malnutrition and dietary interventions in colorectal

157
Malnutrition and dietary interventions in colorectal cancer patients Hanna Ræder PhD Thesis Department of Nutrition Institute of Basic Medical Sciences Faculty of Medicine UNIVERSITY OF OSLO 2018

Upload: others

Post on 05-Dec-2021

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Malnutrition and dietary interventions in colorectal

Malnutrition and dietary interventions in colorectal cancer patients

Hanna Ræder

PhD Thesis

Department of Nutrition Institute of Basic Medical Sciences

Faculty of Medicine

UNIVERSITY OF OSLO

2018

Page 2: Malnutrition and dietary interventions in colorectal

© Hanna Ræder, 2018 Series of dissertations submitted to the Faculty of Medicine, University of Oslo ISBN 978-82-8377-284-5 All rights reserved. No part of this publication may be reproduced or transmitted, in any form or by any means, without permission. Cover: Hanne Baadsgaard Utigard. Print production: Reprosentralen, University of Oslo.

Page 3: Malnutrition and dietary interventions in colorectal

III

ACKNOWLEDGEMENTS

This thesis was performed at the Department of Nutrition, University of Oslo and at The National Advisory Unit on Disease-related Malnutrition at the Oslo University Hospital. The work was funded by Helse Sør-Øst.

First of all, I would like to express my sincere gratitude to my supervisor professor Rune Blomhoff, for giving me this opportunity. Thank you for all the support and excellent guidance through all these years. I admire your enthusiasm, creativity, wide knowledge and your positive energy and good sense of humor. I would also like to thank my co-supervisor Sigbjørn Smeland for all the support, constructive feedbacks and valuable discussions during my work.

Christine Henriksen; your involvement in my work has been outstanding. Thank you for always being so positive, kind and for sharing so much of your time and expertise. I highly appreciate our friendship.

I would also like to thank all the great members of “Typisk Norsk-gjengen”; Hege Berg Henriksen, Ane Sørlie Kværner, Anne Juul Skjetne, Mari Bøe Sebelien, Ingvild Paur, Siv Kjølsrud Bøhn, Torgrim Langleite, Siv Åshild Billington, Kari Holte, Geir Florholmen, Magnhild Håskjold, Kristin S. Hustad, Jonas Grenne, Anne-Rikke O`de Fey Vilbo and Katrine Rolid. It has been a privilege to be part of this fantastic team and I have enjoyed all the time we have spent together.

Warm thanks to Kristine Lillebø Holm for the contribution in the Norwegian translation of the PG-SGA, and to Ingvild Paur for constructive feedbacks and excellent technical assistance in my thesis.

Thanks also to all the former and present members of Rune Blomhoff`s research group, for contributing to a nice and inspiring work environment. During my time as a Phd student I have met so many friendly colleagues at the Department of Nutrition and at the Oslo University Hospital. Thank you for all the nice lunches and coffee breaks and discussions about science and every-day life.

Thanks to my friends Ragnhild, Christina, Mari, Henriette, Ellen, Karen and Elisabeth for enriching my life with a lot of laughter and good moments.

I would also like to thank my family-in-law for all your support and interest in my work. Special thanks go to my lovely parents, Jaroslava and Helge, and my two brothers Helge and Sten, for your inspiration, endless love and for always believing in me.

Finally, to my closest family; my dear Lars and our wonderful sweethearts Elise and Helena – thank you for always reminding me of the most important things in life. I love you.

Oslo, February 2018 Hanna Ræder

Page 4: Malnutrition and dietary interventions in colorectal

IV

TABLE OF CONTENTS

Acknowledgements .................................................................................................................. III

Table of contents ...................................................................................................................... IV

List of tables and figures .......................................................................................................... VI

List of papers ........................................................................................................................... VII

Abbreviations ........................................................................................................................ VIII

1 Introduction ...................................................................................................................... 11

1.1 Dietary aspects of cancer ........................................................................................... 11

1.2 Colorectal cancer ....................................................................................................... 12

1.2.1 Diagnosis and staging ......................................................................................... 13

1.2.2 Incidence, mortality and survival ....................................................................... 15

1.2.3 Etiology .............................................................................................................. 18

1.2.4 Treatment of localized CRC ............................................................................... 19

1.2.5 Follow-up ........................................................................................................... 19

1.3 Colorectal cancer-associated malnutrition and sarcopenia ........................................ 20

1.3.1 Definitions and diagnostic criteria of malnutrition ............................................ 20

1.3.2 Fat-free mass depletion ...................................................................................... 24

1.3.3 Sarcopenia and sarcopenic obesity ..................................................................... 25

1.3.4 Malnutrition and sarcopenia in CRC patients ................................................... 27

1.4 Nutritional risk screening and assessment tools in oncology .................................... 29

1.5 The role of diet after CRC diagnosis ......................................................................... 34

1.5.1 Factors affecting change in dietary habits .......................................................... 36

2 Aims ................................................................................................................................. 39

3 Summary of papers ........................................................................................................... 40

3.1 Paper 1 ....................................................................................................................... 40

3.2 Paper 2 ....................................................................................................................... 41

3.3 Paper 3 ....................................................................................................................... 42

4 General discussion ............................................................................................................ 43

4.1 Study designs ............................................................................................................. 43

4.2 The Norwegian dietary guidelines and colorectal cancer survival (CRC-NORDIET)

study: a food-based multicenter randomized controlled trial ............................................... 45

4.2.1 Study participants ............................................................................................... 46

Page 5: Malnutrition and dietary interventions in colorectal

V

4.2.2 The control group ............................................................................................... 48

4.2.3 Blinding .............................................................................................................. 48

4.2.4 The impact of physical activity .......................................................................... 49

4.2.5 Long-term follow-up .......................................................................................... 50

4.2.6 The dietary intervention ..................................................................................... 50

4.2.7 Implementation of dietary intervention .............................................................. 52

4.2.8 Individualized nutritional counseling by a registered clinical dietitian ............. 52

4.2.9 Telephone and web-based approach .................................................................. 53

4.2.10 Motivational interviewing .................................................................................. 54

4.2.11 Long-term adherence to the dietary intervention ............................................... 55

4.2.12 Endpoints and power calculations in the CRC-NORDIET study ...................... 56

4.2.13 Status of the ongoing CRC-NORDIET study .................................................... 57

4.3 Nutritional assessment by PG-SGA .......................................................................... 58

4.4 Assessment of fat-free mass ...................................................................................... 60

4.4.1 Methods for assessment of FFM ........................................................................ 60

4.4.2 DXA ................................................................................................................... 61

4.4.3 BIA ..................................................................................................................... 62

4.5 The ability of PG-SGA to identify low FFM ............................................................ 63

4.6 The ability of BIA to assess FFM .............................................................................. 65

4.7 The clinical implications of this work ....................................................................... 67

5 Conclusions ...................................................................................................................... 70

6 Future perspectives ........................................................................................................... 71

7 References ........................................................................................................................ 72

Page 6: Malnutrition and dietary interventions in colorectal

VI

LIST OF TABLES AND FIGURES

Table 1. Pathological staging according to the TNM classification system

Table 2. The ESPEN diagnostic criteria for malnutrition

Table 3. The clinical characteristics for the diagnosis of malnutrition in the Consensus

Statement by the Academy/ASPEN

Table 4. The ICD-10 classification of malnutrition

Table 5. An overview of screening and assessment tools in oncology

Table 6. The Norwegian food-based dietary guidelines

Table 7. Study populations in the CRC study vs CRC reference population in Norway

Table 8. Status of participation in the ongoing CRC-NORDIET study

Figure 1. An anatomic overview of colon and rectum

Figure 2. The colorectal cancer stages

Figure 3. Age-standardized incidence, mortality and 5-year survival curves for CRC

Figure 4 A) Gender and age distribution in patients treated by surgery for primary colon

cancer stage I-III

Figure B) Gender and age distribution in patients treated by surgery for primary rectum

cancer stage I-III

Figure 5. Malnutrition sub categories.

Figure 6. The two-compartment model of body composition.

Figure 7. Nutrition disorders and nutrition related conditions.

Figure 8. Frequency of use and content of domains within the definition of malnutrition

Figure 9. The CRC-NORDIET study design

Page 7: Malnutrition and dietary interventions in colorectal

VII

LIST OF PAPERS

Paper 1

Henriksen HB*, Ræder H*, Bøhn SK, Paur I, Kværner AS, Billington SÅ, Eriksen MT,

Wiedsvang G, Erlund I, Færden A, Veierød MB, Zucknick M, Smeland S, Blomhoff R. The

Norwegian dietary guidelines and colorectal cancer survival (CRC-NORDIET) study: a food-

based multicenter randomized controlled trial. BMC Cancer. 2017. Jan 30; 17(1):83

*These authors contributed equally

Paper 2

Ræder H, Henriksen C, Bøhn SK, Vilbo AR, Henriksen HB, Kværner AS, Rolid K, Paur I,

Smeland S, Blomhoff R. The ability of PG-SGA to detect low FFM in colorectal cancer patients.

Manuscript.

Paper 3

Ræder H*, Kværner AS*, Henriksen C, Florholmen G, Henriksen HB, Bøhn SK, Paur I, Smeland

S, Blomhoff R. Validity of bioelectrical impedance analysis in estimation of fat-free mass in

colorectal cancer patients. Clin Nutr, 2018. Feb; 37 (1): 292-300

*These authors contributed equally

Page 8: Malnutrition and dietary interventions in colorectal

VIII

ABBREVIATIONS

ACS American Cancer Society AICR American Institute for Cancer Research AND Academy of Nutrition and Dietetics ASPEN American Society for Parenteral and Enteral Nutrition BIA Bioelectrical impedance analysis BMI Body mass index CALGB Cancer and Leukemia Group B CBT Cognitive Behaviour Therapy CMS Consensus Molecular Subtypes CPS Cancer Prevention Study CRP C-reactive protein CRC Colorectal cancer CRC-NORDIET The Norwegian dietary guidelines and colorectal cancer survival CT Computerized tomography CVD Cardiovascular diseases DFS Disease-free survival DRM Disease-related malnutrition DXA Dual energy x-ray absorptiometry ESPEN European Society for Clinical Nutrition and Metabolism EWGSOP European Working Group on Sarcopenia in Older People FFM Fat-free mass FFMI Fat-free mass index FM Fat mass GI Gastrointestinal HR Hazard ratio ICD International classification of diseases and related health problems IL Interleukin LBM Lean body mass MD Mediterranean diet MI Motivational interview MNA Mini Nutrition Assessment MRI Magnetic resonance imaging MST Malnutrition Screening Tool MSTC Malnutrition Screening Tool for Cancer Patients MUST Malnutrition Universal Screening Tool NCP Nutrition Care Process NFBDG Norwegian food-based dietary guidelines NF-κβ Nuclear factor kappa-light-chain-enhancer of activated B cells NRS-2002 Nutrition Risk Screening 2002 NSQ Nutritional Screening Questionnaire OS Overall survival PG-SGA Patient-generated subjective global assessment RCD Registered clinical dietitian RCT Randomized controlled trial

Page 9: Malnutrition and dietary interventions in colorectal

IX

RDN Registered dietary nutritionist RENEW Reach out to Enhance Wellness RT Radiotherapy SGA Subjective Global Assessment SMM Skeletal muscle mass SO Sarcopenic obesity TBW Total body water TNFα Tumor necrosis factor alpha TNM Tumor node metastasis WCRF World Cancer Research Fund WHR Waist hip-ratio

Page 10: Malnutrition and dietary interventions in colorectal

10

Page 11: Malnutrition and dietary interventions in colorectal

11

1 INTRODUCTION

1.1 Dietary aspects of cancer

There is strong evidence for the link between lifestyle and the risk of developing cancer [1].

World Cancer Research Fund (WCRF) recently stated that 1/3 of the most common cancers

could be prevented through optimal diet, weight regulation and physical activity [2]. A diet

based on plant foods and a limited intake of salt, high-calorie foods, sugary drinks and

alcohol has shown a protective effect in several common cancers. Based on the extensive

evidence for the role of diet in cancer prevention, comprehensive and detailed dietary

recommendations for cancer prevention are currently available [1].

On the other hand, the role of diet for cancer patients is far less investigated. What is the

optimal diet for cancer patients? Should there be similar dietary recommendations for all

cancer patients, independently of cancer type and disease extent? May diet after diagnosis

have impact on recurrence from the primary cancer, new morbidity including secondary

cancer and survival?

The cancer prevalence continues to grow due to increase in incidence and improved survival.

The first is mainly due to an increase in population and a change in age distribution towards

older patients at higher risk for developing cancer. Despite this strong increase in cancer

prevalence and the known impact of diet on cancer development, few studies have

addressed the impact of diet on survival and the risk of developing a second serious chronic

disease in cancer patients after cancer diagnosis.

The nutritional challenges following a cancer diagnosis vary according to several factors such

as type of cancer, extent of disease, age, presence of comorbidities and the general health

status. Moreover, nutritional problems often change during the course of survivorship. At

the time of diagnosis, cancer patients are particularly vulnerable to unintentional weight loss

and malnutrition due to loss of appetite and decreased food intake caused by the tumor

and/or cancer treatment. After treatment is completed, some patients recover within weeks

or months, whereas others, even patients in complete remission, experience persistent side

effects from cancer treatment and difficulties in regaining weight and lean body mass.

Page 12: Malnutrition and dietary interventions in colorectal

12

Prolonged depletion of lean body mass (LBM) may lead to progressive functional impairment

and decreased quality of life. Since cancer survivors have a life-long increased risk of

developing second primary cancers [3], as well as increased risk of chronic diseases such as

cardiovascular disease, obesity, chronic pulmonary disease and diabetes [4], they may

benefit from guidance on a healthy diet targeting prevention of these diseases. However,

documentation of the impact of diet is so far inadequate and data from randomized

controlled trials are limited.

1.2 Colorectal cancer

Colorectal cancer (CRC) is cancer with origin in the colon or rectum. Tumors in colon are

most commonly localized in the right-sided colon and sigmoideum (Figure 1). More than 90

% of the colorectal cancers are adenocarcinomas originating from epithelial cells of the

mucosa layer of the intestine wall. Less common types of colorectal carcinomas are

neuroendocrine, squamous cell, adenosquamous, spindle cell and undifferentiated

carcinomas [5].

Figure 1. An anatomic overview of colon and rectum. ©2017, WebMD, LCC. All rights reserved.

Page 13: Malnutrition and dietary interventions in colorectal

13

Colorectal carcinomas develop from dysplastic adenomatous polyps, the most common form

of premalignant precursor lesions, through a multistep process known as the “adenoma-

carcinoma sequence”, where inactivation of a variety of tumor suppressor genes and DNA-

repair genes and activation of oncogenes occurs in a sequential manner [6]. These genetic

alterations may be inherited or acquired, and the colorectal carcinogenesis may take more

than 10 years [7].

1.2.1 Diagnosis and staging

Asymptomatic CRC can be diagnosed by different screening procedures including fetal blood

examination or colonoscopy and a national screening programme for Norway is planned to

start in 2019. Most CRC cases are, however, currently diagnosed after the onset of

symptoms such as rectal bleeding, anemia, abdominal pain, diarrhea, constipation, and

weight loss [8, 9]. The clinical presentation is depending on the localization of the cancer.

Change in bowel habits and abdominal pain due to obstruction are more common in

patients with left-sided tumours than patients with right-sided tumours [9]. Hematochezia is

typical for tumours located in the rectum. Anemia is most frequent in patients with tumours

in coecum and ascendens affecting 75% of these patients [10]. About 15-25 % of the CRC

cases are presented with acute ileus, severe bleedings or bowel perforation [11].

Approximately 20 % have metastases to intraabdominal lymph nodes, peritoneum, liver or

lungs at the time of diagnosis [12].

Staging of the cancer, i.e. grading of the cancer with regard to size and extent, performed at

the time of diagnosis is important to initiate appropriate treatment as well as to determine

prognosis. The Tumor Node Metastasis (TNM) staging system developed by The Union for

International Cancer Control and the American Joint Comitee on Cancer [13] has been used

for more than 50 years and describes the magnitude of the tumor (T), the extent of spread

to lymph nodes (N) and the presence of metastasis (M). Based on the combination of T, N

and M, i.e. the TNM status, an overall disease stage is determined (Table 1). Stage 0

corresponds to carcinoma in situ, stage I and II correspond to localized cancer, stage III

spread to regional lymph nodes and stage IV is metastatic cancer (Figure 2).

Page 14: Malnutrition and dietary interventions in colorectal

14

Table 1. Pathological staging according to the TNM classification system

Stage T N M

0 Tis N0 N0

I T1 N0 M0 T2 N0 M0

IIA T3 N0 M0 IIB T4a N0 M0 IIC T4b N0 M0

IIIA T1-T2 N1/N1c M0 T1 N2a M0

IIIB T3-T4a N1/N1c M0 T2-T3 N2a M0 T1-T2 N2b M0

IIIC T4a N2a M0 T3-T4a N2b M0 T4b N1-N2 M0

IVA Any T Any N M1a

IVB Any T Any N M1b TNM: Tumor Node Metastasis. The table is a modified version of a table included in a free poster by American Joint Committee on Cancer; Colon and Rectum Cancer Staging 7th edition. Disease stage (I-IV) is determined based on the combination of Tumor (T), nodes (N) and metastasis (M). Stage 0 corresponds to carcinoma in situ, stage I, and II corresponds to localized cancer, stage III spread to regional lymph nodes and stage IV is metastatic cancer. M1a and M1b denote metastases to one or more than one organs, respectively.

Page 15: Malnutrition and dietary interventions in colorectal

15

Figure 2. The colorectal cancer stages. Colorectal carcinomas develop from adenomatous polyps to

adenocarcinomas through four stages. In stage 0, carcinoma in situ, the cancer has not grown

beyond mucosa, i.e. the inner layer of the colon/rectum wall. In stage 1, the cancer has grown

through mucosa. It has not spread to nearby lymph nodes or distant sites. In stage 2, the cancer has

grown further into the mucosa layers. It has not yet spread to lymph nodes or distant sites. In stage

3, the tumor has grown through the mucosa into submucosa and spread to lymph nodes. It has not

yet spread to distant sites. In stage 4, the cancer has spread to 1 or more distant organs. Liver and

peritoneum are the most affected organs. ©2017, WebMD, LCC. All rights reserved.

Recently, an international effort based on sharing of large-scale data and coordinated

analytics, has resulted in the definition of four consensus molecular subtypes (CMS) [14, 15].

The four CMS represent the best current description of CRC heterogeneity, and form the

basis for the future clinical stratification and subtype-based interventions.

1.2.2 Incidence, mortality and survival

CRC is the third most common cancer in the world, with approximately 1.4 million new cases

in 2012 [16]. Norway is one of the top ten countries with highest incidence in the world [16]

and according to the annual report from the Cancer Registry of Norway, 4268 new cases of

CRC were reported in 2015 [17]. About 80 % of these cases are stage I-III whereas about

20 % are metastatic disease.

Page 16: Malnutrition and dietary interventions in colorectal

16

There has been a significant increase in incidence (rates per 100 000) of CRC for both

genders from 1965 to today (Figure 3). For the last decade there has been a slight, but

steady increase in incidence of colon cancer, whereas the rates for rectum cancer have

stabilized.

Figure 3. Age-standardized incidence, mortality and 5-year survival curves for CRC. A) Colon cancer (ICD-10 C18) and B) Rectum and rectosigmoid cancer (ICD-10 C19-20). Tables from the report Cancer in Norway 2015 [17].

A

A

B

Page 17: Malnutrition and dietary interventions in colorectal

17

The main reason for the increase in incidence is the aging population, as the disease is

strongly associated with increased age. Median age at diagnosis for colon and rectum cancer

is 73 and 69 years, respectively. According to the 2016 Cancer in Norway report [18], more

than 2/3 of the patients operated for localized colon cancer were 75 years old or older.

Figures 4 A and 4 B show the age distribution in patients treated for localized colon and

rectum cancer, respectively. The mortality rates for colon cancer have been stabile from the

mid 1960ies in contrast to a significant mortality reduction for rectum cancer the last 25

years (Figure 3).

Figure 4 A) Gender and age distribution in patients treated by surgery for primary colon cancer

stage I-III. Printed with permission from The Norwegian Colorectal Cancer Registry report (2015)

[18]. B) Gender and age distribution in patients treated by surgery for primary rectum cancer stage

I-III. Printed with permission from The Norwegian Colorectal Cancer Registry report (2015) [18].

A

A

B

Page 18: Malnutrition and dietary interventions in colorectal

18

Similar to other high-income countries, there has been a significant improvement in survival

from CRC in Norway. Since the late 1970s, 5-year relative survival has increased from 40 to

60 % and from 37 to 66 % for colon and rectum cancer, respectively. The improvement in

survival from colon cancer is explained by earlier diagnosis and the introduction of adjuvant

chemoterapy for stage III patients in the late 1990s [19]. For rectum cancer, the

implementation of total mesorectal excision in 1993, advancements in radiology

(introduction of magnetic resonance imaging (MRI)) and increased use of chemotherapy and

radiotherapy have contributed to the increase in survival rates.

