nutritional intervention although most of the patients had already completed their cancer treatment.

 Despite that almost one third of the patients were found to have low FFM, PG-SGA did not identify these patients with sufficient sensitivity. The PG-SGA categorization classified only 50 % of the patients as malnourished (PG-SGA B). Moreover, only 64 % of the patients with low FFM were evaluated as muscle depleted by the physical examination in the PG-SGA.

 Both BIA devices showed good agreement compared to the reference method DXA, when using the appropriate BIA equation. Hence, BIA may be a useful tool to identify patients with low FFM and should accompany PG-SGA in the nutritional assessment of CRC patients. It is however, important to note that selection of BIA device and BIA equation may result in significantly different estimates of FFM, and the same BIA device and equation should be used when assessments are repeated during follow-up of the patient.

71

6 F UTURE PERSPECTIVES

Future analyses from the CRC study will examine the ability of the CRC-NORDIET intervention to improve nutritional status as well as to maintain or increase FFM. In cancer patients with metastatic disease, it is well established that the catabolic state favors an ongoing loss of skeletal muscle, a process that cannot be reversed by conventional nutrition care. In contrast, there is limited data concerning the ability of nutritional interventions to improve nutritional status and preserve or improve muscle mass in patients with localized cancer.

The dietary intervention in the CRC-NORDIET study was not specifically designed to achieve beneficial outcomes in terms of muscle mass. However, the dietary counseling aims to ensure adequate energy and protein intake, which is of importance to prevent weight loss and depletion of fat-free mass. Moreover, the recommended diet in the CRC-NORDIET targets a healthy body composition, including prevention of overweight and obesity. Future analyses from the CRC study will examine changes in fat-free mass assessed by BIA and DXA from baseline and during follow-up. Comparison of BIA and DXA at several time points will provide interesting information regarding the agreement between methods.

Although this work suggest that BIA should be applied in combination with PG-SGA to sufficiently identify patients with FFM depletion, it will be interesting to investigate whether the combination of PG-SGA and BIA will predict morbidity and mortality better than each method separately.

72

7 R EFERENCES

1. Food, nutrition, physical activity, and the prevention of cancer: a global perspective.

2007, Washington DC: American Institute for Cancer Research. 517 s. : fig. + 1 optisk plate (CD-ROM).

2. Cancer Prevention and Survival: Summary of global evidence on diet, weight, physical activity & what increases or decreases your risk of cancer. World Cancer Research Fund. 2016.

3. Ng, A.K. and L.B. Travis, Second primary cancers: an overview. Hematol Oncol Clin North Am, 2008. 22(2): p. 271-89, vii.

4. Demark-Wahnefried, W., et al., Riding the crest of the teachable moment: promoting long-term health after the diagnosis of cancer. J Clin Oncol, 2005. 23(24): p. 5814-30.

5. Fleming, M., et al., Colorectal carcinoma: Pathologic aspects. J Gastrointest Oncol, 2012. 3(3): p. 153-73.

6. Arnold, C.N., et al., Molecular pathogenesis of colorectal cancer: implications for molecular diagnosis. Cancer, 2005. 104(10): p. 2035-47.

7. Brenner, H., M. Kloor, and C.P. Pox, Colorectal cancer. Lancet, 2014. 383(9927): p.

1490-502.

8. Hamilton, W., et al., Clinical features of colorectal cancer before diagnosis: a population-based case-control study. Br J Cancer, 2005. 93(4): p. 399-405.

9. Majumdar, S.R., R.H. Fletcher, and A.T. Evans, How does colorectal cancer present?

Symptoms, duration, and clues to location. Am J Gastroenterol, 1999. 94(10): p. 3039-45.

10. Edna, T.H., et al., Prevalence of anaemia at diagnosis of colorectal cancer:

assessment of associated risk factors. Hepatogastroenterology, 2012. 59(115): p. 713-6.

11. Nesbakken, A. and M. Gaard, [Surgical treatment of colon cancer]. Tidsskr Nor Laegeforen, 2007. 127(22): p. 2942-5.

12. Nasjonalt handlingsprogram med retningslinjer for diagnostikk, behandling og oppfølging av kreft i tykktarm og endetarm. 2015, Helsedirektoratet.