Survival is however highly dependent on the TNM stage at diagnosis. In Norway during 2010-

2014, the estimated 5-year relative survival in localized CRC was 77-78 % compared to 12-18

% for metastatic CRC [20].

1.2.3 Etiology

Both genetic and environmental factors play roles in the etiology of CRC, however, only 3-5

% of the CRC cases are attributed to hereditary syndromes, where Familial adenomatous

polyposis and Hereditary non-polyposis colorectal cancer (Lynch syndrome) are the two

major ones [7]. Family history of CRC, i.e. having one or more first-degree relatives with CRC

is present in about 25 % of the patients [21].

A majority of the CRC cases are attributed to unhealthy lifestyle. Smoking, overweight,

abdominal fat distribution, diabetes type 2, physical inactivity and an unhealthy diet are

established risk factors [1, 22]. According to the last update from WCRF and American

Institute for Cancer Research (AICR), i.e. the colorectal cancer continuous update report

released in 2017, there is strong evidence for the protective effects of whole-grains, foods

containing dietary fiber and physical activity, and moreover, for the increased risk by

consuming red and processed meat and alcoholic drinks [23]. It is concluded by the

WCRF/AICR that about 45 % of CRC cases could be prevented by improved lifestyle [1].

Page 19: Malnutrition and dietary interventions in colorectal

19

1.2.4 Treatment of localized CRC

The major treatment modalities for CRC are surgery and chemotherapy and for rectal cancer

also radiotherapy. The majority of the CRC patients undergo surgery, which is the only

curative treatment. In Norway, treatment and follow-up of CRC is practiced in accordance

with the standardized national guidelines from the Norwegian directorate for health [12].

For colon cancer, surgery includes total removal of the tumor-bearing segment and the

corresponding lymph nodes. In most patients undergoing uncomplicated colectomy it is

possible to reestablish intestinal continuity by the use of primary anastomosis [12]. In some

cases, a temporary colostomy or ileostomy is necessary. The standard surgical procedure for

curative resection of rectum cancer is total mesorectal excision, a technique that removes

the tumor-bearing part of the rectum with the surrounding mesorectal fat, mesorectal

lymph nodes and its border, the mesorectal fascia. According to the national guidelines, pre-

operative radiochemotherapy is standard of care for patients with locally advanced rectum

cancer [12]. For rectum cancer, patients aged < 75 years with stage III colon cancer are

offered adjuvant chemotherapy whilst adjuvant chemotherapy is not standard of care for

patients with rectal cancer.

1.2.5 Follow-up

After surgery, patients with localised CRC are systematically followed until 5 years with

regard to local recurrence (i.e. reocurrence of cancer in the same site as the primary tumor)

or distant metastases (i.e. spread of the disease to other organs, including distant lymph

nodes) [12]. According to The Norwegian Colorectal Cancer Registry, 16 % of the patients

with localised colon cancer disease-free after surgery, develop distant metastases during the

first 5 years. Liver and peritoneum are the most commonly affected organs for colon cancer

metastases. Twenty-two % of the patients with localized rectum cancer develop metastases

[18]. Metastases are most commonly localized to the liver or lungs. Estimated proportion

affected by local recurrence is 5 % and less than before introduction of the total mesorectal

excision procedure.

Page 20: Malnutrition and dietary interventions in colorectal

20

1.3 Colorectal cancer-associated malnutrition and

sarcopenia

1.3.1 Definitions and diagnostic criteria of malnutrition

Malnutrition is often observed in CRC patients. There is however currently no universally

accepted definition of malnutrition. ESPEN (The European Society of Clinical Nutrition and

Metabolism) recently published guidelines on definitions and terminology of clinical

nutrition to be used in clinical practice and research [24]. According to these guidelines,

malnutrition is defined as “a state resulting from lack of intake or uptake of nutrition that

leads to altered body composition (decreased fat-free mass (FFM)) and body cell mass

leading to diminished physical and mental function and impaired clinical outcome from

disease” [24]. To diagnose malnutrition, ESPEN has published a set of diagnostic criteria [25]

(Table 2). These criteria are different from the criteria proposed by the Academy of Nutrition

and Dietetics (Academy) and American Society for Parenteral and Enteral Nutrition (ASPEN)

(Table 3) [26]. Whereas low body mass index (BMI) is a central part of the ESPEN criteria, it is

not included in the ASPEN criteria. Moreover, the ASPEN criteria encompass energy intake,

loss of body fat, fluid accumulation and hand grip strength which are not included in the

ESPEN criteria. Weight loss and low muscle mass/FFM are however present in both set of

criteria.

Table 2. The ESPEN diagnostic criteria for malnutrition

1) BMI < 18.5

2) Weight loss (unintentional) > 10 % indefinite of

time, or > 5 % over the last 3 months combined

with either BMI < 20 kg/m 2 if < 70 years of age,

or <22 if > 70 years of age or FFMI < 15 and 17

kg/m2 in women and men, respectively

BMI; body mass index, FFMI; fat-free mass index. Two alternative ways to diagnose malnutrition according the ESPEN Consensus Statement [25]. Prior to diagnosis, it is mandatory to be considered “at risk” by a validated risk screening tool.

Page 21: Malnutrition and dietary interventions in colorectal

21

Table 3. The clinical characteristics for the diagnosis of malnutrition in the Consensus

Statement by the Academy/ASPEN.

Malnutrition in the context of chronic illness

Clinical characteristic

Nonsevere (moderate) malnutrition

Severe malnutrition

Energy intake <75% of estimated energy requirement for ≥1 month

≤75% of estimated energy requirement for ≥1 month

Weight loss 5% in 1 month 7.5% in 3 months 10 % in 6 months 20 % in 1 year

>5% in 1 month >7.5% in 3 months >10% in 6 monts >20% in 1 year

Body fat Mild Severe

Muscle mass Mild Severe

Fluid accumulation

Mild Severe

Reduced grip strength

NA Measurably reduced

NA, not applicable The table is a modified version from the table published in The 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) [26]. A minimum of 2 of the 6 characteristics in the table must be fulfilled for the diagnosis of malnutrition. Chronic is defined as a disease/condition lasting 3 months or longer. Weight loss must be evaluated in light of under or overhydration. Body fat (loss of subcutaneous fat), muscle mass and fluid accumulation is evaluated by a physical exam. Hand grip strength is evaluated in accordance with normative standards presented by the manufacturer of the measurement device.

Page 22: Malnutrition and dietary interventions in colorectal

22

The national guidelines for prevention and treatment of malnutrition [27] have based the

malnutrition codes in the International Classification of Diseases (ICD-10) [28] on low BMI,

weight loss and reduced food intake (Table 4).

Table 4. The ICD-10 classification of malnutrition

Malnutrition code Criteria

E46: Unspecified protein-energy malnutrition NRS 2002 ≥ Score 3

MUST score > 1

MNA score < 11

SGA B

E44: Moderate protein-energy malnutrition Weight loss > 10 % the preceding 3-6 months or > 5 % the last 2 months

BMI < 18.5 kg/m2 (> 70 years old: BMI < 20)

BMI < 20 kg/m2 (>70 years: BMI < 22) and weight loss > 5 % the last 6 months

Food intake <1/2 of calculated requirements the last week

E43: Severe protein-energy malnutrition Weight loss > 15 % the preceding 3-6 months or > 5 % the last month

BMI < 16 kg/m2 (> 70 years old: BMI < 18.5)

BMI < 18.5 kg/m2 (>70 years: BMI < 20) and weight loss > 5 % the last 3 months

Food intake <1/4 of calculated requirements the last week

BMI: Body mass index, MNA; Mini Nutrition Assessment, MUST; Malnutrition Universal Screening Tool, NRS; Nutritional Risk Screening. The World Health Organization ICD-10 classification (version 2016) [28] of malnutrition and the criteria as defined by the National guidelines for prevention and treatment of malnutrition [27].

Page 23: Malnutrition and dietary interventions in colorectal

23

According to the definitions and terminology by ESPEN, malnutrition can be further

categorized into sub categories according to etiology (Figure 5). Malnutrition may be caused

by disease, i.e. disease-related malnutrition (DRM), or other factors such as hunger or

psychological factors. DRM may occur with or without inflammation. When inflammation is

present production of inflammatory mediators induces anorexia and affects the metabolic

pathways leading to increased tissue break down (muscle mass and/or adipose tissue). DRM

with inflammation is also known as cancer cachexia [24].

Figure 5. Malnutrition sub categories. Printed with permission from ESPEN guidelines on definitions

and terminology of clinical nutrition (2017) [24].

Page 24: Malnutrition and dietary interventions in colorectal

24

1.3.2 Fat-free mass depletion

According to the definition of malnutrition as described in the ESPEN guidelines,

malnutrition is characterized by decreased FFM [24]. Low FFM is according to the ESPEN

consensus diagnostic criteria [25] defined as FFM index (FFMI), i.e. FFM adjusted for height

(FFM (kg)/height(m2)), below 15 kg/m2 and 17 kg/m2, in females and males, respectively.

The two-compartment model simply divides the body into fat mass (FM) and FFM. FFM

consists of all that are not fat; minerals, glycogen, proteins and water (Figure 6). On tissue

and organ level, FFM is comprised of skeletal muscle, bone, organs and total body water. In

the literature, FFM is often used synonymously with LBM. However, this is not correct since

LBM does not include bone mineral compartment. FFM represents the sum of LBM and bone

[29].

Figure 6. The two-compartment model of body composition. The body is simply divided into fat and fat-free mass. Body fat consists of storage fat and essential fat. Storage fat is localized subcutaneously and surrounds the internal organs whereas essential fat is utilized by different organs. Fat-free mass consists of water, protein, minerals and glycogen. The amount of water in the adult human body is in the range 50-65 %. Printed with permission from Glucocorticoid therapy and body composition, Nature Reviews Rheumatology [30].

Page 25: Malnutrition and dietary interventions in colorectal

25

The two-compartment model is shown to contribute with important information regarding

nutritional status, since the amount of FFM reflects the amount of skeletal muscle and

moreover, that changes in FFM mainly reflect changes in skeletal muscle mass, since the

other components of FFM are considered to be stable. Nevertheless, in advanced cancer

changes in FFM may be attributed to changes in organs and tumor as well as depletion of

skeletal muscle. Lieffers and colleges observed increases in liver and tumor mass in CRC

patients with unresectable liver metastases, quantified by the use of computer tomography

(CT) images [31].

Since the cut-offs for FFMI were published in 2015 [25], validation studies have confirmed

the prognostic impact of the malnutrition criteria on clinical outcomes [32] and survival [33].

FFM can be estimated by the use of different modalities, including air displacement

plethysmography, labeled water-isotope dilution techniques, dual energy x-ray

absorptiometry (DXA) and bioelectrical impedance analysis (BIA). DXA and BIA were used in

the current work and will be further discussed in the chapter General discussion.

1.3.3 Sarcopenia and sarcopenic obesity

Sarcopenia and frailty may be considered nutritional related disorders in addition to

malnutrition/undernutrition, overweight, obesity, micro-nutrient abnormalities and re-

feeding syndrome (Figure 7). This conceptual categorization of nutrition disorders is

however schematic and the conditions may in practice be overlapping.

Figure 7. Nutrition disorders and nutrition related conditions. Printed with permission from ESPEN

guidelines on definitions and terminology of clinical nutrition (2017) [24].

Page 26: Malnutrition and dietary interventions in colorectal

26

Sarcopenia is defined as “a syndrome characterized by progressive and generalized loss of

skeletal muscle mass and strength with increased risk of adverse outcomes such as physical

disability, poor quality of life and death” [34]. Frailty is a geriatric syndrome characterized by

unintentional weight loss, exhaustion, weakness, slow walking speed and low physical

activity, associated with increased risk of adverse health outcomes such as falls,

hospitalization, institutionalization and mortality [35]. Sarcopenia and frailty are both

associated with advanced age and share several characteristics, such as weight loss,

weakness and reduced physical performance. Frailty is however also characterized by other

factors related to age such as cognitive status and social status (e.g. loneliness and

depression).

Sarcopenia was originally described by Irwin Rosenberg in 1997 as “age-related loss of

muscle mass” [36]. It is now recognized that sarcopenia is not only restricted to the elderly,

and the syndrome may be caused by secondary factors to age such as physical inactivity,

inadequate dietary intake, malabsorption, advanced organ failure and cancer [34]. The

pathogenesis of sarcopenia is complex and multifactorial, encompassing atrophy of muscle

fibers, increased muscle fat infiltration, increased proteolysis and decreased protein

synthesis [37] . In patients with cancer, the process and onset of sarcopenia may be

accelerated due to impaired dietary intake, malabsorption, physical inactivity and the impact

of inflammation.

During the last 20 years, the concept of sarcopenia has been widely discussed and various

definitions have been proposed. In 2010, experts from the European Union Geriatric

Medicine society, European Society for Clinical Nutrition and Metabolism, the International

Academy of Nutrition and Aging, and the International Association of Gerontology and

Geriatrics – Europe Region, gathered to create a study group, the European Working Group

on Sarcopenia in Older People (EWGSOP) in order to propose diagnostic criteria for use in

both clinical practice and research [34]. The experts agreed that the diagnosis should be

based on the following criteria; 1) low muscle mass 2) low muscle strength 3) Low physical

performance, where criterion 1 and either criterion 2 or 3 must be present to fulfill the

diagnosis. Skeletal muscle depletion in combination with excessive adipose tissue,

recognized as “sarcopenic obesity” (SO), is considered to be particularly detrimental since

Page 27: Malnutrition and dietary interventions in colorectal

27

the health risks from obesity and sarcopenia are combined. Moreover, excessive adipose

tissue may lead to increased fat infiltration in skeletal muscle, a condition termed

myosteatosis, leading to decreased muscle density and impaired physical function [38]. SO is

demonstrated to be a significant predictor for functional status and lower survival in patients

with gastrointestinal cancers [39, 40].

1.3.4 Malnutrition and sarcopenia in CRC patients

The prevalence of malnutrition in CRC patients are reported to be in the range of 23-84 %

[41-47] depending on methodology used to assess nutritional status and disease stage. Most

data on prevalence and outcomes of malnutrition in CRC patients are collected at the time of

diagnosis or during CRC treatment. Prior to treatment, the tumor may cause symptoms such

as constipation, abdominal pain and bleedings, subsequently affecting dietary intake and

absorption of nutrients. It is documented that significant weight loss prior to chemotherapy

is prevalent in more than one third of the patients with CRC [48]. Losing weight prior to

chemotherapy increases the risk of chemotherapy-induced toxicity such as stomatitis,

plantar-palmar syndrome, diarrhea, nausea, neuropathy, fever or fatigue, which

subsequently may lead to dose reductions, delays or discontinuation of chemotherapy, and

consequently the effect of treatment. Several studies have shown the impact of weight loss

and malnutrition on toxicity and mortality in CRC patients receiving adjuvant or palliative

chemotherapy [48-50].

CRC patients on radiotherapy (RT) may experience symptoms such as anorexia, nausea,

diarrhea and vomiting. These symptoms may be persistent and affect long-term outcomes.

In a randomized controlled trial in CRC patients referred to RT, patients not receiving

individualized dietary counseling targeting RT induced symptoms had poorer food intake,

more symptoms, poorer nutritional status and lower scores on quality of life (QoL) than

patients who received intensive counseling during RT at the end of RT [51]. Furthermore, the

long-term (median 6.5 years) follow-up of this study showed that the patients with a poorer

dietary intake, worse nutritional status and lower QoL scores at the end of radiotherapy had

a significantly shorter median survival and increased incidence of late effects of toxicity from

radiotherapy [45].

Page 28: Malnutrition and dietary interventions in colorectal

28

The prevalence of sarcopenia in CRC patients is high. Based on various definitions and cut-

offs used, prevalence of sarcopenia is reported to be in the range 39-71 % [50, 52, 53]. The

majority of the studies have used CT to measure low skeletal muscle mass and defined

sarcopenia by the use of cut-offs for sarcopenia published by Prado and coworkers in 2008

[40], which were derived in a mixed sample of patients with tumors in the gastrointestinal

and respiratory tracts. Studies have reported that sarcopenia in CRC patients is associated

with postoperative complications [39, 52-55], increased chemotherapy toxicity [50, 56] and

higher mortality [39, 57]. The impact of sarcopenia on survival has also been shown in

studies with mixed populations of patients were gastrointestinal (GI) cancers were included

[40, 58].

Taken together, the literature shows that malnutrition and sarcopenia assessed at the time

of diagnosis are associated to adverse short-term outcome measurements and survival in

CRC patients. The prevalence of malnutrition and sarcopenia assessed later in the course of

the disease, as well as the impact on quality of life and survival is however not well

investigated. Persistent nutritional problems may lead to continued loss of body weight and

poor nutritional status. In addition, body weight increase in terms of fat instead of FFM may

lead to increased fat infiltration in skeletal muscle, which may accelerate functional decline.

Moreover, most of the studies investigating associations between malnutrition and

sarcopenia and to clinical outcome measurements and survival in CRC patients have included

high proportions of patients with locally advanced or metastatic disease. Only one study [57]

has investigated the impact of sarcopenia in patients with non-metastatic CRC and this study

showed that sarcopenia was found to be associated with reduced survival in this population.

Due to the limited number of studies, data on prevalence and clinical impact of malnutrition

and sarcopenia in non-metastatic patients is currently scarce.

Page 29: Malnutrition and dietary interventions in colorectal

29

1.4 Nutritional risk screening and assessment tools

in oncology

The primary nutritional evaluation is screening, which aims to systematically identify

individuals who are malnourished or are at risk of malnutrition. The initial screening may be

undertaken by a nurse and allows for a rapid decision of whether the patient should be

further evaluated by a nutritional professional with specialized expertise, for instance a

registered clinical dietitian. An in-depth evaluation should include assessment of symptoms

affecting food intake and an evaluation of food intake, appetite, weight status, muscle mass,

subcutaneous fat stores, fluid status, comorbidities and laboratory status [59].

According to the Oncology Evidence-Based Nutrition Practice Guideline for Adults from the

Academy of Nutrition and Dietetics, “adult oncology patients should be screened using a

malnutrition tool validated in the setting in which the tool is intended for use” [60]. More

specifically, Malnutrition Screening Tool (MST) [61], Malnutrition Screening Tool for Cancer

patients (MSTC) [62] and Malnutrition Universal Screening Tool (MUST) [63] are the tools

recommended for hospitalized patients, whereas MST is recommended for

ambulatory/outpatient settings. For nutritional assessment, the Academy recommends that

“registered dietary nutritionists (RDN) should use an assessment tool validated in the setting

in which the tool is intended for use as part of the complete nutrition assessment”. These

include the Patient-generated subjective global assessment (PG-SGA) [64] and the Subjective

Global Assessment [65].

According to ESPEN guidelines on nutrition in cancer patients [66], the recommendation

concerning screening is as follows; “To detect nutritional disturbances at an early stage, we

recommend to regularly evaluate nutritional intake, weight change and BMI, beginning with

cancer diagnosis and repeated depending on the stability of the situation” [66]. Next, “In

patients with abnormal screening, we recommend objective and qualitative assessment of

nutritional intake, nutritional impact symptoms, muscle mass, physical performance and the

degree of systemic inflammation” [66]. The ESPEN guidelines include various methods for

the purpose of screening, such as BMI, evaluation of weight loss, evaluation of food intake,

or screening tools such as Nutritional Risk Screening 2002 (NRS-2002)[67] , MUST, MST or

Mini Nutritional Assessment Short Form Revised [68]. Regarding nutritional assessment,

Page 30: Malnutrition and dietary interventions in colorectal

30

ESPEN recommends that patients identified as “at risk” should be assessed with attention to

factors such as dietary intake, body composition, physical activity and predominant

metabolic pattern (i.e. metabolic derangements caused by systemic inflammation) without

recommending any specific nutritional assessment tool [66].

In Norway, NRS 2002, MUST, MNA [68] and SGA are the available recommended tools to use

for identification of patients with increased risk of malnutrition [27]. An overview of the

most commonly used screening and assessment tools in oncology is given in Table 5.

Table 5. An overview of screening and assessment tools in oncology

Die

tary

inta

ke

Nu

trit

ion

al im

pac

t sy

mp

tom

s

Ap

pe

tite

We

igh

t lo

ss

BM

I

Bo

dy

com

po

siti

on

An

thro

po

me

try

Ph

ysic

al f

un

ctio

n/m

ob

ility

Psy

cho

logi

cal s

tre

ss

Dis

eas

e

Ne

uro

psy

cho

logi

cal

pro

ble

ms

Me

tab

olic

str

ess

Screening tools

NRS 2002 X X X

MST X X

MUST X X X

MSTC X X X

PG-SGA SF

X X X X

Assessment tools

SGA X X X X X X X

PG-SGA X X X X X X X

MNA X X X X X X X

Mini Nutrition Assessment (MNA); MST: Malnutrition Screening Tool; MSTC: Malnutrition Screening Tool for Cancer Patients; MUST: The Malnutrition Universal Screening Tool; NRS: Nutritional Risk Screening; PG-SGA: Patient-Generated Subjective Global Assessment; PG-SGA SF: Patient-Generated Subjective Global Assessment Short Form; SGA: Subjective Global Assessment Body composition includes a visual examination of muscle and fat depletion Metabolic stress includes questions regarding use of corticosteroids or the presence of fever Neuropsychological problems include depression and dementia

Page 31: Malnutrition and dietary interventions in colorectal

31

The various methods used in nutritional assessment of cancer patients differ with regard to

which nutritional aspects being emphasized. Sealy and colleges investigated to which extent

commonly used methods for nutritional screening and assessment in cancer patients

adequately cover the dimensions of the malnutrition definitions by ESPEN and ASPEN,

respectively [69]. Based on the malnutrition definitions, four dimensions were defined; A)

Assessment of nutrient balance B) Assessment of body weight, body area and body

composition C) Assessment of muscle, immune and cognitive function and D) Measurement

of inflammatory factors. They systematically reviewed the largest databases for studies

including assessment of malnutrition, between 1998 and 2013. The authors discovered 37

different methods used for malnutrition assessment. Only four out of 37 methods covered

all dimensions of malnutrition, namely the Nutritional Screening Questionnaire (NSQ), MNA,

SGA and PG-SGA, of which SGA and PG-SGA were the most frequently used (Figure 8).

Page 32: Malnutrition and dietary interventions in colorectal

32

Figure 8. Frequency of use and content of domains within the definition of malnutrition. BIA, Bio-Impedance Measurement; BMI, body mass index; CHI, Creatine Height Index; GNS, General Nutritional Status; HAS, French National Authority for Health; MNA, Mini Nutritional Assessment; MSTC, Malnutrition Screening Tool for Cancer; MUST, Malnutrition Universal Screening Tool; NRI, Nutritional Risk Index; NSQ, Nutritional Screening Questionnaire; NST, Nottingham Screening Tool; PNI, Prognostic Nutritional Index; PINI, Prognostic Inflammatory and Nutritional Index; PG-SGA, Patient-Generated Subjective Global Assessment; SGA, Subjective Global Assessment; SNAQ, Short Nutritional Assessment Questionnaire. Figure is printed with permission from Content validity across malnutrition assessment in patients with cancer is limited, Journal of Clinical Epidemiology [69].

Page 33: Malnutrition and dietary interventions in colorectal

33

The selection of assessment tool to identify malnutrition has implications for whether cancer

patients are adequately recognized as malnourished and subsequently receive appropriate

nutritional therapy. For instance, it is not given that assessment tools originally designed to

identify malnutrition in cancer patients in the lower range of BMI are suitable to detect

malnutrition in cancer patients with overweight or obesity. The growing prevalence of

overweight and obesity among cancer patients challenges the use of the traditional markers

of malnutrition such as low BMI and weight loss. For instance, recent studies have

demonstrated that substantial loss of muscle mass may be masked in overweight and obese

cancer patients [40, 70], and cancer patients with identical BMI may have significantly

different levels of skeletal muscle [71]. By using an assessment tool designed to capture

changes in body weight without taking the aspect of body composition into consideration,

important information may be missed since the weight change may consist of increases in

fat and not muscle mass [72]. Hence, nutrition assessment tools in oncology practice should

be adequately designed to identify malnutrition in patients with muscle mass depletion,

particularly in overweight and obese cancer patients. Although the nutritional assessment

tools recommended by ESPEN and by the Academy of Nutrition and Dietetics are validated in

many cancer populations, they are not sufficiently validated in cancer populations with these

nutritional concerns.

In the current thesis, PG-SGA was selected as nutritional assessment tool to evaluate its

ability to detect low FFM. BIA and DXA were selected as methods to measure FFM. These

methods are described in more details in the chapter General Discussion.

Page 34: Malnutrition and dietary interventions in colorectal

34

1.5 The role of diet after CRC diagnosis

Malnutrition and sarcopenia are common features of CRC. However, the role of malnutrition

and dietary interventions after CRC diagnosis is not well investigated. Current available data

on the impact of diet on CRC recurrence and survival in this patient population, are based on

two large US cohorts, the CALGB (Cancer and Leukemia Group B) Diet and Lifestyle

Companion Study [73] and the American CPS (Cancer Prevention Study) II Nutrition Cohort

[74].

Meyerhardt and coworkers analyzed dietary patterns in stage III colon cancer patients

treated by surgery and chemotherapy, and found that a western dietary pattern, i.e. a diet

characterized by high intakes of red and processed meat, fat, refined grains and dessert, was

associated with increased risk of cancer recurrence and mortality. In contrast, a prudent

dietary pattern characterized by high intakes of fruits and vegetables, poultry and fish was

associated to favorable outcome [75].

In participants recruited to the American CPS study, intake of red and processed meat after

diagnosis was not associated with CRC-specific or all-cause mortality [76]. However, analyses

from the same study revealed that individuals with high intakes of red and processed meat

both before and after diagnosis had a 79 % increased risk of CRC-specific mortality compared

with individuals with low intakes at both time points. Since the participants to a minor

extent changed their intake of red and processed meat after diagnosis, the study did not

provide data to evaluate the effect of change in dietary habits.

Glycemic load and total intake of carbohydrates related to CRC survival were analyzed from

the CALGB study cohort. Both higher dietary glycemic load and total carbohydrate intake

were associated to increased risk of recurrence and mortality in overweight or obese

patients with colon cancer, but not in patients with BMI < 25 [77]. Moreover, patients who

consumed two or more sugar-sweetened beverages per day after a colon cancer diagnosis

had a 67 % increased risk of recurrence compared to patients who consumed less than two

per month [78].

Taken together, these observational studies suggest that diet after a CRC diagnosis may

affect risk of CRC recurrence and mortality. More data from additional cohort studies and

Page 35: Malnutrition and dietary interventions in colorectal

35

randomized controlled trials (RCTs) are however needed to be able to draw robust

conclusions about the role of diet in CRC survivors. So far, only one RCT has been conducted

in CRC patients to investigate the effect of a dietary intervention on survival. In this study,

they aimed to test the effect of individualized dietary counseling with focus on symptom

alleviation and/or arrest and adequate energy intake [45]. To date, no RCTs have tested the

effect of a food-based dietary intervention on disease outcomes and survival in CRC patients.

The CRC-NORDIET study was therefore designed to test the effect of a diet in accordance

with the Norwegian food-based dietary guidelines (NFBDG) (Table 6). These guidelines were

developed to prevent chronic diseases in the general population and are based on a

comprehensive, systematic review of the evidence linking diet to the risk of chronic diseases

[79].

Table 6. The Norwegian food-based dietary guidelines

Dietary recommendation

1. A primarily plant-based diet is recommended, including plenty of vegetables,

fruit, berries, wholegrain and fish, and limited quantities of red and processed

meat, salt, added sugar and energy-rich foods

2. It is recommended to maintain a balance between energy intake and energy

expenditure

3. Eat at least five portions of vegetables, fruit and berries every day

4. Eat at least four portions of wholegrain products every day

5. Eat the equivalent of two or three portions of fish per week

6. Low-fat dairy products should be included in your daily diet

7. It is recommended to eat lean meat and lean meat products, and limit the

intake of red meat and processed meat

8. It is recommended to use cooking oil, liquid margarine or soft margarine

9. Water is recommended as the primary choice of drinks

10. Limit your intake of added sugar

11. Limit your intake of salt

12. Dietary supplements may be necessary to ensure an adequate intake of

nutrients for some groups in the population

13. It is recommended that everyone participates in least 30 minutes of physical

activity per day

For specific and detailed quantitative recommendations, see the Norwegian food-based dietary guidelines [79].

Page 36: Malnutrition and dietary interventions in colorectal

36

1.5.1 Factors affecting change in dietary habits

In order to be able to measure effects of dietary interventions, long-term compliance to

dietary recommendations is essential. However, achieving long-term changes in dietary

habits is complex and the individual`s ability or willingness to change diet is dependent on

several factors. For instance, knowledge, beliefs and interest in nutrition may influence the

motivation and susceptibility for change. Moreover, socioeconomic factors including

financial conditions, literacy and support from family or spouse, and psychological factors

such as depression and loneliness may also have impact on the motivation to make changes

of dietary habits. In cancer patients, there may be additional barriers to change diet due to

factors related to the disease and treatment, such as fatigue, pain, GI problems and reduced

physical function. Thus, dietary interventions not taking into account these factors may be

less likely to succeed in achieving compliance to the dietary advice.

There are several theories regarding human`s ability and potential to change behavior,

including strategies to achieve these changes. The health belief model, social cognitive

theory, theory of planned behavior and transtheoretical model of change are examples of

theories focusing patient-centered factors such as self-efficacy, perceived control, barriers

and benefits of making changes [59]. In the dietary counseling setting, various approaches

based on these models may be used to enhance behavior change. Counseling targeting

“behavior modification” is based on principles from behaviorism, a psychological theory

founded by Watson [80]. According to this theory, it is essential to understand behavior by

identifying the stimulus that triggers a certain response. For example, a patient eats more

chocolate if chocolate is available in the kitchen. In this case, clinical dietitian according to a

behavioristic approach will recommend the patient to avoid having chocolate in the kitchen

and thus remove the stimulus that triggers the undesirable outcome. Within this approach,

use of positive and negative reinforcements (i.e. feedbacks) are essential strategies to

modify the patient`s behavior. One of the main limitations with this method is that effect of

reinforcements may be temporary and that the desirable behavior terminates when the

feedbacks disappear. In addition, the method has been criticized for leaving the

responsibility of change to the counselor and not the patient and thus ascribing the patient a

passive role in the process of change. Approaches emphasizing patient-driven behavior

change such as cognitive approaches, take into account the relationship between thoughts,

Page 37: Malnutrition and dietary interventions in colorectal

37

emotions and behavior, in order to increase the patients` awareness of how they act in the

way they do, and how their actions may influence the environment [80]. There are several

related strategies with origin in cognitive theories. One of them, cognitive behavior therapy

(CBT), seeks to identify behavior and thoughts leading to negative outcomes and includes

strategies to change the behavior and thoughts. CBT is commonly used in treatment of

several psychological and psychiatric disorders.

Several approaches used to facilitate a behavior change assume that the patient is

motivated for changing behavior. However, lack of motivation may represent a major

obstacle for achieving effects on desirable outcome. The motivational interviewing (MI)

method developed by Miller and Rollnick, is a widely used and well documented [81]

counseling approach where the primary focus is to enhance readiness for change by helping

patients explore and resolve ambivalence. The unique core concept of MI is the patient-

centered counseling with the motivation for change to be expressed by the patient and not

argued by the counselor. The concept of MI was developed to treat alcohol abuse, but is

now extensively used in several other areas for health behaviors. In the current thesis, MI

was implemented in the nutrition intervention as part of the dietary counseling in the CRC-

NORDIET study (paper 1) and will be further discussed in the General discussion.

Nutritional intervention is a central part of the Nutrition Care Process (NCP) model, which is

the standardized process for nutritional care developed by the Academy of Nutrition and

Dietetics (AND) [82]. The nutrition intervention aims to treat the nutritional problem and

may consist of nutrition and food therapy, nutrition education and counseling. Nutrition

education and counseling are both important parts of the nutrition intervention in order to

achieve the treatment goals. Whereas nutritional education aims to help the patient to gain

knowledge and skills needed to make desirable change, dietary counseling focuses on how

to reach the goals, by identifying favorable conditions for change. The dietary intervention in

the CRC-NORDIET study (paper 1) was therefore designed to include both education, i.e.

educating the patients how to change their dietary habits according to the NFBDG, and

dietary counseling with particular focus on individual needs, nutritional status and

challenges with regard to symptoms and food intake as well as motivational status.

Page 38: Malnutrition and dietary interventions in colorectal

38

Page 39: Malnutrition and dietary interventions in colorectal

39

2 AIMS

In Norway, there has been a steady increase in survival of non-metastatic CRC patients the

last decades. CRC patients have more comorbidities and reduced quality of life compared to

the general population. To date, there is limited data on the role of diet after CRC diagnosis,

and intervention studies investigating whether there exists a causal relationship between

diet and disease outcomes and survival in these patients are urgently needed.

After CRC surgery, patients may experience nutritional problems leading to malnutrition,

depletion of FFM and sarcopenia. It is suggested that malnutrition and FFM depletion may

have impact on quality of life, physical function and survival in CRC patients, and hence

appropriate nutritional interventions are needed to deal with these nutritional problems.

PG-SGA is one of a few nutritional assessment tools targeting all dimensions of malnutrition,

including depletion of fat and muscle. Moreover, the tool is recommended by The Oncology

Evidence-Based Nutrition Practice Guideline for Adult from the Academy of Nutrition and

Dietetics to use in cancer patients. However, the knowledge about its ability to identify low

FFM is scarce.

The overall aims of this thesis focusing on non-metastatic CRC patients were to:

1. Develop and establish a RCT with individualized nutrition intervention

2. Evaluate the ability of the nutritional assessment tool PG-SGA to identify low FFM

3. Determine the prevalence of malnutrition, low FFM and sarcopenia

4. Evaluate the ability of two different BIA devices to assess FFM

Page 40: Malnutrition and dietary interventions in colorectal

40

3 SUMMARY OF PAPERS

3.1 Paper 1

This paper presents the study protocol of the Norwegian Dietary Guidelines and Colorectal

Cancer Survival study. Men and women aged 50-80 years diagnosed with primary invasive

CRC (Stage I-III) are invited to this randomized controlled, parallel two-arm trial 2-9 months

after curative surgery. The intervention group (n=250) receives an intensive dietary

intervention lasting for 12 months and a subsequent maintenance intervention for 14 years.

The control group (n=250) receives no dietary intervention other than standard clinical care.

Both groups are offered equal general advice of physical activity. Patients are followed-up at

6 months and 1, 3, 5, 7, 10 and 15 years after baseline. The study center is located at the

Department of Nutrition, University of Oslo, and patients are recruited from two hospitals

within the South-Eastern Norway Regional Health Authority. Primary outcomes are disease-

free survival and overall survival. Secondary outcomes are time to recurrence, cardiovascular

disease-free survival, compliance to the dietary recommendations and the effects of the

intervention on new comorbidities, intermediate biomarkers, nutrition status, physical

activity, physical function and quality of life.

The current study is designed to gain a better understanding of the role of a healthy diet

aimed at dampening inflammation and oxidative stress on long-term disease outcomes and

survival in colorectal cancer patients. Since previous research on the role of diet for

colorectal cancer survivors is limited, the study may be of great importance for this cancer

population.

Page 41: Malnutrition and dietary interventions in colorectal

41

3.2 Paper 2

The aim of this study was to investigate the ability of PG-SGA to detect low FFM in patients

with non-metastatic CRC. Ninety-seven patients were included and categorized as well

nourished (PG-SGA A, n=67) or malnourished (PG-SGA B, n=30). BIA was used to assess FFM.

Low FFM was defined as low FFMI according to cut-off values recently proposed by ESPEN.

Twenty-nine percent of the patients were identified with low FFMI. The proportion with low

FFMI was significantly higher among patients classified as malnourished by PG-SGA

compared to well nourished (p=0.015). The sensitivity was however low, as the PG-SGA

categorization classified only 50.0 % of the patients with low FFMI as malnourished (PG-SGA

B). Moreover, 60.7 % of the patients with low FFMI had a total PG-SGA score indicating need

for a nutritional intervention (i.e. >4 points). Physical examination in the PG-SGA identified

only 64.3 % of the patients with low FFMI as muscle depleted. In conclusion, our results

indicate that the PG-SGA does not identify with sufficient sensitivity patients with low FFMI

among patients with non-metastatic CRC. In clinical practice, PG-SGA should be

accompanied by muscle mass assessments by BIA or other methods in order to detect low

FFM in this patient group.

Page 42: Malnutrition and dietary interventions in colorectal

42

3.3 Paper 3

The aim of this study was to validate a whole-body and a segmental BIA device against DXA

in CRC patients, and to investigate the ability of different empiric equations for BIA to predict

DXA FFM (FFMDXA).

Forty-three non-metastatic CRC patients (aged 50-80 years) were enrolled in this study.

Whole-body and segmental BIA FFM estimates (FFMwhole-bodyBIA, FFMsegmentalBIA) were

calculated using 14 empiric equations, including the equations from the manufacturers,

before comparison to FFMDXA estimates.

Strong linear relationships were observed between FFMBIA and FFMDXA estimates for all

equations (R2=0.94-0.98 for both devices). However, there were large discrepancies in FFM

estimates depending on the equations used with mean differences in the ranges -6.5-6.8 kg

and -11.0-3.4 kg for whole-body and segmental BIA, respectively. For whole-body BIA, 77 %

of BIA derived FFM estimates were significantly different from FFMDXA, whereas for

segmental BIA, 85 % were significantly different. For whole-body BIA, the Schols* equation

gave the highest agreement with FFMDXA with mean difference ± SD of -0.16 ± 1.94 kg

(p=0.582). The manufacturer`s equation gave a small overestimation of FFM with 1.46 ± 2.16

kg (p<0.001) with a tendency towards proportional bias (r=0.28, p=0.066). For segmental

BIA, the Heitmann* equation gave the highest agreement with FFMDXA (0.17 ± 1.83 kg

(p=0.546)). Using the manufacturer’s equation, no difference in FFM estimates was observed

(-0.34 ± 2.06 kg (p=0.292)), however, a clear proportional bias was detected (r=0.69,

p<0.001). Both devices demonstrated acceptable ability to detect low FFM compared to DXA

using the optimal equation.

In a population of non-metastatic CRC patients, mostly consisting of Caucasian adults and

with a wide range of body composition measures, both the whole-body BIA and segmental

BIA device provide FFM estimates that are comparable to FFMDXA on a group level when the

appropriate equations are applied. At the individual level (i.e. in clinical practice) BIA may be

a valuable tool to identify patients with low FFM as part of a malnutrition diagnosis.

Page 43: Malnutrition and dietary interventions in colorectal

43

4 GENERAL DISCUSSION

4.1 Study designs

This thesis includes a study protocol for the RCT “The Norwegian dietary guidelines and

colorectal cancer survival (CRC-NORDIET) study: a food-based multicenter randomized

controlled trial” (paper 1) and two cross-sectional studies (paper 2 and 3) with populations

drawn from the patients included in the CRC-NORDIET study.

The RCT was established due to paucity of data on the role of diet after CRC diagnosis, and

more specifically, lack of intervention studies investigating whether a causal relationship

exists between diet and disease outcomes and survival in colorectal cancer patients. Current

evidence supporting diet after CRC diagnosis is based on epidemiological studies indicating

that diet is of importance for CRC survivors, however, RCTs are needed to confirm that diet

improves survival and disease outcomes in this population. RCTs are considered the most

rigorous method for investigating the cause-and-effect relationship between a dietary

intervention and outcome [83]. The main strength of this study design is the random

allocation to treatment group(s) and control group(s), ensuring that the potential

confounding variables, both known and unknown factors, are equally distributed among

intervention subjects and controls.

The evidence for the impact of diet on disease in cancer prevention is based on

observational and biological studies, since long-term RCTs are not appropriate for this

purpose in a healthy population. Cancer patients are, however, suitable as a target for RCTs,

since these patients are more likely to develop new morbidities and have increased risk of

death, compared to people without a cancer diagnosis. Moreover, RCTs allow for the

opportunity to strongly influence the participants` diet, and thus generate differences

between intervention subjects and control subjects that are sufficiently large to observe

effects of the dietary intervention on the outcomes of interest.

In paper 2 and 3, we used a cross-sectional study design. Cross-sectional studies are often

described as taking a “snapshot” of a group of individuals, since data is collected from one

time point and thus reflects the situation at this particular time point [84]. Cross-sectional

Page 44: Malnutrition and dietary interventions in colorectal

44

studies are particularly useful to estimate prevalence of various conditions or outcomes, and

factors associated with the outcomes. Since measurement of exposure and outcomes are

performed simultaneously, no conclusions about temporal relationship may be drawn from

this type of studies. Moreover, one limitation with these studies is that the outcome of

measurements may vary from one point of time to another, and hence influence the

generalizability of the results. The prevalence of low FFMI and malnutrition (e.g. PG-SGA B)

assessed in paper 2 and 3 could have been different if measured at another point of time. In

paper 2, were we aimed to investigate the ability of PG-SGA to detect low FFMI in patients

with non-metastatic CRC, another prevalence of PG-SGA B and low FFMI would affect the

results of these analyses.

The patients included in paper 3 were CRC patients who had undergone surgery for CRC

within the last 4 years. Hence, the population consisted of patients enrolled in this study

within some months after surgery as well as patients who were enrolled more than 3 years

after surgery. Variability in timing of entry to the study may have implications for the

prevalence of malnutrition and presence of symptoms related to surgery and cancer

treatment. Since these factors may influence the validity of the results, we cannot rule out

the possibility that our results would be different if the measurements were performed in a

CRC patient population with for instance a higher proportion of patients recruited shortly

after surgery.