13. AJCC Cancer Staging. 23 May 2017]; Available from: https://cancerstaging.org.

14. Dienstmann, R., et al., Consensus molecular subtypes and the evolution of precision medicine in colorectal cancer. Nat Rev Cancer, 2017. 17(2): p. 79-92.

15. Guinney, J., et al., The consensus molecular subtypes of colorectal cancer. Nat Med, 2015. 21(11): p. 1350-6.

16. World Cancer Reserach Fund International. 25 May 2017]; Available from:

http://www.wcrf.org/.

17. Kreftregisteret, Cancer in Norway 2015. 2015.

18. Årsrapport 2015 for tykk- og endetarmskreft 2015, Kreftregisteret.

19. Nedrebo, B.S., et al., Survival effect of implementing national treatment strategies for curatively resected colonic and rectal cancer. Br J Surg, 2011. 98(5): p. 716-23.

20. Cancer Registry of Norway. 2015; Available from: http://kreftregisteret.no/.

21. Johns, L.E. and R.S. Houlston, A systematic review and meta-analysis of familial colorectal cancer risk. Am J Gastroenterol, 2001. 96(10): p. 2992-3003.

22. Colorectal Cancer 2011 Report: Food, Nutrition, Physical Activity, and the Prevention of Colorectal Cancer. 2011.

23. Continous Update Project. Diet, nutrition, physical activity and colorectal cancer.

2017.

73

24. Cederholm, T., et al., ESPEN guidelines on definitions and terminology of clinical nutrition. Clin Nutr, 2017. 36(1): p. 49-64.

25. Cederholm, T., et al., Diagnostic criteria for malnutrition - An ESPEN Consensus Statement. Clin Nutr, 2015. 34(3): p. 335-40.

26. White, J.V., et al., Consensus statement: Academy of Nutrition and Dietetics and American Society for Parenteral and Enteral Nutrition: characteristics recommended for the identification and documentation of adult malnutrition (undernutrition). JPEN J Parenter Enteral Nutr, 2012. 36(3): p. 275-83.

27. Helsedirektoratet, Nasjonale faglige retningslinjer for forebygging og behandling av underernæing. 2013.

28. Organization, W.H. International classification of diseases. Available from:

http://www.who.int/classifications/icd/en/.

29. Prado, C.M. and S.B. Heymsfield, Lean tissue imaging: a new era for nutritional assessment and intervention. JPEN J Parenter Enteral Nutr, 2014. 38(8): p. 940-53.

30. Buttgereit, F. and G.R. Burmester, Rheumatoid arthritis: Glucocorticoid therapy and body composition. Nat Rev Rheumatol, 2016. 12(8): p. 444-5.

31. Lieffers, J.R., et al., A viscerally driven cachexia syndrome in patients with advanced colorectal cancer: contributions of organ and tumor mass to whole-body energy demands. Am J Clin Nutr, 2009. 89(4): p. 1173-9.

32. Sanz-Paris, A., et al., Application of the new ESPEN definition of malnutrition in geriatric diabetic patients during hospitalization: A multicentric study. Clin Nutr, 2016. 35(6): p. 1564-1567.

33. Rondel, A.L., et al., The new ESPEN diagnostic criteria for malnutrition predict overall survival in hospitalised patients. Clin Nutr, 2016.

34. Cruz-Jentoft, A.J., et al., Sarcopenia: European consensus on definition and

diagnosis: Report of the European Working Group on Sarcopenia in Older People.

Age Ageing, 2010. 39(4): p. 412-23.

35. Fried, L.P., et al., Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci, 2001. 56(3): p. M146-56.

36. Rosenberg, I.H., Sarcopenia: origins and clinical relevance. J Nutr, 1997. 127(5 Suppl): p. 990s-991s.

37. Laviano, A., C. Gori, and S. Rianda, Sarcopenia and nutrition. Adv Food Nutr Res, 2014. 71: p. 101-36.

38. Visser, M., et al., Muscle mass, muscle strength, and muscle fat infiltration as predictors of incident mobility limitations in well-functioning older persons. J Gerontol A Biol Sci Med Sci, 2005. 60(3): p. 324-33.