Page 45: Malnutrition and dietary interventions in colorectal

45

4.2 The Norwegian dietary guidelines and

colorectal cancer survival (CRC-NORDIET) study: a

food-based multicenter randomized controlled trial

Paper 1 presents the study design of The Norwegian dietary guidelines and colorectal cancer

survival (CRC-NORDIET) study. The CRC-NORDIET study is a multicenter RCT with two parallel

study arms (Figure 9). The intervention group receives an intensive dietary intervention and

general advice on physical activity, whereas the control group receives standard dietary

advice and general advice on physical activity. The intervention starts 2-9 months after

surgery (i.e. baseline) and consists of two periods; an intensive period that lasts 12 months,

and a subsequent maintenance period which lasts for additional 14 years. The details of the

intervention are given in the section “Methods and design” of paper 1. Some of the aspects

of the study design are discussed in the next sections.

Figure 9. The CRC-NORDIET study design

Page 46: Malnutrition and dietary interventions in colorectal

46

4.2.1 Study participants

Patients eligible for the CRC-NORDIET study are men and women, 50-80 years, newly

diagnosed with primary CRC, staged I-III according to the TNM system [13]. Precise inclusion

and exclusion criteria are given in paper 1. Patients with stage IV are excluded from the

study due to the short expected survival, as the study aims to test the long-term effects of

diet. However, by including stage I-III we increase the generalizability of our findings since

patients with these stages constitute the majority of the CRC patients.

The two patient populations included in paper 2 and 3, respectively, were recruited from the

ongoing CRC-NORDIET study. Table 7 shows the distribution of sites and stages in the study

populations from paper 2 and paper 3, compared to a reference population, i.e. patients

with stage I-III treated by surgery for primary tumor Norway based on data from the latest

report from the Norwegian Colorectal Cancer Registry. The distribution of colon,

rectosigmoid and rectum cancer in the study populations in paper 2 and 3, respectively, was

similar to the reference population. However, there were also some differences between the

study populations and the reference population. For instance, the proportion of patients

with stage I in paper 2 and the proportion of stage III patients in paper 3 were low compared

to the reference population.

Page 47: Malnutrition and dietary interventions in colorectal

47

Table 7. Study populations in the CRC-NORDIET study vs CRC reference population in Norway

Study population, paper 2 (n=97)

Study population, paper 3 (n=43)

CRC reference population* (n=2878)

Age,years (median)

67 (CRC) 67 (CRC) 73 (colon) / 69 (rectum)

Women Men

46 (47.4 %) 51(52.6 %)

26 (60.5 %) 17 (39.5 %)

1413 (49.1 %) 1465 (50.9 %)

Colon cancer 54 (58.7 %) 26 (60.5 %) 2014 (70 %)

Rectosigmoid cancer

6 (6.5 %) 2 (4.7 %) 127 (4 %)

Rectum cancer 32 (34.8%) 15 (34.9 %) 737 (26 %)

Stage I 10 (11.2%) 15 (37.5 %) 584 (24.3 %)

Stage II 46 (51.7 %) 18 (45.0%) 937 (38.9 %)

Stage III 33 (37.1 %) 7 (17.5 %) 886 (36.8 %) CRC: Colorectal cancer *Numbers are taken from the latest report from the Norway Colorectal Cancer Registry (2015) [18]. In paper 2, localization (colon/rectosigmoid/rectum cancer) was available for 92 out of 97 patients, while TNM stage was available for 89 out of 97 patients. In paper 3, localization (colon/rectosigmoid/rectum cancer) was available for all patients, while TNM stage was available for 40 out of 43 patients.

Patients included in paper 2, were enrolled between August 2013 and March 2015. A total of

one hundred and six patients with CRC stage I-III were assessed with the PG-SGA tool. Of

these, nine patients were excluded from the analyses due to lack of data needed to

determine FFMI. The patients included in paper 3 were enrolled between December 2015

and February 2016. Forty-five patients completed the DXA scans. Of these, two patients

were excluded from BIA assessments due to pacemakers. Furthermore, two patients were

excluded from the segmental BIA assessments due to upper extremity amputations and

technical issues during the assessments, respectively. Thus, 43 completed whole-BIA

assessment and 41 completed segmental BIA assessment.

In contrast to previous validation studies, we chose not to exclude patients with

abnormalities in body shape, obesity, orthopedic prosthesis/implants, chronic diseases and

fluid disturbances (presence of oedema) in order to increase generalizability. Patients with

these features have often been excluded due to possible interference with the BIA

Page 48: Malnutrition and dietary interventions in colorectal

48

measurements, resulting in highly selected patient populations. By including a broad

spectrum of patients, we were able to explore the impact of these features on the

agreement of the estimates.

4.2.2 The control group

The control group in the CRC-NORDIET study does not receive placebo in the sense of

placebo as defined by the English oxford dictionary as “a medicine or procedure described

for the psychological benefit to the patient rather than for any physiological effect”. Placebo-

controlled RCT is most often not possible when studying food-, or exercise-based

interventions, since placebo-foods or placebo-exercise do not exist. We decided that the

control group should receive standard clinical care as well as a booklet with standard dietary

advice for ethical reasons. There is a risk that the control patients will improve their dietary

habits, particularly the most health-conscious participants. We suggest that providing these

dietary advice as a booklet is not sufficient to generate the same effects as the intensive

dietary intervention with all the selected strategies included in this study. Hence, it is not

expected that it will preclude the interpretation of results. Furthermore, we decided to give

the control subjects, identically with the intervention group, free access to the training

studio and general advice physical activity, as well as health reports following every visit at

the study center and invitation to group meetings. We believe that all these aspects are

important in order to increase the motivation to participate in our study as well as to reduce

drop out in the long run.

4.2.3 Blinding

In RCTs, “blinding” refers to the procedure of ensuring that participants, health-care

professionals, data collectors or data analyzers are not informed about which intervention is

received [85]. The purpose with blinding is to reduce the risk that knowledge about the

intervention received affects the outcome measurements. Because of the nature of the

intervention in the CRC-NORDIET study, blinding of participants and health professionals

involved in the assessments and the different activities with the patients (e.g. group

meetings and dietary counseling), is not feasible. However, laboratory personnel and those

who are involved in analysis and interpretation of data are blinded to the group assignment.

Page 49: Malnutrition and dietary interventions in colorectal

49

The effect of blinding or lack of blinding on intervention effects have been investigated [86].

Some studies suggest that lack of double-blinding may lead to overestimation of

intervention effects compared to adequately blinded trials [87-89], however others have not

found the same results [90]. Wood et al found that the bias associated with lack of blinding

were restricted to studies with subjectively assessed outcomes, whereas there was no

evidence for bias in studies with objective outcomes [86]. The primary outcomes in CRC-

NORDIET study are objective outcomes, however, self-reporting questionnaires used in

some of the analyses for the assessment of the secondary endpoints may be a possible

source for performance bias.

4.2.4 The impact of physical activity

Physical activity is one of the established protective factors for CRC and furthermore, several

observational studies suggest that being physical active after a diagnosis of CRC may play an

important role for survival. In a recent meta-analysis of six large prospective cohort studies

by Schmid and coworkers it was found that high vs low physical activity after CRC diagnosis

was associated with a 42 % lower risk of total mortality (Hazard ratio (HR):0.58, 95 % CI,

0.48-0.70) and a 39 % lower risk of CRC-specific mortality (HR: 0.61, 95 % CI 0.40, 0.92) [91].

For the present, no RCTs have been completed to draw any conclusions on the effect of

physical activity on risk for mortality in patients after CRC diagnosis. One ongoing trial, the

CHALLENGE (Colon Health and Life-Long Exercise Change) trial is designed to investigate

whether supervised physical activity have impact on survival in patients with high-risk stage

II and III colon cancer [92]. Since physical activity probably is important to improve survival in

patients diagnosed with cancer, we chose to give identical advice on physical activity to both

intervention and control subjects, in order to isolate the effect of diet. Previous RCTs

conducted in CRC survivors [93-95] have studied the effect of diet in combination with

physical activity and thus, no conclusions for the role of diet can be made based on these

studies.

Ideally, there should be no skewness with regard to physical activity between the two groups

in our study design. However, in order to control for the confounding effects of physical

activity, we have included several methods to monitor physical activity.

Page 50: Malnutrition and dietary interventions in colorectal

50

4.2.5 Long-term follow-up

When conducting an RCT to study the effect of diet on disease outcomes, it is difficult to

estimate the time between dietary exposure and expected change in incidence of disease.

Hence, RCTs should ideally be of a long duration. In our study, patients are followed until 15

years after baseline (see power calculations at 5, 10 and 15 years in paper 1). Hence, we can

expect that the outcomes of interest (i.e. local recurrence, metastasis, new cancers, new

morbidity or death) may occur within the time frame of follow-up. Based on data from the

Norwegian cancer registry for the period 2013-2015, the 5-year relative survival for CRC

patients stage I-III was reported to be 84 % and 85 % and for colon and rectum cancer

respectively, meaning that 16 % and 15 %, respectively, died during the first 5 years after

surgery [18]. Data from the same period showed that metastasis occurred in 16 % and 22 %

of the patients with colon and rectum cancer, respectively and local recurrence occurred in

5% of the patients with rectum cancer within the first 5 years.

4.2.6 The dietary intervention

The recommended diet in the CRC-NORDIET study is in accordance with the NFBDG (Table

6). These guidelines were developed to prevent diet-related chronic diseases in the general

population [79] and are based on a comprehensive, systematic review of the evidence

linking diet to the risk of chronic diseases. Dampening of chronic low-grade inflammation

and oxidative stress are likely mechanisms mediating these effects. The rationale for testing

NFBDG in a CRC population is based on the hypothesis that a diet in accordance with these

guidelines will have beneficial effects in CRC progression and diet-related comorbidities

mediated through dampening of inflammation and oxidative stress. Chronic inflammation

plays an important role in CRC-related comorbidities such as cardiovascular diseases,

metabolic syndrome, obesity, type 2 diabetes and sarcopenia [96]. Moreover, experimental

and clinical research demonstrates that inflammation profoundly affects all phases of

cancer, from the initiation of cancer to early growth, progression and the process of

metastases. Inflammation favors the processes leading to cancer and create a tissue

microenvironment that allow the tumor to grow and metastasize [97]. Hence, we

hypothesize that adherence to our dietary recommendations, will modulate the

Page 51: Malnutrition and dietary interventions in colorectal

51

inflammatory processes and thereby reduce cancer recurrence and development of new

chronic diseases.

Within the NFBDG, we emphasize specific anti-inflammatory and antioxidant-rich foods and

drinks (details given in paper 1), previously identified by our research group through clinical

trials and in vivo and in vitro models [96, 98-104]. The potential of diet to reduce

inflammation and oxidative stress has also been investigated by others. In the PREDIMED

study, adherence to the Mediterranean diet (MD) (i.e a diet rich in vegetables, fruits, whole

grains, legumes, olive oil and fish, and limited intake of saturated fat and red meat), which is

similar to the NFBDG, was demonstrated to increase the total antioxidant capacity [105]. The

MD is furthermore shown to reduce the expression of genes related to inflammation (e.g.

NF-kB, monocyte chemoattractant protein 1, TNF-α, and IL-6) in older people as

demonstrated by Camargo et al [106]. High intakes of fruits and vegetables provide high

levels of different polyphenols with both antioxidant and anti-inflammatory properties. For

instance, polyphenols from olive oil are shown to modulate enzymes in the eicosanoid

pathway leading to a reduced production of inflammatory mediators such as metabolites of

arachidonic acid (i.e. prostaglandins and leukotrienes) [107]. In addition, olive oil

polyphenols reduce inflammatory angiogenesis, which is central in the pathogenesis of

cancer and atherosclerosis [108]. With regard to other components with anti-inflammatory

properties, dietary fibre is shown to decrease inflammatory mediators such as CRP, IL-6 and

TNF-α [109-111] whereas high intakes of fatty acids from marine fish and nuts (i.e. omega 3

fatty acids) are demonstrated to balance the pro-inflammatory omega-6 fatty acids, and

hence modulate inflammation and blood coagulation [112]. For review of the impact of MD

on inflammation, see the review paper published by Ostan et al [96].

The CRC-NORDIET study tests the effect of a whole-diet based on the assumption that a

whole diet approach provides more beneficial effects than single nutrient interventions. It is

previously shown in our research group that various plant compounds have the ability of

creating synergistic effects in in cell experiments using human cancer cells, through

modulation of NF-kB [101]. The hypothesis that a healthy dietary pattern may be beneficial

for cancer patients is supported by epidemiological studies showing that a prudent dietary

pattern (e.g. high intakes of vegetables, legumes, fruits, fish and low intakes of red and

Page 52: Malnutrition and dietary interventions in colorectal

52

processed meat) after cancer diagnosis is associated with reduced risk of recurrence and

mortality.

4.2.7 Implementation of dietary intervention

The dietary intervention in the CRC-NORDIET study is designed to attain long-term

compliance to the NFBDG. One of the major challenges in lifestyle intervention studies is to

achieve sustainable changes in their daily life habits, including diet. In cancer patients, there

may be additional barriers to change diet due to different conditions and consequences

related to the disease and treatment, such as fatigue, pain, gastrointestinal problems, and

depression. However, studies have reported that cancer patients may be particular

motivated to change diet immediately after diagnosis [4, 113, 114]. We have therefore

designed our intervention to be initiated short time after diagnosis, i.e. within the time

frame where the patients are most likely to be interested in changing diet. Furthermore, we

have chosen multiple strategies to implement the dietary recommendations. All the

strategies were carefully selected to achieve compliance to the dietary intervention;

individual counseling by registered clinical dietitians (RCDs) (both face-to-face and telephone

based), delivery of free foods, grocery discount cards, group meetings, cooking course,

printed materials and access to a CRC-NORDIET web page. To our knowledge, no previous

intervention studies conducted in CRC patients have included so many strategies to increase

compliance to the dietary advice. Some of these strategies will be discussed in the next

section.

4.2.8 Individualized nutritional counseling by a registered clinical

dietitian

The nutritional face-to-face counseling aims to meet the individual needs as well as to

educate the patients on how to change their diet in accordance with the NFBDG.

Individualized dietary advice is given based on comprehensive evaluation of dietary intake,

nutritional status assessed by PG-SGA and weight history as well as characterization of

individual needs and problems (e.g. loss of appetite, gastrointestinal symptoms and fatigue).

Furthermore, dietary advice is individualized by taking into account the patient`s personal

eating habits and preferences.

Page 53: Malnutrition and dietary interventions in colorectal

53

Interventions with individualized dietary counseling are previously reported to be effective

in achieving adherence to dietary advice in cancer patients [51, 115-117]. In a RCT by

Ravasco and coworkers, CRC patients receiving dietary counseling during radiotherapy

weekly for 6 weeks significantly increased their energy intake compared to patients with no

dietary counseling [51]. Similar to our intervention, dietary advice was individualized and

based on a thorough evaluation of clinical and nutritional status, adjusted to personal eating

patterns and treatment-related symptoms. Evidence for the effect of individualized dietary

counseling on dietary intake is also supported by RCTs conducted in patients with head and

neck cancer [115, 117] and lung cancer [118, 119].

4.2.9 Telephone and web-based approach

Patients in the intervention group receive face-to-face dietary counseling at the study center

three times the first year, and five times during the maintenance period (year 2-15). In

addition, the patients receive telephone-based counseling in between the meetings at the

study center, consisting of three phone calls during the first year, and then every second

year during the maintenance period (year 2-15). The primary purpose with this follow-up is

to monitor the patients` body weight status, dietary pattern and motivational status. Good

et al recently published a review evaluating the efficacy of telephone delivered interventions

in cancer survivors [120]. Most of the 27 included RCTs in this review aimed to achieve

changes in physical activity, or a combination of physical activity and diet, whereas five

targeted weight control. Approximately 75 % of the reported trials were rated as successful

(i.e. at least one significant end-of-intervention effect was observed). Evidence supporting

the telephone-based approach is also published by others [121].

Two RCT`s performed in CRC patients which at least partially, succeeded in achieving

changes in dietary behavior are the CanChange trial [122] and the Reach out to Enhance

Wellness (RENEW) trial [95] in which both were based on interventions delivered by

telephone. In the CanChange trial, 410 CRC survivors were randomized to either health

coaching focusing at diet, physical activity, weight control, alcohol and smoking, or usual

care. The intervention was delivered as telephone sessions biweekly over 6 months. At 12

months, significant intervention effects were observed for moderate physical activity, BMI,

energy from total fat and energy from saturated fat. No significant effects were observed for

Page 54: Malnutrition and dietary interventions in colorectal

54

fruits, fiber, alcohol or smoking. In the RENEW study [95] including 641 older overweight or

obese survivors of breast, colorectal and prostate cancer, the 12 months home-based

intervention consisted of 15 telephone sessions and 8 automated prompts during 12

months. Significant intervention effects were found for almost all the targeted behaviors;

intake of fruits and vegetables, saturated fat, weight loss and physical exercise.

Time and resources to travel are reported barriers for participation in clinical trials [123-

125], and the telephone-based approach is hence an easily accessible way to participate in a

trial. In addition, the patients may communicate with the RDNs by e-mail and they have

access to a dynamic website. Internet-based approaches are effective channels to obtain

frequent contact with the participants, and potentially to maintain behavior change.

However, evidence supporting newer modalities such as web sites, mail, SMS, mobile/smart

phone applications and blogs is currently limited [120] in cancer patients. When choosing

effective strategies to communicate with the patients it is important to bear in mind the age

group in the study population. Since the patient population in our study is in the range 50-80

years, we chose a combination of up to date modalities, such as mail, SMS and web site and

more “old-fashion” modalities such as printed materials and telephone-based counseling. All

information provided by web/mail/SMS is also given as printed materials to ensure that all

patients in the intervention group have access to the same information.

4.2.10 Motivational interviewing

In the CRC-NORDIET study, the RCDs use principles from motivational interviewing (MI) to

explore the potential to increase motivation. The degree of motivation (i.e. “very

motivated”, “motivated”, “less motivated” and “not motivated”) is taken into account in

each of the counseling sessions. Since the intervention in the CRC-NORDIET study lasts for 15

years, we have to expect that motivation may change during participation and it is likely that

the participants will face challenges to maintain a healthy diet. We suggest that it is more

strategic to focus on a few realistic goals instead of aiming at changing the whole diet. Each

counseling session is therefore intended to result in a few dietary goals, and it is emphasized

that the patient defines his or her own goals. Both the previously mentioned CanChange

study and the RENEW study and several others [114] have based their interventions on

theoretical frameworks and theories, such as the social cognitive theory [126] and MI [127]

Page 55: Malnutrition and dietary interventions in colorectal

55

in order to facilitate changes in lifestyle habits. Lifestyle trials anchoring their interventions

into solid theories for behavioral change are reported to be more likely to achieve success

[114].

4.2.11 Long-term adherence to the dietary intervention

Since most of the RCTs conducted in CRC patients are designed to investigate effects on

short-term outcomes such as quality of life and physical function, studies investigating long-

term adherence to dietary recommendations are limited in these patients. The earlier

mentioned study by Ravasco and coworkers demonstrated that CRC patients receiving

individualized dietary intervention during radiotherapy maintained their dietary intake in

accordance to the recommendations more than 5 years after completion of radiotherapy

[45]. The 6-week long nutritional intervention in this trial was delivered as weekly counseling

sessions with registered dietitians and aimed to educate patients how to modify their

nutritional intake to ensure sufficient energy and nutrient intake, as well as to select

appropriate foods to target symptoms from radiotherapy. Our CRC-NORDIET study is

designed to provide new knowledge about the long-term effects on adherence to the dietary

recommendations. To increase the chances to obtain sustainable effects on dietary

behavior, we have chosen to extend the intervention with a maintenance period of 14

additional years after the first intensive period of 12 months. During this maintenance

period, intervention subjects receive individual counseling (face-to-face and telephone-

based) once a year, invitation to several inspiration meetings and access to the CRC-

NORDIET web site throughout the entire study period. Moreover, patients receive reports

with feedbacks from the non-biological assessments (e.g. body weight, body composition

analysis, blood pressure) after the assessments at study center and this may serve as

additional inspiration for participation and retention in the study. Compliance to the

intervention is fundamental in order to assess any effects of an intervention. We have

therefore developed a specific questionnaire for assessment of compliance to the

intervention, and this questionnaire will be validated within the first period of the study.

Compliance to the intervention will also be measured by objective measures such as

biological markers of food intake (e.g. carotenoids, fatty acids) which will be assessed at

regular time points during the study period.

Page 56: Malnutrition and dietary interventions in colorectal

56

4.2.12 Endpoints and power calculations in the CRC-NORDIET

study

The primary endpoints in the CRC-NORDIET study are

1. Disease-free survival (DFS) (events defined as detection of local recurrence or

metastasis or any second primary other cancer or death from any cause)

2. Overall survival (OS) (event defined as death from any cause)

DFS was defined in agreement with the proposed guidelines by Punt el al [128]. The primary

endpoints will be assessed 5, 10, and 15 years, respectively, of follow up after baseline.