39. Malietzis, G., et al., Influence of body composition profile on outcomes following colorectal cancer surgery. Br J Surg, 2016. 103(5): p. 572-80.

40. Prado, C.M., et al., Prevalence and clinical implications of sarcopenic obesity in patients with solid tumours of the respiratory and gastrointestinal tracts: a population-based study. Lancet Oncol, 2008. 9(7): p. 629-35.

41. Barret, M., et al., Nutritional status affects treatment tolerability and survival in metastatic colorectal cancer patients: results of an AGEO prospective multicenter study. Oncology, 2011. 81(5-6): p. 395-402.

42. Burden, S.T., et al., Nutritional status of preoperative colorectal cancer patients. J Hum Nutr Diet, 2010. 23(4): p. 402-7.

43. Daniele, A., et al., Assessment of Nutritional and Inflammatory Status to Determine the Prevalence of Malnutrition in Patients Undergoing Surgery for Colorectal Carcinoma. Anticancer Res, 2017. 37(3): p. 1281-1287.

74

44. Gupta, D., et al., Malnutrition was associated with poor quality of life in colorectal cancer: a retrospective analysis. J Clin Epidemiol, 2006. 59(7): p. 704-9.

45. Ravasco, P., I. Monteiro-Grillo, and M. Camilo, Individualized nutrition intervention is of major benefit to colorectal cancer patients: long-term follow-up of a randomized controlled trial of nutritional therapy. Am J Clin Nutr, 2012. 96(6): p. 1346-53.

46. Read, J.A., et al., Evaluation of nutritional and inflammatory status of advanced colorectal cancer patients and its correlation with survival. Nutr Cancer, 2006. 55(1):

p. 78-85.

47. Thoresen, L., et al., Nutritional status, cachexia and survival in patients with advanced colorectal carcinoma. Different assessment criteria for nutritional status provide unequal results. Clin Nutr, 2013. 32(1): p. 65-72.

48. Andreyev, H.J., et al., Why do patients with weight loss have a worse outcome when undergoing chemotherapy for gastrointestinal malignancies? Eur J Cancer, 1998.

34(4): p. 503-9.

49. Aaldriks, A.A., et al., Frailty and malnutrition predictive of mortality risk in older patients with advanced colorectal cancer receiving chemotherapy. J Geriatr Oncol, 2013. 4(3): p. 218-26.

50. Barret, M., et al., Sarcopenia is linked to treatment toxicity in patients with metastatic colorectal cancer. Nutr Cancer, 2014. 66(4): p. 583-9.

51. Ravasco, P., et al., Dietary counseling improves patient outcomes: a prospective, randomized, controlled trial in colorectal cancer patients undergoing radiotherapy. J Clin Oncol, 2005. 23(7): p. 1431-8.

52. Lieffers, J.R., et al., Sarcopenia is associated with postoperative infection and delayed recovery from colorectal cancer resection surgery. Br J Cancer, 2012. 107(6): p. 931-6.

53. Reisinger, K.W., et al., Functional compromise reflected by sarcopenia, frailty, and nutritional depletion predicts adverse postoperative outcome after colorectal cancer surgery. Ann Surg, 2015. 261(2): p. 345-52.

54. Huang, D.D., et al., Sarcopenia, as defined by low muscle mass, strength and physical performance, predicts complications after surgery for colorectal cancer. Colorectal Dis, 2015. 17(11): p. O256-64.

55. Mauricio, S.F., et al., Different nutritional assessment tools as predictors of

postoperative complications in patients undergoing colorectal cancer resection. Clin Nutr, 2017.

56. Prado, C.M., et al., Body composition as an independent determinant of

5-fluorouracil-based chemotherapy toxicity. Clin Cancer Res, 2007. 13(11): p. 3264-8.

57. Black, D., et al., Prognostic Value of Computed Tomography: Measured Parameters of Body Composition in Primary Operable Gastrointestinal Cancers. Ann Surg Oncol, 2017.

58. Martin, L., et al., Cancer cachexia in the age of obesity: skeletal muscle depletion is a powerful prognostic factor, independent of body mass index. J Clin Oncol, 2013.