The 16 secondary endpoints in the study are presented in paper 1. Time to recurrence and

all endpoints including survival will be assessed 5, 10, and 15 years of follow up after

baseline. The remaining secondary endpoints will be assessed after 6 months, 1 year and 3

years after baseline.

The sample size was calculated with a logrank test, based on a statistical power of 80 % and

a significance level of 5 %. Five-year OS data from the Norwegian cancer registry report from

2013 [20], 68%, was used as the expected OS for the control group in the study. A sample

size of 250 participants in each study group was needed to detect a 25% reduction in

mortality in the intervention group, with a statistical power of 80 % (corresponding to a

hazard ratio of 0.71). To achieve a 25 % reduction in events of DFS 5 years after baseline,

190 patients were needed in each study group corresponding to 80 % power, to detect a HR

of 0.70 (Paper 1). An expected reduction in mortality of 25 % was based on observational

studies comparing high vs low adherence to a healthy diet and effects on survival from

cancer [20, 76, 129-132], due to limited number of RCTs in this field.

Page 57: Malnutrition and dietary interventions in colorectal

57

4.2.13 Status of the ongoing CRC-NORDIET study

Recruitment of patients to the CRC-NORDIET study was initiated in 2012. Table 8 shows the

number of patients enrolled and as well as the number of patients lost from the study due to

voluntary quit, exclusion or death. At the end of January 2018, 340 patients were enrolled,

and out of these, 324 patients are still participating in the study. Of the 324 patients who are

still in the study, 175 and 165 are intervention patients and controls, respectively.

It is estimated that calculated sample size of 500 participants will be reached by the end of

2020. Of the 340 patients enrolled in the study so far, only 16 patients (4.7 %) have

voluntary quitted. Based on these observations we suggest that study participation is

conceived as feasible by the patients.

Table 8. Status of participation in the CRC-NORDIET study

N

Enrolled in the study A B

340 175 165

Voluntary quitted A B

16 6 10

Excluded* A B

11 4 7

Dead A B

9 7 2

Total lost A B

36 17 19

Status of study participation was analyzed the 31 th of January 2018. *See description of inclusion and exclusion criteria in paper 1. A: Intervention group B: Control group

Page 58: Malnutrition and dietary interventions in colorectal

58

4.3 Nutritional assessment by PG-SGA

PG-SGA was originally developed to evaluate nutritional risk or presence of malnutrition in

an oncologic setting [133]. It was modified from the original SGA [65] with additional cancer-

related nutritional impact symptoms, assessment of recent weight loss (i.e. weight loss the

recent two weeks) reflecting anabolic or catabolic state and a change from clinician-

generated to patient-generated approach (i.e. patients completing the first part of the tool

instead of the clinician). See paper 2 for details. The original PG-SGA was developed to

categorize the patient as PG-SGA A; no nutritional risk/well nourished, PG-SGA B; moderate

risk of malnutrition/malnourished, or PG-SGA C; severe risk of malnutrition/severe

malnourished. The scored PG-SGA was later introduced to facilitate monitoring of subtle

changes in nutritional status as well as effects of nutritional interventions, to be utilized both

in the clinical and research settings. More recently, several studies have evaluated the

shorted version of PG-SGA (i.e. the PG-SGA Short Form, the patient-generated part of the

tool) that is meant to be used as a screening tool. Hence, the PG-SGA is considered to be a

“4-in-1 instrument” to be used both to the purpose of screening, assessment, monitoring

and triaging for interventions [134].

PG-SGA is now widely used, and the full version is recommended by the Academy of

Nutrition and Dietetics as one of the nutritional assessment tools to use in clinical oncology.

Since the patients may complete the patient-centered part of the tool prior to consultation

with the clinical dietitian, valuable time may be saved. The complete assessment takes only

approximately 5 minutes and gives a detailed overview of the patient`s nutritional status

immediately. The PG-SGA scoring is useful in the follow-up of these patients, by using the

scores to monitor changes during and after nutritional therapy. Hence, in the CRC-NORDIET

study, changes in PG-SGA scores will be analyzed and compared between the intervention

group and control group, to investigate the effect of the intervention on nutritional status.

Importantly, PG-SGA has been found to predict clinical outcomes such as length of hospital

stay [135, 136], rehospitalization [137], postoperative complications [138], quality of life

[139] and survival [136, 140-143].

Page 59: Malnutrition and dietary interventions in colorectal

59

In the recent publication by Sealy and colleges, it was demonstrated that PG-SGA is one of

few assessment tools in oncology that include all domains within the malnutrition definitions

by ESPEN and ASPEN [69]. One of these domains is assessment of body weight, body area

and body composition. The physical examination in the PG-SGA consists of visual inspection

and palpation of muscles, subcutaneous fat and edema and seeks to evaluate nutritional

depletion. To which degree this examination is able to detect low muscle mass/FFM in

comparison to a reference method is however not well investigated.

To the best of our knowledge, no studies have investigated the ability of the PG-SGA tool to

detect nutritional depletion in patients with non-metastatic CRC. Hence, the primary aim of

paper 2 was to examine the ability of the PG-SGA to detect low FFM in these patients, with

BIA as reference method for assessment of FFM (see section 4.6). Although PG-SGA scores

previously are shown to be associated nutritional measures such as BMI and weight loss,

nutritional deficits may occur across the categories of BMI, and malnutrition may be present

in patients with absence of weight loss. With the growing prevalence of overweight and

obesity for several cancer sites, it is important to investigate whether the PG-SGA tool is

capable of detecting nutritional depletion also in these patients. Several studies aimed to

explore the concurrent validity of PG-SGA have used SGA or malnutrition ICD-10 codes as

reference or “gold standard”. In these studies PG-SGA score have demonstrated strong

diagnostic accuracy with high sensitivity and specificity [135, 137, 144]. However, neither

SGA nor ICD-10 classification of malnutrition are appropriate as measures of nutritional

deficits. Both methods reflect the same risk factors as evaluated by the PG-SGA such as

reduced food intake and weight loss. Furthermore, the ICD-10 codes for malnutrition

emphasize low BMI, even though nutritional deficits also may be present in individuals with

normal or high BMI.

Page 60: Malnutrition and dietary interventions in colorectal

60

4.4 Assessment of fat-free mass

4.4.1 Methods for assessment of FFM

The field of body composition in medical research has grown rapidly the last decades and

brought valuable insight about the impact of body composition on morbidity and mortality

in cancer patients. To date, body composition methodology encompasses a large number of

methods that differ with regard to precision, costs and availability. Advanced imaging

techniques such as DXA, CT and MRI are considered gold standard methods for assessment

of FFM due to their ability to precisely measure different tissues within FFM, such as skeletal

muscle, fat, bone and organs. However, access to these methods is often limited in clinical

practice and BIA is considered a more available alternative to assess FFM. Since BIA

estimates FFM indirectly, it is less precise compared to the gold standard methods. In the

current work, BIA and DXA were chosen to assess FFM, and certain methodological aspects

regarding these methods will be discussed in the next sections.

Although all are considered gold standard methods, the imaging techniques DXA, CT and

MRI have their strengths and limitations, and it is important to note that they measure

different compartments of the human body. In order to discuss DXA as a reference method

for FFM assessments, it is relevant to compare the method in light of the other gold

standard methods. CT and MRI allow for segmentation and quantification of different tissues

from either cross sectional area in single images or series of images that encompass entire

organs. Within these cross sectional images, the amount of skeletal muscle (including psoas,

paraspinal muscles, transversus abdominus, external and internal obliques and rectus

abdominus) and adipose tissues (including the subcutaneous, visceral and intramuscular

adipose tissues) can be precisely determined. For CT, images from the third lumbar (L3)

vertebra has been defined as the landmark of interest since the composition of skeletal

muscle and adipose tissue at this area is found to be highly representative for whole-body

tissue quantities [33, 34]. The cross-sectional areas (m2) can be computed for each tissue by

the use of specific CT imaging software, and further incorporated into prediction equations

to estimate whole-body skeletal muscle mass and adipose tissue [145-147].However, use of

CT in body composition assessment is limited due to the high radiation exposure, and only

images taken as part of routine care may be utilized for this purpose. Furthermore, the

Page 61: Malnutrition and dietary interventions in colorectal

61

estimation of whole-body tissue quantities is based on regression equations developed in a

selected patient population and may hence not be valid in all populations. In contrast to CT,

MRI is based on for whole-body assessments and repeated measurements than CT.

However, there are also some limitations related to the use of MRI in clinical practice. It is

less available, requires high technical competence and the analysis is time consuming.

4.4.2 DXA

DXA was originally developed to measure bone density and is primarily used to diagnose

osteoporosis. DXA is now also used to characterize body composition as the method also

allows for whole-body and regional determination of FFM, bone mass, and FM. The sum of

lean soft tissue (i.e. FFM without bone) in arms and legs is referred to as total appendicular

skeletal muscle. With its high precision is considered one of the gold standard methods for

measurement of fat and lean mass. In contrast to CT, the radiation exposure is very low, and

DXA is thus more suitable for repeated measurements. The repeatability is found to be very

high [148]. Since we had access to a DXA device in our research setting and considered DXA

as the most suitable imaging method for assessment of fat-free mass, DXA was used as

reference method in the validation study in paper 3. There are, however, some

methodological aspects related to DXA that need to be further discussed. Similar to BIA, also

DXA is based on an assumption that lean soft tissue is constantly hydrated at 73 %. Thus, the

DXA estimates may be less reliable in patients with fluid and electrolyte disturbances. Small

variations in hydration status (i.e. 68-78 %) are however not considered to significantly

influence the estimates [149]. On the other hand, severe overhydration such as ascites and

edema, may have substantial effect on the measurements. In paper 3, only 12 % of the

patients were observed with edema and only four of the patients had a hydration status

exceeding the normal range for the ration between total body water (TBW) measured with

BIA (i.e. the segmental BIA) and FFM measured with DXA (data not shown). Hence, severe

overhydration was not a major concern in our study and had minimal impact on the

interpretation of our results.

Page 62: Malnutrition and dietary interventions in colorectal

62

4.4.3 BIA

Compared to imaging techniques such as CT, MRI and DXA that directly measure lean body

mass with high precision, BIA measures these compartments indirectly by measuring the

impedance of a low-voltage current passing through the body. The impedance consists of

two components, the resistance, which is the opposition of an ionic solution in both intra-

and extracellular spaces and the reactance representing the capacitance from the cell

membranes [37]. The resistance and reactance values are further incorporated in linear

regression equations to calculate TBW or FFM.

These equations have been developed in different populations and combine BIA impedance

data with variables such as height, weight data, age and gender to calculate the various body

compartments. One of the main limitations with BIA is related to the complexity of these

equations. Although a prediction equation is developed and validated in a specific

population it will not necessarily suit other populations. In addition to the equations

incorporated in the various BIA devices, there are a large number of published equations

available in the literature. It is therefore confusing for the clinicians to know which equation

should be used in which patient. Moreover, BIA relies on several assumptions such as normal

body composition and normal fluid and electrolyte status. Since the validity of BIA is mainly

tested in healthy populations where these assumptions are met, the clinical value of BIA in

patient populations has in general been considered limited.

The existing BIA devices are either based on a whole-body approach or a segmental

approach. With the whole-body BIA approach, the human body is viewed as cylindrical

conductor with a uniform cross-sectional area. This model is demonstrated to be valid in

healthy individuals with BMI in the range 16.0-34.0 kg/m2, provided that hydration is normal

and the BIA equation used is applicable to the population studied [150]. In contrast to the

whole-body BIA, the segmental BIA devices measure impedance in the various segments,

such as arms, legs and trunk.

BIA has several advantages compared to DXA, CT and MRI. It is relative cheap, requires

minimal operator training and provides the results immediately. Some BIA devices are

portable and may be used in settings where DXA, CT and MRI is not available, such as in

Page 63: Malnutrition and dietary interventions in colorectal

63

primary care, hospital wards and institutions such as elderly care and nursing homes. BIA

may unquestionable be an easy accessible and useful tool for body composition analysis in

clinical practice, given that the various devices and equations are valid.

4.5 The ability of PG-SGA to identify low FFM

In paper 2, 69 % were evaluated as well nourished (PG-SGA A) and 31 % as malnourished

(PG-SGA B). No patients were classified as severely malnourished (PG-SGA C). The

prevalence of low FFM and sarcopenia in this population was 29 % and 22 %, respectively.

The primary aim of the study was to examine the ability of the PG-SGA to identify low FFM.

Our results showed that despite that almost one third of the patients were found to have

low FFM, PG-SGA did not identify these patients with sufficient sensitivity.

In paper 2, only half of the patients with low FFMI were evaluated as malnourished by the

PG-SGA (i.e. the sensitivity of the PG-SGA categories). When we analyzed the various

components of PG-SGA in patients with low FFMI, we observed that the majority of these

patients were weight stable or gaining weight at the time of assessments, accompanied by a

normal food intake (i.e. stable or increased) and no symptoms affecting food intake.

According to the developers of PG-SGA, the fundamental concept of the PG-SGA assessment

is to consider the patient in terms of anabolic vs catabolic [134] . They recommend the

weight history component of PG-SGA to be used as an indicator of anabolism or catabolism,

and by using recent weight change (weight loss the last month and/or the last 2 weeks),

improvements or deterioration may be detected. However, since increase in body weight

may consist of FM rather than FFM, monitoring only body weight will not precisely reveal

the specific shifts between lean tissues and adipose tissues.

In both study populations (i.e. paper 2 and 3, respectively), we observed a high prevalence of

overweight and obesity. In paper 2, fifty-nine percent of the patients had BMI above the

normal range, whereas only 8 % were underweight. These findings are in line with the

findings by Thoresen in Norwegian metastatic CRC patients [47] and by several others [39,

41, 42, 46, 151]. Moreover, thirty-nine percent of the women and 41 % of the men in paper

3 were classified as abdominally obese (waist circumference ≥88 cm for women and ≥102 cm

Page 64: Malnutrition and dietary interventions in colorectal

64

for men). These findings were not unexpected, since body fatness and abdominal fatness are

established risk factors for CRC.

In paper 2 we demonstrated that 29 % of the patients were identified with low FFMI

assessed with BIA. About 32 % of these patients had BMI in the overweight range.

Interestingly, we observed that PG-SGA physical examination (i.e. visual inspection and

palpation of muscles in temporal areas, deltoids and quadriceps) detected only 64 % of the

patients with low FFMI (as assessed with BIA), implying that 36 % of the patients were not

identified. The physical examination part of the PG-SGA is recognized as challenging, and it is

emphasized that training is necessary both to increase the precision as well as to ensure

reliability [134]. However, it is also acknowledged that muscle mass depletion may be

difficult to detect in the clinical setting. Whereas clinical signs of wasting may be obvious in

patients with BMI in the lower range, it may be masked in patients with normal or high BMI.

In paper 2 we observed that mean BMI was found to be significantly higher in those patients

who were not identified as muscle depleted by the PG-SGA. Furthermore, we observed a

higher proportion of overweight patients among these patients compared to those who

were captured as depleted. These findings indicate that PG-SGA is not sensitive enough to

detect low FFM among patients with non-metastatic CRC, particularly in overweight and

obese patients. To the best of our knowledge, only one study has previously examined the

ability of nutritional risk screening and nutritional assessment tools to detect malnutrition

according to low FFMI. In this study, Elkan and coworkers compared the sensitivity and

specificity of BMI, SGA, MNA, MUST and NRS-2002 in patients with rheumatoid arthritis

[152]. In line with our findings, SGA showed poor sensitivity (46%) to detect low FFM in

these patients.

Although PG-SGA is a recommended tool for nutritional assessments in oncology practice,

there is lack of research concerning the ability of this tool to identify malnourished patients

in terms of low FFM, and our results contribute to new knowledge within this field.

Page 65: Malnutrition and dietary interventions in colorectal

65

4.6 The ability of BIA to assess FFM

In paper 3, we tested the ability of two different BIA devices to assess FFM compared to DXA

as reference method. Main finding from this study were that both devices showed good

agreement compared to DXA, when using the appropriate BIA equation.

In paper 2, a whole-body BIA device was used to assess FFM. Since the whole-body BIA

approach relies on the assumptions on normal body composition and hydration status, the

estimates of FFM could potentially be unreliable in our population consisting of CRC

patients. Hence, we performed a validation study by testing the validity of the same whole-

body BIA device used in paper 2 against DXA as a reference method in a separate population

of CRC patients. The reason why this validation was not performed in the same patient

population as those included in paper 2 was that DXA was not available at that time. Further,

we compared the whole-body BIA device with a segmental BIA device. It has been suggested

that a segmental BIA is better suited than a whole-body BIA in patients with overweight and

obesity since the device measures impedance in the various segments and takes into

account that the trunk only contributes in a minor extent to the whole-body resistance.

However, there is limited data supporting that this approach is superior to the whole-body

approach. The results from this validation study published in paper 3 showed that both

devices, the whole-body and the segmental BIA device, demonstrated good agreement with

DXA, when the appropriate BIA equation was used. Hence, we could not confirm the

superiority of segmental BIA over whole-body BIA in estimation of FFM in this patient

population.

As part of this validation study, we tested a set of BIA equations, both the equations

incorporated in the BIA software of each of the devices (i.e. the manufacturer`s equation) as

well as a selection of equations from the literature. Our results showed that use of the

various equations resulted in significant different estimates of FFM. These results are in line

with the findings reported in a recently published review by Haverkort based on studies

conducted in surgical and oncological patients [153]. Both our study and the review by

Haverkort hence conclude that selection of equation in BIA analysis has significant

implications for the accuracy of estimates. Since the BIA equations are developed in specific

populations based on reference methods, they are specifically suited for these populations.

Page 66: Malnutrition and dietary interventions in colorectal

66

The equations selected in our study were a number of equations mainly developed in

healthy populations and previously tested in cancer patients [154-163]. One of the equations

was the Geneva equation [158] which is recommended by ESPEN. Interestingly, we found

that two of the existing equations, the Schols and the Heitmann equations, applied in the

whole-body and segmental BIA respectively, gave the highest agreement with the estimates

from DXA. The Schols equation was developed in patients with chronic pulmonary disease

[163], while the Heitmann equation was developed in a healthy population consisting of

adult Danes 35-65 years of age [157]. Few others have validated these equations in patient

populations. Mourtzakis compared FFM estimates calculated with Schols equation with DXA

as reference in a mixed population consisting of 51 patients with locally advanced or

metastatic non-small cell lung and colorectal cancer [164]. Similar to our population, the

patient population in this study was dominated by overweight and obese patients. However,

in contrast to our results, they observed large discrepancy between FFM calculated with

Schols equation and and DXA. The Heitmann equation was validated in a study with 26

patients undergoing major abdominal surgery, and similar to our findings, they found good

agreement between BIA and DXA estimates [165].

The manufacturer`s equations in the whole-body and the segmental BIA, respectively, were

not the most suitable equations. Using the manufacturer`s equation in the whole-body BIA

gave a small overestimation of FFM by 1.5 kg with a tendency towards proportional bias.

Hence, by using estimates based on this equation in paper 2, it is possible that the

prevalence of patients with low FFM would be higher if the optimal equation (i.e. the

Schols*) was used. In paper 3 we found that the prevalence of low FFM was 33 % based on

DXA estimates compared to 26 % and 35 % by the use of the manufacturer`s and the Schols

equations, respectively.

On the individual level, FFM estimates were within -2.8-5.7 kg and -4.4-3.7 kg, using the

whole-body and segmental BIA device, respectively, for approximately 95 % of the patients.

Moreover, we observed that both devices resulted in proportional bias. The clinical

implications of these findings are therefore that FFM estimates may be less accurate in

individuals with increased FFM, and that single measurements should be interpreted with

Page 67: Malnutrition and dietary interventions in colorectal

67

care. Repeated measurements in clinical practice, with the same BIA advice and equation

may however, contribute with important insight into changes in FFM, and FM.

Our results showed that despite including patients with abnormal body shapes, obesity,

presence of chronic diseases and orthopedic prosthesis/implants, we observed high

agreement between FFM estimates from both BIA devices and DXA. These findings support

BIA as a good alternative to DXA.

4.7 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].

Page 68: Malnutrition and dietary interventions in colorectal

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.

Page 69: Malnutrition and dietary interventions in colorectal

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.

Page 70: Malnutrition and dietary interventions in colorectal

70

5 CONCLUSIONS

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.

Page 71: Malnutrition and dietary interventions in colorectal

71

6 FUTURE 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.

Page 72: Malnutrition and dietary interventions in colorectal

72

7 REFERENCES

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.

Page 73: Malnutrition and dietary interventions in colorectal

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.

Page 74: Malnutrition and dietary interventions in colorectal

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.

Page 75: Malnutrition and dietary interventions in colorectal

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.

Page 76: Malnutrition and dietary interventions in colorectal

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

expression in prostate cancer cells, xenografts, and xenograft microenvironment. Nutr

Cancer, 2015. 67(2): p. 305-15.

Page 77: Malnutrition and dietary interventions in colorectal

77

103. Paur, I., L.M. Austenaa, and R. Blomhoff, Extracts of dietary plants are efficient

modulators of nuclear factor kappa B. Food Chem Toxicol, 2008. 46(4): p. 1288-97.

104. Paur, I., T.R. Balstad, and R. Blomhoff, Degree of roasting is the main determinant of

the effects of coffee on NF-kappaB and EpRE. Free Radic Biol Med, 2010. 48(9): p.

1218-27.

105. Zamora-Ros, R., et al., Mediterranean diet and non enzymatic antioxidant capacity in

the PREDIMED study: evidence for a mechanism of antioxidant tuning. Nutr Metab

Cardiovasc Dis, 2013. 23(12): p. 1167-74.

106. Camargo, A., et al., Expression of proinflammatory, proatherogenic genes is reduced

by the Mediterranean diet in elderly people. Br J Nutr, 2012. 108(3): p. 500-8.

107. Santangelo, C., et al., Polyphenols, intracellular signalling and inflammation. Ann Ist

Super Sanita, 2007. 43(4): p. 394-405.

108. Scoditti, E., et al., Mediterranean diet polyphenols reduce inflammatory angiogenesis

through MMP-9 and COX-2 inhibition in human vascular endothelial cells: a

potentially protective mechanism in atherosclerotic vascular disease and cancer. Arch

Biochem Biophys, 2012. 527(2): p. 81-9.

109. Chuang, S.C., et al., The intake of grain fibers modulates cytokine levels in blood.

Biomarkers, 2011. 16(6): p. 504-10.

110. Ma, Y., et al., Association between dietary fiber and serum C-reactive protein. Am J

Clin Nutr, 2006. 83(4): p. 760-6.

111. Ma, Y., et al., Association between dietary fiber and markers of systemic inflammation

in the Women's Health Initiative Observational Study. Nutrition, 2008. 24(10): p. 941-

9.

112. Kalogeropoulos, N., et al., Nutritional evaluation and health promoting activities of

nuts and seeds cultivated in Greece. Int J Food Sci Nutr, 2013. 64(6): p. 757-67.

113. Davies, N.J., L. Batehup, and R. Thomas, The role of diet and physical activity in

breast, colorectal, and prostate cancer survivorship: a review of the literature. Br J

Cancer, 2011. 105 Suppl 1: p. S52-73.

114. Stull, V.B., D.C. Snyder, and W. Demark-Wahnefried, Lifestyle interventions in

cancer survivors: designing programs that meet the needs of this vulnerable and

growing population. J Nutr, 2007. 137(1 Suppl): p. 243s-248s.

115. Isenring, E.A., J.D. Bauer, and S. Capra, Nutrition support using the American

Dietetic Association medical nutrition therapy protocol for radiation oncology

patients improves dietary intake compared with standard practice. J Am Diet Assoc,

2007. 107(3): p. 404-12.

116. Isenring, E.A., S. Capra, and J.D. Bauer, Nutrition intervention is beneficial in

oncology outpatients receiving radiotherapy to the gastrointestinal or head and neck

area. Br J Cancer, 2004. 91(3): p. 447-52.

117. Ravasco, P., et al., Impact of nutrition on outcome: a prospective randomized

controlled trial in patients with head and neck cancer undergoing radiotherapy. Head

Neck, 2005. 27(8): p. 659-68.

118. Evans, W.K., et al., A randomized study of oral nutritional support versus ad lib

nutritional intake during chemotherapy for advanced colorectal and non-small-cell

lung cancer. J Clin Oncol, 1987. 5(1): p. 113-24.

119. Ovesen, L., et al., Effect of dietary counseling on food intake, body weight, response

rate, survival, and quality of life in cancer patients undergoing chemotherapy: a

prospective, randomized study. J Clin Oncol, 1993. 11(10): p. 2043-9.

120. Goode, A.D., et al., Telephone, print, and Web-based interventions for physical

activity, diet, and weight control among cancer survivors: a systematic review. J

Cancer Surviv, 2015. 9(4): p. 660-82.

Page 78: Malnutrition and dietary interventions in colorectal

78

121. Eakin, E.G., et al., Telephone interventions for physical activity and dietary behavior

change: a systematic review. Am J Prev Med, 2007. 32(5): p. 419-34.

122. Hawkes, A.L., et al., Effects of a telephone-delivered multiple health behavior change

intervention (CanChange) on health and behavioral outcomes in survivors of

colorectal cancer: a randomized controlled trial. J Clin Oncol, 2013. 31(18): p. 2313-

21.

123. Brawley, O.W., The study of accrual to clinical trials: can we learn from studying

who enters our studies? J Clin Oncol, 2004. 22(11): p. 2039-40.

124. Lara, P.N., Jr., et al., Evaluation of factors affecting awareness of and willingness to

participate in cancer clinical trials. J Clin Oncol, 2005. 23(36): p. 9282-9.

125. Mills, E.J., et al., Barriers to participation in clinical trials of cancer: a meta-analysis

and systematic review of patient-reported factors. Lancet Oncol, 2006. 7(2): p. 141-8.

126. Bandura, A., Human agency in social cognitive theory. Am Psychol, 1989. 44(9): p.

1175-84.

127. Miller, W.R. and S. Rollnick, Motivational interviewing : helping people change. 3rd

ed. ed. Applications of motivational interviewing. 2013, New York: Guilford.

128. Punt, C.J., et al., Endpoints in adjuvant treatment trials: a systematic review of the

literature in colon cancer and proposed definitions for future trials. J Natl Cancer

Inst, 2007. 99(13): p. 998-1003.

129. Hastert, T.A., et al., Adherence to the WCRF/AICR cancer prevention

recommendations and cancer-specific mortality: results from the Vitamins and

Lifestyle (VITAL) Study. Cancer Causes Control, 2014. 25(5): p. 541-52.

130. Kabat, G.C., et al., Adherence to cancer prevention guidelines and cancer incidence,

cancer mortality, and total mortality: a prospective cohort study. Am J Clin Nutr,

2015. 101(3): p. 558-69.

131. Trichopoulou, A., et al., Adherence to a Mediterranean diet and survival in a Greek

population. N Engl J Med, 2003. 348(26): p. 2599-608.

132. Vergnaud, A.C., et al., Adherence to the World Cancer Research Fund/American

Institute for Cancer Research guidelines and risk of death in Europe: results from the

European Prospective Investigation into Nutrition and Cancer cohort study1,4. Am J

Clin Nutr, 2013. 97(5): p. 1107-20.

133. Ottery, F.D., Definition of standardized nutritional assessment and interventional

pathways in oncology. Nutrition, 1996. 12(1 Suppl): p. S15-9.

134. Jager-Wittenaar, H. and F.D. Ottery, Assessing nutritional status in cancer: role of the

Patient-Generated Subjective Global Assessment. Curr Opin Clin Nutr Metab Care,

2017. 20(5): p. 322-329.

135. Bauer, J., S. Capra, and M. Ferguson, 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(8): p. 779-85.

136. Rodrigues, C.S., M.S. Lacerda, and G.V. Chaves, Patient Generated Subjective

Global Assessment as a prognosis tool in women with gynecologic cancer. Nutrition,

2015. 31(11-12): p. 1372-8.

137. Marshall, S., et al., Malnutrition in Geriatric Rehabilitation: Prevalence, Patient

Outcomes, and Criterion Validity of the Scored Patient-Generated Subjective Global

Assessment and the Mini Nutritional Assessment. J Acad Nutr Diet, 2016. 116(5): p.

785-94.

138. Harter, J., S.P. Orlandi, and M.C. Gonzalez, Nutritional and functional factors as

prognostic of surgical cancer patients. Support Care Cancer, 2017. 25(8): p. 2525-

2530.

Page 79: Malnutrition and dietary interventions in colorectal

79

139. Isenring, E., J. Bauer, and S. Capra, 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(2): p. 305-9.

140. Hsieh, M.C., et al., A Prognostic Model Using Inflammation- and Nutrition-Based

Scores in Patients With Metastatic Gastric Adenocarcinoma Treated With

Chemotherapy. Medicine (Baltimore), 2016. 95(17): p. e3504.

141. Kim, H.S., et al., Patient-Generated Subjective Global Assessment as a prognosis tool

in patients with multiple myeloma. Nutrition, 2017. 36: p. 67-71.

142. Persson, C., P.O. Sjoden, and B. Glimelius, The Swedish version of the patient-

generated subjective global assessment of nutritional status: gastrointestinal vs

urological cancers. Clin Nutr, 1999. 18(2): p. 71-7.

143. Vigano, A.L., et al., The abridged patient-generated subjective global assessment is a

useful tool for early detection and characterization of cancer cachexia. J Acad Nutr

Diet, 2014. 114(7): p. 1088-98.

144. Laky, B., et al., Comparison of different nutritional assessments and body-

composition measurements in detecting malnutrition among gynecologic cancer

patients. Am J Clin Nutr, 2008. 87(6): p. 1678-85.

145. Helseundersøkelsen i Nord-Trøndelag (HUNT). Available from:

http://www.ntnu.edu/hunt/databank.

146. Shen, W., et al., Total body skeletal muscle and adipose tissue volumes: estimation

from a single abdominal cross-sectional image. J Appl Physiol (1985), 2004. 97(6): p.

2333-8.

147. Shen, W., et al., Visceral adipose tissue: relations between single-slice areas and total

volume. Am J Clin Nutr, 2004. 80(2): p. 271-8.

148. Baracos, V., et al., Advances in the science and application of body composition

measurement. JPEN J Parenter Enteral Nutr, 2012. 36(1): p. 96-107.

149. Genton, L., et al., Dual-energy X-ray absorptiometry and body composition:

differences between devices and comparison with reference methods. Nutrition, 2002.

18(1): p. 66-70.

150. Kyle, U.G., et al., Bioelectrical impedance analysis-part II: utilization in clinical

practice. Clin Nutr, 2004. 23(6): p. 1430-53.

151. McSorley, S.T., et al., The relationship between tumour stage, systemic inflammation,

body composition and survival in patients with colorectal cancer. Clin Nutr, 2017.

152. Elkan, A.C., et al., Malnutrition in women with rheumatoid arthritis is not revealed by

clinical anthropometrical measurements or nutritional evaluation tools. Eur J Clin

Nutr, 2008. 62(10): p. 1239-47.

153. Haverkort, E.B., et al., Bioelectrical impedance analysis to estimate body composition

in surgical and oncological patients: a systematic review. Eur J Clin Nutr, 2015.

69(1): p. 3-13.

154. Deurenberg, P., et al., Assessment of body composition by bioelectrical impedance in a

population aged greater than 60 y. Am J Clin Nutr, 1990. 51(1): p. 3-6.

155. Deurenberg, P., et al., Sex and age specific prediction formulas for estimating body

composition from bioelectrical impedance: a cross-validation study. Int J Obes, 1991.

15(1): p. 17-25.

156. Gray, D.S., et al., Effect of obesity on bioelectrical impedance. Am J Clin Nutr, 1989.

50(2): p. 255-60.

157. Heitmann, B.L., Prediction of body water and fat in adult Danes from measurement of

electrical impedance. A validation study. Int J Obes, 1990. 14(9): p. 789-802.

158. Kyle, U.G., et al., Single prediction equation for bioelectrical impedance analysis in

adults aged 20--94 years. Nutrition, 2001. 17(3): p. 248-53.

Page 80: Malnutrition and dietary interventions in colorectal

80

159. Kyle, U.G., et al., New bioelectrical impedance formula for patients with respiratory

insufficiency: comparison to dual-energy X-ray absorptiometry. Eur Respir J, 1998.

12(4): p. 960-6.

160. Lohman, T.G., Preliminary results from the 1986 interlaboratory investigation on

bioelectrical impedance. 1988.

161. Lukaski, H.C., et al., Validation of tetrapolar bioelectrical impedance method to

assess human body composition. J Appl Physiol (1985), 1986. 60(4): p. 1327-32.

162. Segal, K.R., et al., Lean body mass estimation by bioelectrical impedance analysis: a

four-site cross-validation study. Am J Clin Nutr, 1988. 47(1): p. 7-14.

163. Steiner, M.C., et al., Bedside methods versus dual energy X-ray absorptiometry for

body composition measurement in COPD. Eur Respir J, 2002. 19(4): p. 626-31.

164. Mourtzakis, M., et al., A practical and precise approach to quantification of body

composition in cancer patients using computed tomography images acquired during

routine care. Appl Physiol Nutr Metab, 2008. 33(5): p. 997-1006.

165. Jensen, M.B., et al., Components of variance when assessing the reproducibility of

body composition measurements using bio-impedance and the Hologic QDR-2000

DXA scanner. Clin Nutr, 1997. 16(2): p. 61-5.

Page 81: Malnutrition and dietary interventions in colorectal

I

Page 82: Malnutrition and dietary interventions in colorectal
Page 83: Malnutrition and dietary interventions in colorectal

STUDY PROTOCOL Open Access

The Norwegian dietary guidelines andcolorectal cancer survival (CRC-NORDIET)study: a food-based multicentrerandomized controlled trialHege Berg Henriksen1†, Hanna Ræder1†, Siv Kjølsrud Bøhn1, Ingvild Paur1, Ane Sørlie Kværner1,Siv Åshild Billington1, Morten Tandberg Eriksen2,3, Gro Wiedsvang2, Iris Erlund4, Arne Færden5,Marit Bragelien Veierød6, Manuela Zucknick6, Sigbjørn Smeland3,7 and Rune Blomhoff1,7*

Abstract

Background: Colorectal cancer survivors are not only at risk for recurrent disease but also at increased risk ofcomorbidities such as other cancers, cardiovascular disease, diabetes, hypertension and functional decline. In thistrial, we aim at investigating whether a diet in accordance with the Norwegian food-based dietary guidelines andfocusing at dampening inflammation and oxidative stress will improve long-term disease outcomes and survival incolorectal cancer patients.

Methods/design: This paper presents the study protocol of the Norwegian Dietary Guidelines and Colorectal CancerSurvival study. Men and women aged 50–80 years diagnosed with primary invasive colorectal cancer (Stage I-III) areinvited to this randomized controlled, parallel two-arm trial 2–9 months after curative surgery. The intervention group(n = 250) receives an intensive dietary intervention lasting for 12 months and a subsequent maintenance interventionfor 14 years. The control group (n = 250) receives no dietary intervention other than standard clinical care. Both groupsare offered equal general advice of physical activity. Patients are followed-up at 6 months and 1, 3, 5, 7, 10 and 15 yearsafter baseline. The study center is located at the Department of Nutrition, University of Oslo, and patients are recruitedfrom two hospitals within the South-Eastern Norway Regional Health Authority. Primary outcomes are disease-freesurvival and overall survival. Secondary outcomes are time to recurrence, cardiovascular disease-free survival,compliance to the dietary recommendations and the effects of the intervention on new comorbidities, intermediatebiomarkers, nutrition status, physical activity, physical function and quality of life.

Discussion: The current study is designed to gain a better understanding of the role of a healthy diet aimedat dampening inflammation and oxidative stress on long-term disease outcomes and survival in colorectalcancer patients. Since previous research on the role of diet for colorectal cancer survivors is limited, the studymay be of great importance for this cancer population.

Trial registration: ClinicalTrials.gov Identifier: NCT01570010.

Keywords: Colorectal cancer, Disease-free survival, Overall survival, Time to recurrence, Cardiovascular disease-free survival, Comorbidity, Inflammation, Oxidative stress, Antioxidant-rich foods, Food-based dietary guidelines

* Correspondence: [email protected]†Equal contributors1Department of Nutrition, Institute of Basic Medical Sciences, University ofOslo, Oslo, Norway7Division of Cancer Medicine, Oslo University Hospital, Oslo, NorwayFull list of author information is available at the end of the article

© The Author(s). 2017 Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, andreproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link tothe Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver(http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Henriksen et al. BMC Cancer (2017) 17:83 DOI 10.1186/s12885-017-3072-4

Page 84: Malnutrition and dietary interventions in colorectal

BackgroundThe incidences of colorectal cancer (CRC) are 5–10times higher in Europe, North America and Oceaniathan in countries in Africa, south Asia and CentralAmerica [1], and the incidence in Norway is among thehighest in the world [2]. Established risk factors for CRCare age, family history of CRC, inherited syndromes (Fa-milial adenomatous polyposis, Lynch syndrome) and in-flammatory bowel disease. In addition, severalmodifiable lifestyle-related risk factors are associatedwith CRC. Those include smoking, body fatness, abdom-inal fatness, diabetes, physical inactivity and an un-healthy diet (high consumption of alcohol, red andprocessed meat, and low consumption of foods contain-ing dietary fibre) [3, 4]. World Cancer Research Fund(WCRF)/American Institute for Cancer Research (AICR)estimates that about 45% of all CRC cases could be pre-vented by improved lifestyle [3].About 40% of CRC patients [5] have at least one con-

comitant disease (e.g. hypertension, cardiovascular dis-ease (CVD), diabetes, chronic obstructive pulmonarydisease or other malignancies) at the time of diagnosisand increased risk of developing additional comorbidi-ties after CRC diagnosis [6–10]. These comorbid condi-tions may preclude or reduce effect of treatment, andconsequently reduce disease-specific and total survival[8, 11, 12].While it is well established that an unhealthy diet in-

creases risk of CRC (e.g. see the latest update fromWorld Cancer Research Fund, 2011 [4]) there are fewstudies that have focused on the effect of diet on diseaseoutcomes and survival [13–15]. In paucity of data, healthauthorities in most countries recommend the same dietto CRC survivors (i.e. patients living with a CRC diagno-sis, including those who have recovered) as to peoplewithout a cancer diagnosis [3].Inflammation and oxidative stress are central under-

lying disease mechanisms in cancer and several otherchronic diseases. Recent research suggests that there aretwo major molecular pathways leading to CRC, both ofwhich involve inflammation and oxidative stress asmajor driving forces. The majority of CRC cases may bedue to molecular events that result in chromosomal in-stability, while about 20-30% of CRCs are due to genehypermethylation (called CpG island methylator ph-enotype (CIMP)) [16–18]. A large proportion of theCRC cases due to CIMP display microsatellite instability[18, 19]. In total, about 70 mutations in different geneshave been identified as relevant for these two pathwaysto CRC, and it is assumed that each individual CRCtumor accumulates an average of 9 CRC pathogenic mu-tations out of this total pool of 70 mutations [16].The heterogeneous pathogenesis of CRC comply with

the hallmarks of cancer defined by Hanahan and

Weinberg [20] and the cancer genome landscape as de-fined by Vogelstein et al [21]. Underlying these hall-marks of cancer, Hanahan and Weinberg proposed thatgenome instability and inflammation are two underlyingdriving forces [20]. These two processes or mechanismsare closely intertwined, since inflammation is a majorcause of oxidative stress, and oxidative stress is a majorcause of genome instability. Although inflammation andoxidative stress ultimately may be related to all CRCcases, the degree of inflammation and oxidative stressmay vary significantly with the molecular signaturepresent in the individual CRC patient [22].In clinical trials and various models systems, we have

identified a number of plant foods (e.g. berries, nuts,spices, coffee and specific fruits and vegetables) with thepotential of dampening inflammation and oxidativestress [23–29]. Furthermore, a number of studies havealso suggested that adherence to a prudent diet (e.g.Mediterranean diet) reduce inflammation and oxidativestress [30, 31]. We suggest that a prudent diet rich inspecific plant-foods may be beneficial for CRC patients,especially those CRC cases with molecular signaturescreating major inflammation and oxidative stress.No intervention studies have investigated the role of

diet in disease outcomes and survival in CRC-patientsafter diagnosis. Furthermore, no previous diet interven-tion study has focused on dampening inflammation andoxidative stress in this cancer population. This paperpresents the background and design of a randomizedcontrolled food-based diet intervention that examinesthe effects on disease outcomes and survival in CRC sur-vivors. The diet intervention includes foods and drinksthat have been suggested to dampen inflammation andoxidative stress. While specific anti-inflammatory andantioxidant-rich foods are emphasized in each food cat-egory, the complete intervention is fully in accordancewith the prudent diet recommended by the Norwegianfood-based dietary guidelines (NFBDG) [32] (i.e. a dietsimilar to the Mediterranean diet).