31(12): p. 1539-47.

59. Mahan, L.K., Raymond, Janice L.,, Krause`s food and nutrition care process 14. ed.

2017, St.Louis, Missouri.

60. Thompson, K.L., et al., Oncology Evidence-Based Nutrition Practice Guideline for Adults. J Acad Nutr Diet, 2017. 117(2): p. 297-310.e47.

61. Ferguson, M., et al., Development of a valid and reliable malnutrition screening tool for adult acute hospital patients. Nutrition, 1999. 15(6): p. 458-64.

62. Kim, J.Y., et al., Development and validation of a nutrition screening tool for hospitalized cancer patients. Clin Nutr, 2011. 30(6): p. 724-9.

75

63. Stratton, R.J., et al., Malnutrition in hospital outpatients and inpatients: prevalence, concurrent validity and ease of use of the 'malnutrition universal screening tool' ('MUST') for adults. Br J Nutr, 2004. 92(5): p. 799-808.

64. Patient-Generated Subjective Global Assessment. 23.05.2017]; Available from:

http://pt-global.org/.

65. Detsky, A.S., et al., What is subjective global assessment of nutritional status? JPEN J Parenter Enteral Nutr, 1987. 11(1): p. 8-13.

66. Arends, J., et al., ESPEN guidelines on nutrition in cancer patients. Clin Nutr, 2017.

36(1): p. 11-48.

67. Kondrup, J., et al., Nutritional risk screening (NRS 2002): a new method based on an analysis of controlled clinical trials. Clin Nutr, 2003. 22(3): p. 321-36.

68. Mini Nutritional Assessment. [cited 2018 20.02.2018]; Available from:

http://www.mna-elderly.com/default.html.

69. Sealy, M.J., et al., Content validity across methods of malnutrition assessment in patients with cancer is limited. J Clin Epidemiol, 2016. 76: p. 125-36.

70. Tan, B.H., et al., Sarcopenia in an overweight or obese patient is an adverse prognostic factor in pancreatic cancer. Clin Cancer Res, 2009. 15(22): p. 6973-9.

71. Prado, C.M., L.A. Birdsell, and V.E. Baracos, The emerging role of computerized tomography in assessing cancer cachexia. Curr Opin Support Palliat Care, 2009. 3(4):

p. 269-75.

72. Thibault, R. and C. Pichard, The evaluation of body composition: a useful tool for clinical practice. Ann Nutr Metab, 2012. 60(1): p. 6-16.

73. Saltz, L.B., et al., Irinotecan fluorouracil plus leucovorin is not superior to

fluorouracil plus leucovorin alone as adjuvant treatment for stage III colon cancer:

results of CALGB 89803. J Clin Oncol, 2007. 25(23): p. 3456-61.

74. Calle, E.E., et al., The American Cancer Society Cancer Prevention Study II Nutrition Cohort: rationale, study design, and baseline characteristics. Cancer, 2002. 94(2): p.

500-11.

75. Meyerhardt, J.A., et al., Association of dietary patterns with cancer recurrence and survival in patients with stage III colon cancer. Jama, 2007. 298(7): p. 754-64.

76. McCullough, M.L., et al., Association between red and processed meat intake and mortality among colorectal cancer survivors. J Clin Oncol, 2013. 31(22): p. 2773-82.

77. Meyerhardt, J.A., et al., Dietary glycemic load and cancer recurrence and survival in patients with stage III colon cancer: findings from CALGB 89803. J Natl Cancer Inst, 2012. 104(22): p. 1702-11.

78. Fuchs, M.A., et al., Sugar-sweetened beverage intake and cancer recurrence and survival in CALGB 89803 (Alliance). PLoS One, 2014. 9(6): p. e99816.

79. Kostråd for å fremme folkehelsen og forebygge kroniske sykdommer : metodologi og vitenskapelig kunnskapsgrunnlag. 2011, Oslo: Helsedirektoratet.

80. Nielsen, L., Kostvejledning. 2016, København.

81. Hettema, J., J. Steele, and W.R. Miller, Motivational interviewing. Annu Rev Clin Psychol, 2005. 1: p. 91-111.

82. The Nutrition Care Process. Available from:

http://www.eatrightpro.org/resources/practice/practice-resources/nutrition-care-process.