ObjectivesOutcomes are inconsistently defined in many clinicalcancer trials [33, 34]. For the primary outcomes, wehave used the proposed guidelines for outcomes asdescribed by Punt et al [34]. The two primary out-comes are (to be assessed when all patients havecompleted 5, 10, and 15 years, respectively, of follow-up after baseline):

1. Disease-free survival (DFS) (events are defined asdetection of local recurrence or metastasis or anysecond cancer or death from any cause)

2. Overall survival (OS) (event is defined as deathfrom any cause)

Henriksen et al. BMC Cancer (2017) 17:83 Page 2 of 17

Page 85: Malnutrition and dietary interventions in colorectal

Secondary outcomes are:

I. Time to recurrence (events are defined as detectionof local recurrence or metastasis)

II. CVD -free survival (events of CVD (ICD-10;chapter I) or death from any cause)

III. CRC-specific survival (death due to CRC)IV. Total cancer-specific survival (death due to CRC or

any other cancer)V. Inflammatory disease-specific survival (death due

to inflammatory disease)VI. Cardiovascular (CVD)-specific survival (death due

to CVD)VII.New morbidity of other diet-related chronic

diseases (e.g. ischemic coronary heart disease,cerebrovascular disease, thromboembolic disease,type 2 diabetes, obesity, hypertension and chronicobstructive pulmonary disease)

VIII.Dietary intake and nutritional statusIX. Physical activity and functionX. Nutrition biomarkers (e.g., carotenoids, fatty acids,

25-hydroxy vitamin D)XI. Body compositionXII. Anthropometric measures (e.g. weight, waist and

hip circumference)XIII.Biomarkers for inflammation and oxidative stress

(e.g. isoprostanes, cytokines)XIV.Transcription- and epigenetic profilesXV. Biomarkers for cardiovascular disease, metabolic

syndrome, type 2-diabetes, thromboembolicdisease and cancer (e.g. blood pressure, total/LDL-cholesterol, HbA1c, CRP, IL-6, IL-10, TNFα)

XVI.Health related quality of life and fatigue

The secondary outcomes will be assessed after 5, 10,and 15 years and described in detail in subsequent re-ports. In addition, intervention effects on secondary out-comes VII-XVI will also be assessed at 6 months, 1 yearand 3 years follow-up.

Methods and DesignStudy designThe CRC-NORDIET study is a multicentre, randomizedcontrolled trial (RCT), with two parallel study arms. Theintervention group receives an intensive dietary inter-vention and general advice on physical activity (seebelow), whereas the control group only receives standardgeneral dietary advice and general advice on physicalactivity. Newly diagnosed CRC patients undergoing sur-gery are recruited to the study. In addition, an age-matched CRC-free reference group (will be publishedelsewhere) will also be included. The intervention starts2–9 months after surgery (i.e. baseline), and consists oftwo periods: an intensive period that lasts 12 months,

and a subsequent maintenance period which lasts anadditional 14 years. Patients are invited to the studycentre, situated at the Department of Nutrition, Univer-sity of Oslo, at baseline, 6 and 12 months after baseline,and 3, 5, 7, 10 and 15 years after baseline. Additionalfollow-ups by regular mail, phone and e-mail, occurthroughout the study. The study flow diagram is pre-sented in Fig. 1. The design and handling of data of theCRC-NORDIET study is in fully agreement with theCONSORT statement [35].

Patients and eligibilityMen and women 50 to 80 years of age with newly diag-nosed primary invasive colorectal cancer (ICD-10 18-20), staged I-III (TNM-staging system [36]) are eligiblefor the study. The patients must be able to read andunderstand Norwegian and to provide a signed informedwritten consent. Patients unable to perceive informationand understand the intervention due to diagnosed de-mentia, or altered mental status as well as patients par-ticipating in other RCTs in conflict with our trial areexcluded from the study. Precise inclusion and exclusioncriteria are presented in Table 1.

Recruitment and randomizationPatients are recruited from Oslo University Hospital andAkershus University Hospital within the South-EasternNorway Regional Health Authority. Screening for eli-gible patients is performed by research investigators incooperation with hospital personnel by monthly reviewsof surgery lists and medical records. Eligible patients areinvited within 9 months from surgery.Patients accepting the invitation sign an informed con-

sent. Signed informed consent gives permission to thestudy personnel to take biological samples, performphysical measurements, and retrieve information frommedical records, health registries and questionnaires. In-formation about storage of biological materials and useof individual data retrieved during the whole study foranalysis and publishing purposes is also included in theinformed consent letter.Prior to baseline of the intervention, patients are ran-

domized to either intervention group A or control groupB in blocks of four. The random number sequence iscomputer-generated for each hospital. The personwho generates the allocation sequence is neither thesame person who determines eligibility nor the personthat informs patients about their allocated studygroup. The patients are informed about the studygroup assignment at the baseline visit. Due to the na-ture of the intervention, neither the registered dieti-tians, nor the other research coworkers who meet thepatients at the study centre, nor the patients them-selves are blinded to group allocation.

Henriksen et al. BMC Cancer (2017) 17:83 Page 3 of 17

Page 86: Malnutrition and dietary interventions in colorectal

Intensive period of interventionThe CRC-NORDIET study offers an extensive interven-tion program for patients in group A, consisting of indi-vidual counselling on nutrition and physical activity,grocery discount cards, delivery of free food items, groupmeetings, printed materials, access to a CRC-NORDIETwebpage and contact by telephone and e-mail. The pa-tients in group B are offered the same individual coun-selling on physical activity as group A, as well as generalgroup meetings. An overview of the intervention pro-gram and the instruments used are presented in Table 2and Table 3, and in Additional file 1.

Group A: diet interventionColorectal cancer patients experience different diseasecourses due to different stages at diagnosis, location oftumor, surgical procedure and adjuvant treatment. The dietintervention is therefore designed to meet the patients’ in-dividual needs after surgery. In the initial phase, whensymptoms related to cancer and cancer treatment are mostcommon, the dietary focus is mainly on recovery and treat-ment of symptoms and progressive weight loss. Later,when symptoms and weight loss are treated and undercontrol, and the disease conditions are more stable, themajor focus is long-term disease-free living and secondarypreventions. In this phase, we emphasize a diet which maydampen chronic inflammation and oxidative stress, fully inaccordance with the NFBDG. A number of strategies areimplemented to improve compliance to the recommendeddiet of the CRC patients in group A (see below).

The dietary recommendations in the CRC-NORDIETintervention The NFBDG, published in 2011, was de-veloped to prevent chronic diseases in the general popula-tion [32]. These guidelines are based on a comprehensive,systematic review of the evidence linking diet to risk ofchronic diseases, including cancer. The guidelines do notprovide a detailed diet plan, but define major aspects ofthe diet (Additional file 2). In the current study, theparticular focus will be on the following NFBDGrecommendations

1) daily intake of fruits, berries and vegetables(≥500 g/day)

2) weekly intake of 300-450 g fish3) daily intake of 70-90 g wholegrains4) limiting red and processed meat to maximum

500 g/week5) keeping body weight within normal range of body

mass index (BMI)6) reduce intake of added sugar to < 10 E%7) reduce salt intake to less than 6 g/day8) achieving an average of at least 30 min of

moderate (3–6 metabolic equivalents (METs))physical activity per day or 150 min of moderatephysical activity per week

The NFBDG can be implemented in different ways.For example, the recommendations of eating 500 gfruits, berries and vegetables every day may includedifferent selections of individual foods, all compliant

Fig. 1 Study flow diagram

Henriksen et al. BMC Cancer (2017) 17:83 Page 4 of 17

Page 87: Malnutrition and dietary interventions in colorectal

to the quantitative advice. However, not all of thesefoods may dampen inflammation and oxidativestress. Since inflammation and oxidative stress areubiquitous as common basic pathogenic mechanism,we have selected to compose the intervention notonly according to the NFBDG, but also by emphasiz-ing those foods with strongest evidence for dampen-ing low grade chronic inflammation and oxidativestress: We have identified foods and drinks that havehigh contents of redox-active compounds and/orhave antioxidative effects individually or in combin-ation in in vitro models, animal models, clinical tri-als and/or epidemiological studies [23–26, 28, 29,37–56] (detailed list with references in Additionalfile 3):

� Drinks (e.g. coffee, black tea)� Fruits and vegetables (e.g. onions, broccoli,

tomatoes, carrots, pomegranates, garlic,oranges, olives)

� Berries (e.g. blueberries/bilberries, blackberries,and raspberries)

� Nuts (e.g. walnuts, almonds, and hazel nuts)� Herbs and spices (e.g. thyme, oregano, clove,

cinnamon, and rosemary)� Whole grain (e.g. barley)� Miscellaneous (dark chocolate)

Furthermore, we have also identified that the fol-lowing foods and drinks may have anti-inflammatoryeffects individually or in combination in cell cul-tures, animal models, clinical trials and/or epidemio-logical studies (detailed list with references inAdditional file 3):

Table 2 Instruments used to facilitate compliance in intervention group A during the first 12 months

Baseline (at study centre) 1 month(at home)

3 months(at home)

6 months(at study centre)

9 months(at home)

12 months(at study centre)

Nutritional counselling Face to face individual Phonecall

Phonecall

Face to faceindividual

Phonecall

Face to faceindividual

Free-of-charge food Delivered at the visit Homedelivery

Delivered at thevisit

Home

delivery

Delivered atthe visit

Information/courses

Folder with information on thestudy and the study instruments

Inspirationday andCookingcourse

Discount card Discount card (25% discount onhealthy foods)

CRC-NORDIET Webpage/e-mail

Login-restricted webpage accessand e-mail communication

Physical activity Access to free training facilities(“Pusterommet”)

Reports from non-biologicalmeasurements

Reports sent to the patients afterevery visit

Table 1 Inclusion and exclusion criteria

Inclusioncriteria

Primary adenocarsinoma colorectal cancer(ICD-10 C18-C20):

C18 Malignant neoplasm of colon

C18.0 Caecum

C18.1 Appendix

C18.2 Ascending colon

C18.3 Hepatic flexure

C18.4 Transverse colon

C18.5 Splenic flexure

C18.6 Descending colon

C18.7 Sigmoid colon (sigmoid (flexure)

C18.8 Overlapping lesion of colon

C18.9 Colon, unspecified

C19 Malignant neoplasm of rectosigmoidjunction

C20 Malignant neoplasm of rectum

TNM stage I-III

Age 50–80 years old

Exclusioncriteria

Colorectal adenoma, carcinoid, abdominalcarcinomatosis or sarcoma

Unable to read and understand Norwegian

Unable to perceive information and understandthe intervention as such due to dementia oraltered mental status

Unable to follow the dietary intervention due tomedical/clinical conditions e.g. total parentalnutrition, permanently institutionalized

Participation in another study in conflict with theintention of the CRC-NORDIET study

Henriksen et al. BMC Cancer (2017) 17:83 Page 5 of 17

Page 88: Malnutrition and dietary interventions in colorectal

� Coffee� Fruits and vegetables (e.g. tomatoes, carrots, dog

rose)� Nuts (e.g. walnuts)� Berries (e.g. strawberries, blueberries/bilberries, and

blackberries)� Whole grains� Herbs and spices (e.g. thyme, oregano, and

rosemary)

During the 15 year intervention period, these foodsand drinks are gradually implemented in the advice togroup A.While these antioxidant- and phytochemical rich foods

are advised as part of a balanced diet according to theNFBDG, patients were advised not to take any antioxi-dant supplements [55, 57].

Intervention strategiesThe following instruments are used to facilitate compli-ance to the intervention in group A.

1. Individualized nutrition counselling by a registeredclinical dietitian.

The nutritional counselling aims to meet the individ-ual nutritional needs as well as educate the patients onhow to change dietary habits in accordance with theNGBDG. In order to individualize the dietary advice, theregistered clinical dietitian performs a comprehensiveevaluation in each of the meetings (Fig. 1). The Patient-Generated Subjective Global Assessment (PG-SGA) tool[58] is used to assess nutritional status and nutritionalimpact symptoms. Weight and height measured thesame day is used to calculate BMI, and current weight iscompared with previous weight measurements to calcu-late weight changes. The presence of stoma is recordedas well as treatment status (i.e. whether or not the pa-tient receives adjuvant treatment). Dietary intake is

assessed by 24-h recall (at baseline). In addition, the reg-istered clinical dietitian characterizes the patient’scurrent diet in relation to the NFBDG, and record use ofsupplements.When the nutritional evaluation is completed, the pa-

tient receives dietary advice based on nutritional statusand weight history. If the patient is malnourished or atrisk of malnutrition (i.e. PG-SGA category B or C), diet-ary counselling primarily focuses on improving nutri-tional status by treating symptoms, ensuring anadequate energy and protein intake, and to prevent fur-ther nutritional deterioration. In terms of progressiveweight loss, patients with PG-SGA B or C with BMI >20are recommended to stabilize their body weight. Patientswith BMI < 20 are recommended to increase their bodyweight within the range of a normal BMI, determined inthe current study as BMI 20–27 for patients aged 50–80years [59, 60]. Well-nourished patients (i.e. PG-SGA cat-egory A) with BMI >27 are recommended to decreasetheir weight within normal BMI range. The recom-mended change (weight gain or weight reduction) is setto maximum 3 kg in 6 months to ensure an optimalchange in body composition.If the patient is evaluated as well-nourished (PG-SGA

A), the dietary counselling primarily focuses on theNFBDG. Examples of week menus are used to illustrateexamples of foods and amounts to be eaten in adherencewith the NFBDG. Food alternatives are given to adjustthe week menu to the patient’s personal eating habitsand preferences.Motivation to change dietary habits in according to

the NFBDG is recorded by asking whether the patientconsiders herself/himself to be either “very motivated”,“motivated”, “less motivated” or “not motivated”. Whenone of the last two categories is present, the registeredclinical dietitian explores the potential to increase motiv-ation by using techniques from Motivational Interviewing(MI) [61]. The degree of motivation (“very motivated”,“motivated”, “less motivated” or “not motivated”) is takeninto account in each of the counselling sessions.Each of the nutritional consultations is intended to re-

sult in a few dietary goals in agreement with the patient.It is emphasized that the patient defines her/his personalgoals to increase the chances that he or she will succeedin changing dietary habits. The registered clinicaldietitian aims at encouraging the patient to achieve thesegoals and the goals will be revised at next session. Thetelephone-based counselling in between the meetings atthe study centre focus at monitoring the patient’s bodyweight status, dietary pattern according to the prede-fined goals and motivational status. In addition to thescheduled consultations at the study centre and by tele-phone, the patients have the opportunity to contact theregistered clinical dietitian by e-mail during the entire

Table 3 Instruments used in the control group during the first12 months

Baseline(at study centre)

1–12 months (1, 3, and 9months at home, 6 and 12months at the study centre)

Information/courses

Folder withinformationon the study

Inspiration day

Physical activity Access to freetraining facilities(“Pusterommet”)

Reports fromnon-biologicalmeasurements

Reports sent tothe patients afterevery visit

Henriksen et al. BMC Cancer (2017) 17:83 Page 6 of 17

Page 89: Malnutrition and dietary interventions in colorectal

intervention period. The same registered clinicaldietitian follows the patient during the entire interven-tion period, when possible.

2. Discount card (25% discount on healthy foods)

The patients in the intervention group are offered a dis-count card from the retailer company, “Norgesgruppen”,which is Norway’s largest enterprise within the grocerymarket, with a market share of 40%. The discount cardcan be used within the first year of the intervention andgives a 25% discount on all fresh vegetables, fruit, berriesand fish and on all food items marked with the keyholesymbol, which is used by the health authorities to labelfood that is considered the most healthy within its foodcategory [62]. The discount card can be used in all foodstores and supermarkets within “Norgesgruppen”.

3. Delivery of specific foods

The CRC-NORDIET is sponsored by several food pro-ducing companies with free food items, specifically se-lected in accordance with the anti-inflammatory andantioxidant-rich foods emphasized in this study, such asjuice, garlic, tomato juice, fish, coffee, tea, cereals, wholegrain bread, oils etc. At all visits to the study centre, thepatients in group A receive a bag containing a mixtureof these food items. In addition, they receive a box withfree food items delivered to their homes two times dur-ing the intensive period of the intervention.

4. CRC-NORDIET website

The patients in the intervention group get access to alogin-restricted, dynamic website with detailed informa-tion about the NFBDG, portion sizes of recommended in-take of fruits and vegetables and whole grain, food recipes,examples of week menus, dietary advice for treatment-related symptoms and advice on physical activity. Inaddition, information about the CRC-NORDIET studyand contact information for the study organizers aregiven. The website is continuously updated.

5. Printed materials

The patients in the intervention group receive printedmaterials at the first visit to the study centre and at allfollow-ups to ensure that also patients who do not usethe internet get all relevant information.

6. Cooking course

During the first 6 months of the intervention, each pa-tient in group A is offered a one-day cooking course.

This course is led by a registered clinical dietitian whofollows a protocol developed for the CRC-NORDIETintervention. The aim of the cooking course is to givethe patients practical experience in making healthydishes and to introduce healthy choices when shoppingfor food. The course consists of a one hour lecture onthe NFBDG and how to implement these guidelines indaily cooking. All recipes can also be found on the CRC-NORDIET web site.

7. Physical activity

The CRC-NORDIET study has an agreement with“Active against cancer” [63], a non-governmental non-profit organization founded in 2007. The organizationoperates a free training studio (“Pusterommet”) for can-cer patients at several hospitals in Norway. The physicaltherapists working at these studios are instructed to giveindividualized advice for exercises during and after can-cer treatment. The CRC-NORDIET patients are encour-aged to utilize this offer.Moreover, the CRC-NORDIET patients are advised to

practice moderate physical activity for at least 30 minper day, or 150 min per week, and they receive a bookleton how to be physically active in daily life. In addition,they are recommended to use local facilities, includingswimming pool, health training centres and walks intheir neighbourhoods.

8. Inspiration day

The patients in Group A are invited to an inspirationday within the first 6 months of the intervention. Theday opens with a 45 min lecture about the aim and back-ground of the CRC-NORDIET study by the projectleader, with special focus on the NFBDG. The patientsare shown examples of different portion sizes of fruitsand vegetables, nuts, whole grain products, the food-dish-model, and have the opportunity to talk to regis-tered clinical dieticians. The last part of the inspirationday focuses on physical activity, and starts with a lectureabout physical activity incorporated in daily life. The pa-tients also meet the physical therapists from “Pusterom-met”. The meeting ends with a lunch and a quiz aboutphysical activity, and each patient receives a pedometeras an incentive to be physically active.

9. Written reports

The patients receive reports from the non-biologicalsamplings (e.g. anthropometric measurements and bloodpressure, described in detail in the following section)performed at the three time points during the intensiveintervention period (baseline, 6 and 12 months after

Henriksen et al. BMC Cancer (2017) 17:83 Page 7 of 17

Page 90: Malnutrition and dietary interventions in colorectal

baseline), as well as a one-year report showing the devel-opment during the last year. Reports from the physicalactivity monitors are given to the patients after the firstintensive year of intervention.

Group B: control group

1. Physical activity

Patients in the control group receive the same basicadvice on physical activity as well as free access to thetraining studio as patients in the intervention group (seeabove).

2. Inspiration day

The inspiration day is structured identically as for groupA, except for the session focusing particularly on diet,which is excluded in the inspiration day for group B.

3. Dietary information

The patients in group B receive a booklet with basicdietary advice at baseline. In contrast to the interventiongroup, the control group receives no individualized diet-ary advice adapted to their eating habits and preferences.If they seek counselling concerning symptoms related tocancer or cancer treatment, the registered clinical dieti-tians provide dietary advice based on information frombooklets and other printed materials already available inthe hospitals. This information and dietary advice is con-sidered as part of the standard care.

4. Written reports

The patients in group B receive written reports simi-larly as group A after all visits during the intensive inter-vention period.

Moderate intervention during maintenanceperiod (year 2–15)During the maintenance period, which starts after thefirst intensive year and lasts for 14 years, both groups re-ceive reports (e.g. anthropometric measurements andblood pressure) following every visit at study centre(year 3, 5, 7, 10 and 15).The patients in group A are invited to an inspiration

day every year during moderate period of intervention.The aim of these meetings is to maintain the focus onfoods dampening inflammation and oxidative stress andthe NFBDG, and to encourage the patients to continuefollowing the guidelines in a long-term perspective. Inaddition, group A are offered dietary counselling at eachvisit at the study centre, as well as a telephone

counselling by the registered clinical dietitians once ayear. They also have access to the CRC-NORDIET web-page which is continuously updated with informationand encouragements (e.g. recipes, nutrition information,motivational tips and relevant popular reports from nu-tritional sciences) until the end of study participation.An overview of the instruments used during the main-tenance period is presented in Table 4.

Assessment of primary outcomesSeveral registries and medical records will be used forassessment of primary outcomes. The registries and timepoints for primary outcome assessment are summarizedin Table 5.

Questionnaires, biological samplings and measurementsGroup A and Group B are undergoing equal regimes ofmeasurements and biological samplings at all visits(Additional file 4). All patients are also asked tocomplete several questionnaires regarding demographicinformation, dietary intake, health status and physicalactivity (described below) (Additional file 4). The ques-tionnaires administered at baseline of intervention arealso completed at 6 months and 12 months follow-up.After the first year, the patients are invited to the studycentre for questionnaires, biological samplings and mea-surements 3, 5, 7, 10 and 15 years after baseline. Inaddition to the visits to the study centre during themaintenance period, finger prick blood sample equip-ment (dried blood-spot cards) and questionnaires aresent to the patients’ home at certain time points andsubsequently returned to the study centre.

Demographic informationA short questionnaire is used to assess demographiccharacteristics including age, gender, marital status, eth-nicity, level of education, working status, family historyof CRC or other type of cancer.