83. Sibbald, B. and M. Roland, Understanding controlled trials. Why are randomised controlled trials important? Bmj, 1998. 316(7126): p. 201.

84. Carlson, M.D. and R.S. Morrison, Study design, precision, and validity in observational studies. J Palliat Med, 2009. 12(1): p. 77-82.

76

85. Gluud, L.L., Bias in clinical intervention research. Am J Epidemiol, 2006. 163(6): p.

493-501.

86. Wood, L., et al., Empirical evidence of bias in treatment effect estimates in controlled trials with different interventions and outcomes: meta-epidemiological study. Bmj, 2008. 336(7644): p. 601-5.

87. Kjaergard, L.L., J. Villumsen, and C. Gluud, Reported methodologic quality and discrepancies between large and small randomized trials in meta-analyses. Ann Intern Med, 2001. 135(11): p. 982-9.

88. Moher, D., et al., Does quality of reports of randomised trials affect estimates of intervention efficacy reported in meta-analyses? Lancet, 1998. 352(9128): p. 609-13.

89. Schulz, K.F., et al., Empirical evidence of bias. Dimensions of methodological quality associated with estimates of treatment effects in controlled trials. Jama, 1995. 273(5):

p. 408-12.

90. Balk, E.M., et al., Correlation of quality measures with estimates of treatment effect in meta-analyses of randomized controlled trials. Jama, 2002. 287(22): p. 2973-82.

91. Schmid, D. and M.F. Leitzmann, Association between physical activity and mortality among breast cancer and colorectal cancer survivors: a systematic review and meta-analysis. Ann Oncol, 2014. 25(7): p. 1293-311.

92. Courneya, K.S., et al., The Colon Health and Life-Long Exercise Change trial: a randomized trial of the National Cancer Institute of Canada Clinical Trials Group.

Curr Oncol, 2008. 15(6): p. 279-85.

93. Anderson, A.S., et al., "It makes you feel so full of life" LiveWell, a feasibility study of a personalised lifestyle programme for colorectal cancer survivors. Support Care Cancer, 2010. 18(4): p. 409-15.

94. Bourke, L., et al., Pragmatic lifestyle intervention in patients recovering from colon cancer: a randomized controlled pilot study. Arch Phys Med Rehabil, 2011. 92(5): p.

749-55.

95. Morey, M.C., et al., Effects of home-based diet and exercise on functional outcomes among older, overweight long-term cancer survivors: RENEW: a randomized controlled trial. Jama, 2009. 301(18): p. 1883-91.

96. Ostan, R., et al., Inflammaging and cancer: a challenge for the Mediterranean diet.

Nutrients, 2015. 7(4): p. 2589-621.

97. Trinchieri, G., Cancer and inflammation: an old intuition with rapidly evolving new concepts. Annu Rev Immunol, 2012. 30: p. 677-706.

98. Andersen, L.F., et al., Consumption of coffee is associated with reduced risk of death attributed to inflammatory and cardiovascular diseases in the Iowa Women's Health Study. Am J Clin Nutr, 2006. 83(5): p. 1039-46.

99. Jacobs, D.R., Jr., L.F. Andersen, and R. Blomhoff, Whole-grain consumption is associated with a reduced risk of noncardiovascular, noncancer death attributed to inflammatory diseases in the Iowa Women's Health Study. Am J Clin Nutr, 2007.

85(6): p. 1606-14.

100. Karlsen, A., et al., Bilberry juice modulates plasma concentration of NF-kappaB related inflammatory markers in subjects at increased risk of CVD. Eur J Nutr, 2010.

49(6): p. 345-55.

101. Kolberg, M., et al., Plant extracts of spices and coffee synergistically dampen nuclear factor-kappaB in U937 cells. Nutr Res, 2013. 33(10): p. 817-30.

102. Kolberg, M., et al., Tomato paste alters NF-kappaB and cancer-related mRNA

102. Kolberg, M., et al., Tomato paste alters NF-kappaB and cancer-related mRNA