Table 4 Instruments offered to the respective groups duringmaintenance period of intervention

Instruments Time points

2, 4, 6, 8, 9, 11,12, 13, 14 years

3, 5, 7, 10,15 years

Dietary counselling at study centre(Group A)

X

Dietary counselling by telephone(Group A)

X X

Inspiration day with extended dietsession (Group A)

X X

CRC-NORDIET Website/e-mail(Group A)

X X

Reports from non-biologicalmeasurements (Group A and B)

X

Henriksen et al. BMC Cancer (2017) 17:83 Page 8 of 17

Page 91: Malnutrition and dietary interventions in colorectal

Assessment of dietary intakeSemi-quantitative food frequency questionnaire (FFQ)The semi-quantitative 282-item FFQ used in CRC-NORDIET is designed to assess habitual diet over thepreceding year, including both frequency of intake andportion sizes. The FFQ is described and validated else-where [64, 65].

Compliance questionnaire The compliance question-naire is a semi-quantitative short 63-item FFQ, devel-oped within this study and designed to assess the dietaryintake (grams per day) and physical activity (minutes perday) for the last 1–2 months. The questions correspondto the food groups and the recommendations regardingphysical activity of the NFBDG. The questionnaire willbe validated within the first period of study.

Food records Food intake is recorded by using a 7-daysweighed food record. The patients are provided with afood diary and a digital scale, and are instructed on howto weigh and record all foods and beverages consumedduring a period of seven days. The food diary include alldays of a week, and can either record seven consecutivedays or be divided into two periods of three and fourdays within two weeks. The food records are performedin a subgroup of patients (will be published elsewhere).

24-h recall A registered clinical dietitian performs a 24-h recall at baseline by asking the patients in the inter-vention group in details about the intake of foods anddrink during the past 24-h period. The 24-h recall is per-formed only in intervention patients since it is an inte-grated part of the nutritional counselling.

Assessment of physical activity and functionRecording of daily physical activity The physical activ-ity monitor SenseWear Mini Armband (BodyMedia,Pittsburgh, Pennsylvania, USA) [66] is used to recorddaily physical activity, inactivity and energy expenditureduring seven consecutive days among all patients in bothstudy arms at all visits. The armband monitors

physiological data such as heat flux, galvanic skin re-sponse, 3-axis accelerometer and skin temperature. Alldata are retrieved from the armband to the computerwith the SenseWear Professional Software [66]. The par-ticipant are instructed how to use the armband, and re-turn it in a stamped envelope to the CRC-NORDIETstudy at the end of the test period. The armband is pre-programmed with the co-predictors such as weight,height, age, gender, smoking status (smoker/non-smoker) and placed around the non-dominant arm.

Self-reported physical activity The patients are askedto complete a questionnaire regarding frequency, intensityand duration of their daily physical activity, as well as dur-ation of sedentary time. These questions are based on thequestionnaire from the HUNT 3 study in Norway [67].

6-min walking test Patients are invited to a 6-min walktest (6MWT) at several time points. The test is per-formed indoors, along a long, flat, straight, enclosed cor-ridor with a hard surface. The walking course is 30 m inlength, and cones mark the turnaround points. A count-down timer (or stopwatch) is used to record the time ofthe test. Prior to the test, the researcher measures theblood pressure of the patient. In addition, the pulse ismonitored before, during and after the test. The patientsare asked to grade its level of shortness of breath andthe level of fatigue by using the Borg scale 6-20 beforeand after the test. Total length of walking (in meters) isrecorded during 6 min of time.

Sit-to-stand test The test is performed by the use of astraight back chair with a solid seat at the height of44 cm. The patients are instructed to sit on the chairwith arms folded across their chest, and then to standup and sit down as quickly and frequently as possiblewithin 30 s, keeping both arms folded across the chest.The number of stands during this period is counted.

Handgrip strength Hand-grip strength is measured bythe MAP 80 K1 Hand grip dynamometer (KERN &SOHN GmbH, Balingen, Germany) and measured as de-scribed in the manufacturer’s protocol [68]. The max-imal strength of hand grip (kg) is recorded. For womenand men, a 40 kg- and 80 kg-spring is used, respectively.The grip strength is measured with one punch and re-peated three times on both hands. The maximum hand-grip strength on both left and right hands are recorded.

Assessment of nutritional statusPatient-Generated Subjective Global Assessment(PG-SGA) Nutritional status is measured by using thescored PG-SGA [58], a nutritional assessment tool spe-cifically developed and validated for cancer patients. A

Table 5 Data source used to assess primary outcomes

Outcome Instrument 5 yearsafterbaseline

10 yearsafterbaseline

15 yearsafterbaseline

DFS Colorectal Cancer Registry ofNorway, Cancer Registry ofNorway, Cause of DeathRegistry in Norway,Norwegian Patient Registry,Norwegian PrescriptionDatabase

X X X

OS Cause of Death Registry inNorway

X X X

DFS disease-free survival, OS overall survival

Henriksen et al. BMC Cancer (2017) 17:83 Page 9 of 17

Page 92: Malnutrition and dietary interventions in colorectal

translated (Norwegian) version is used. Both the globalcategories well-nourished (A), moderate malnourished(B) and severe malnourished (C), as well as the numer-ical scoring system are used to characterize the nutri-tional status.

Anthropometric measurementsBody weight Body weight (kg) is measured by using anon-slip Marsden M-420 Digital Portable Floor Scale(Marshden, Rotherham, South Yorkshire, United Kingdom)or a digital wireless measuring station for height andweight, Seca 285 (Seca, Birmingham, United Kingdom)[69]. Measurements are performed with light clothes andwithout shoes. Body weight is recorded with 2 decimalsand the kind of clothing is recorded.

Height Height (cm) is measured using either a mechan-ical height rod (Kern MSF- 200, [68]) or a digital wire-less stadiometer (Seca 285 [69]). The height is recordedwith one decimal precision.

Waist and hip circumference Waist circumference ismeasured at the midpoint between the lower margin ofthe last palpable rib and the top of the iliac crest,whereas the hip circumference is measured around thewidest portion of the hips. Waist and hip circumferenceare used to calculate the waist hip-ratio (WHR) which isa well-established indicator of abdominal fatness [70].

Body composition analysisBioelectrical impedance analysis (BIA) BIA is per-formed under standardized conditions by the use of BIA101 (SMT Medical, Würzburg, Germany) that applies acurrent of 0,8 μA at a frequency of 50 kHz. Four skinelectrodes are placed on hand and foot of the patientswhen lying in supine position. All measurements areconducted on the patients’ right side as instructed bythe manual. Resistance (Rz) and reactance (Xc) are usedin appropriate and validated equations to calculate bodycomposition compartments such as fat mass, fat freemass and muscle mass. In addition, BIA is also per-formed with Seca mBCA515 (Seca, Birmingham, UnitedKingdom) [71]. Patients carrying a pacemaker are ex-cluded from the BIA measurements.

Dual-energy x-ray absorptiometry (DXA) The LunariDXA (GE Healthcare Lunar, Buckinghamshire, UnitedKingdom) is used to measure bone mineral density andbody composition, including quantification of visceral fat.

Computed tomography (CT) CT images taken rou-tinely for clinical purposes are used for body compos-ition analysis, i.e. quantification of fat (visceral,subcutaneous and intermuscular adipose tissue) and

skeletal muscle. The images are analysed using the Slice-o-matic software, version 4.3 (Tomovision, Montreal,Canada). The third lumbar vertebra (L3) is chosen asstandard landmark since skeletal muscle, lean tissuemass and adipose tissue at this level are significantly cor-related to whole-body tissue in healthy adults [72].

Blood pressureBlood pressure (BP) is measured with the digital bloodpressure patient monitor Carescape V100 (GE Health-care, Fairfield, USA) and performed by trained staff fol-lowing the clinical procedure as described by themanufacturer [73]. After a 5 min resting period in a si-lent room, BP is measured four times on the non-dominant arm with intervals of one minute.

BiobankA variety of biological samples will be collected at differ-ent time points during the study and will be used for thepurposes of measuring surrogate outcomes, biomarkersof food intake and for identification of phenotypes asso-ciated with different responses to the intervention.

Venous blood samples Overnight fasting blood samplesare taken between 07.30 and 10.30 at the study centre bya trained technician. BD Vacutainer® (Becton, Dickinsonand Co, Franklin Lakes, NJ, USA) tubes are used to col-lect ethylene diamine tetraacetic acid (EDTA) samples(no. 367861 and 366643), serum samples (no 368774),lithium heparin samples (no 367526), and citrate sam-ples (no 369714).Serum tubes are placed in room temperature for

30 min. Serum, EDTA and heparin samples are centri-fuged at1500 g, 10 min, 15 °C. Serum, plasma and redblood cells are aliquoted, and immediately stored in at−80 °C until further analysis. Whole blood from EDTAsamples are also aliquoted for e.g. DNA extraction andDNA damage/repair analysis The buffy coat from theheparin samples are either frozen at −80 °C for later ana-lysis or used to obtain isolated peripheral blood mono-nuclear cells (PBMC) through Percoll centrifugation.The isolated PBMCs from heparin samples are used forex vivo experiments. Two citrate tubes are kept 1 h re-spectively at 4 °C and room temperature before centrifu-gation (2500 g, 15 min, 4 °C) to obtain core plasma,plasma and red blood cell aliquots that are stored at-70 °C. One citrate tube is centrifuged (2500 g, 15 min,4 °C) within 30 min of sampling, and core plasma isstored at - 80 °C for further analysis of thromboembolicfactors. The citrate buffy coats are used to obtain iso-lated PBMCs for the study of DNA repair and DNAdamage. PAXgene Blood RNA Tubes (cat.no 762115,PreAnalytiX, Hombrechtikon, Switzerland) are used assource for total blood RNA. The tubes are kept 2 h at

Henriksen et al. BMC Cancer (2017) 17:83 Page 10 of 17

Page 93: Malnutrition and dietary interventions in colorectal

room temperature before they are frozen at -20 °C for24 h and subsequently transferred to −80 °C until timefor RNA isolation.

Isolation of buffy coats from EDTA samples The EDTAbuffy coats are re-solved in 9% NaCl (cat.no 586564,B.Braun Melsungen AB, Melsungen, Germany) solutionbefore added on top of 4 ml Lymphoprep (cat.no1114545, Axis-Shield, Oslo, Norway) in a 15 ml tube forcentrifugation (20 min RT 400 g) to isolate PBMCswhich are further used for a chromatin crosslinking pro-cedure. The crosslink procedure for preparing the cellpellets for ChIP-chip analysis are performed as follows:Firstly, PBMCs are allowed to crosslink with 1% formal-dehyde (final concentration) for 10 min at roomtemperature, adding glycine (0.125 M) for 10 min atroom temperature to stop the crosslinking process. Afterwashing the cell pellets twice with 10 mL of ice-cold 1 ×PBS the pellets are immediately stored in 2 ml plastictubes at −80 °C until proceeding further with protocolsfor Chip-on-Chip analysis at a later time point.

Finger prick blood samples Finger prick blood samplesfor analysis of e.g. biomarkers of dietary intake, oxidativestress and oxidative damage are collected by the driedblood spots (DBS) method as previously described [74].DBS cards (2 cards per patient) are allowed to dry inroom temperature for 2 h and are frozen at −80 °C inairtight aluminium bag with a desiccant until furtheranalysis.

Urine samples Biomarkers of food intake, oxidativestress and other risk factors related to the progression ofCRC will be measured in urine. Urine samples are col-lected from a subpopulation several times during the inter-vention by the methods as previously described [75–77].

Faeces samples Microbiotica and biomarkers related toCRC will be measured in faeces samples which are col-lected from a subpopulation several times during theintervention. The patients will receive a specific faecessample tool kit and are asked to collect the sample athome and mail it to the study centre. Sampling and ana-lysing of the faeces samples will be performed by follow-ing the procedure as described by Naseribafrouei [78].

Tumour tissue Molecular signatures in CRC tumoursthat are linked to inflammation, oxidative stress and en-ergy balance have been shown to predict response tolifestyle intervention. Characterization of tumor markerswill be performed by immunohistochemistry, PCR, se-quencing and q‐PCR (to be published elsewhere). Fur-thermore, we will study whether tumor markers predictresponse to the dietary intervention. Samples of tumor

tissue are collected at surgery in collaboration with thehospitals. Molecular signature data are also obtainedfrom the CRC biobank project at the Oslo UniversityHospital.

Oral glucose tolerance testPrior to the oral glucose tolerance test, the patient isfasting for at least 8 h. Blood samples (serum and PAXtubes) are taken, and blood glucose is measured with ablood glucose meter [79]. The patients are asked todrink 75 g of glucose (D (+)-Glucose (product number:1370485000, Merck-Millipore Corp, Darmstadt,Germany) in 4 dl of boiled water. The glucose liquid isexpected to be consumed total within 5 min. Blood sam-ples will be taken after 2 h. Exclusion criteria for oralglucose tolerance test are Diabetes Type I, use of insulinand/or fasting blood glucose level exceeding 10 mmol/l.

Health related quality of life and fatigueQuality of life will be self-reported and measured usingthe generic, multi-purpose-form questionnaire forHealth related quality of life (HRQOL) called Short form(SF) health survey consisting of 36 items (SF-36) [80].The 36 items are categorized into eight multi-itemscales; 1) physical functioning, 2) role physical, 3) bodilypain, 4) general health, 5) vitality, 6) social functioning,7) role emotional and 8) mental health as well as asingle-item measuring health transition during the lastyear. The data will first be standardized in order to com-pare results across studies [80] and then recoded accord-ing to a syntax developed by Loge et al [81].A validated generic fatigue questionnaire (FQ) is used

to assess the patients subjective fatigue status (11 items)and the duration and extent of fatigue (2 items) [82].The FQ asks about fatigue symptoms experienced dur-ing the last month compared to how the subject feltwhen she/he was last feeling well [82–85]. Each item hasfour response-choices [82]. The scoring of each responseis based on a Likert- (0, 1, 2, 3) and a dichotomized (0,0, 1, 1) scale. The latter is only used for case definition.The total sum of the Likert-scores is designated total fa-tigue (TF) where higher scores imply more fatigue.

Assessment of new morbidity of diet-related chronicdiseases and adverse eventsNew morbidity of diet-related chronic diseases arisingafter CRC diagnosis (e.g. ischemic coronary heart dis-ease, cerebrovascular disease, thromboembolic disease,diabetes, hypertension and chronic obstructive pulmon-ary disease) will be collected from the national healthregistries in Norway, a comorbidity questionnaire devel-oped for this study designed to assess comorbidity basedon data from the third Norwegian population healthstudy (HUNT 3) [67], and from medical records. These

Henriksen et al. BMC Cancer (2017) 17:83 Page 11 of 17

Page 94: Malnutrition and dietary interventions in colorectal

data will be supplemented by data on drug use from theNorwegian Prescription Register. Adverse events are re-corded based on the Common Terminology Criteria forAdverse Events (CTCAE, Version 4.0) [86].

Sample sizeCalculation for primary outcomesThe sample size calculations are based on assuming aWeibull distribution for the survival times in both arms.We further assume a constant hazard ratio for the inter-vention effect over time and that we have the samefollow-up of 5, 10, or 15 years, respectively, for all pa-tients. Sample sizes required to achieve a statisticalpower of 80% and significance level of 5% were calcu-lated with computer simulations using the spower func-tion in R (version 3.2.0) package Hmisc version 3.17–0.Survival rates in the control group [2, 87] and expectedreduction in mortality rates in the intervention groupare taken from the literature (see Discussion, [88–93]).With a 68% 5-year OS in the control group, we have80% power to detect a 25% reduction in mortality due tothe intervention (corresponding to a hazard ratio of0.71). The required total sample size is then 500 (250 ineach study group) (Table 6).Moreover, sample size calculation based on 25% reduc-

tion in events of DFS after 5 years of surgery (59% 5-yearDFS in the control group), we have 80% power to detectHR of 0.70, with 190 patients in each group (Table 6).

Stratified and subgroup analysisAll of the power calculations are based on a heteroge-neous population of CRC patients.Since post-surgery treatment may vary, and colon ver-

sus rectum cancer may respond differently to the dietintervention, we will also perform stratified statisticalanalysis. It is not known whether treatment effects aredifferent in these subgroups. These stratified analyseswill be conducted with primary outcomes at the latertime-points in the study and at all time-points to assessmean differences between the control and the interven-tion groups in biomarker analysis, as these data normallyrequire fewer patients per group.

Statistical analysisData will be analysed using SPSS (IBM SPSS Statistic22). For the survival outcomes (primary outcome 1 and2, and secondary outcomes I-VI) tests will be performedto compare survival rates between the control and inter-vention groups at 5, 10, and 15 years after baseline.Survival probabilities will be estimated with the Kaplan-Meier method. Cox proportional hazards models will beused to identify prognostic and predictive biomarkersfor survival outcomes.For non-survival secondary outcomes, parametric or

non-parametric tests for two-group comparisons will beused to assess group differences at individual time-points. In addition, mixed effect models for longitudinaldata and regression models will be used to evaluate associ-ation and change over time in dietary intake, nutritionalstatus, body composition, molecular tumor characteristics,physical function and activity, quality of life, fatigue andtreatment related outcomes and to examine differencesbetween the intervention and control groups. All statis-tical tests are performed as two-sided tests. Effects areconsidered statistically significant if p <0.05.

DiscussionThe primary aims of the CRC-NORDIET are to studywhether a healthy diet rich in anti-inflammatory andantioxidant-rich foods and based on the NFBDG can im-prove DFS and OS in CRC patients. To our knowledge,this is the first randomized controlled trial designed toinvestigate the effect of a dietary intervention on theseoutcomes in CRC patients, and to investigate the potentialrole of diet in dampening of inflammation and oxidativestress in these patients. The multiple strategies used toachieve compliance to the intervention during the firstyear, followed by the 14 years maintenance and follow-upperiod make the design of this intervention unique.Data on role of diet on disease outcomes and survival

in CRC survivors is limited. To date, several cohortstudies, but no RCTs have investigated the effects offood-based dietary interventions on these outcomes.

Table 6 Sample size in each group (n) and hazard ratios (HRs*)for selected scenarios of reduction in mortality by intervention.The power is 80% and significance level 5%

Reduction in mortalityby intervention

Survival rates inthe control group

Primaryoutcome

20% 25% 30%

n (HR) n (HR) n (HR)

DFS

5 years 320(0.753)

190(0.696)

140(0.641)

0.59

10 years 240(0.716)

140(0.655)

110(0.597)

0.41

15 years 180(0.680)

120(0.616)

90(0.557)

0.29

OS

5 years 390(0.767)

250(0.712)

160(0.658)

0.68

10 years 280(0.732)

180(0.673)

130(0.616)

0.48

15 years 210(0.695)

140(0.632)

100(0.574)

0.34

* HR = hazard ratio of intervention versus control, which corresponds to theassumed survival rate in the control group and assumed reduction in mortalityby interventionDFS disease-free survival, OS overall survival

Henriksen et al. BMC Cancer (2017) 17:83 Page 12 of 17

Page 95: Malnutrition and dietary interventions in colorectal

Data from a US cohort study with stage III colon cancerpatients suggest that high intakes of red and processedmeat, fat, refined grains and dessert, i.e. a Western diet-ary pattern, after diagnosis are associated with a signifi-cantly reduced disease-free and OS [13]. Similar findingsare reported in a Canadian cohort study with CRC pa-tients staged I-III, where patients with the highest intakeof processed meat the previous year before diagnosishad an 82% increased risk of recurrence or death com-pared with patients with the lowest intake [15].Prospective cohort studies have consistently reported

that physical activity after colorectal cancer diagnosis re-duces risk of mortality. In a meta-analysis of six pro-spective cohort studies, including 7522 CRC survivors,the authors observed that the most physical active survi-vors had a 42% lower risk of total mortality compared tothose who were least active. The risk reduction ofcancer-specific mortality was 39% [94]. No RCT has sofar confirmed that physical activity impacts mortality inCRC survivors.Interventions designed to investigate the effect of diet

separated from other lifestyle factors (smoking, physicalactivity, weight regulation) are needed in order to inves-tigate whether there is a causal relationship between dietand survival as well as disease-related outcomes. Ourintervention is intended to change the dietary habits to-wards a diet in agreement with the NFBDG. These diet-ary guidelines are developed to prevent chronic diseases,including cancers, in the general population. Severallarge cohort studies have shown that there is a consist-ent inverse association between adherence to cancer pre-vention guidelines and cancer-specific and all-causemortality [89, 95]. Among cancer survivors, reduction intotal mortality between highest versus lowest score inadherence to diet recommendations has been docu-mented in five different cohort studies, ranging from24% to 36% (follow-up period from 3.7 to 13.6 years)[88–93]. Association of adherence to American CancerSociety guidelines and reduction in death attributed tocancer has been shown to be 25 and 26% in men andwomen, respectively [89]. Hastert et al documented anassociation of adherence to the WCRF/AICR guidelinesand reduction in cancer-specific mortality of 61% in re-spondents with the highest compared to the lowestWCRF/AICR score (follow-up time of 7.7 years) [95].Furthermore, NFBDG include advice regarding red

and processed meat, dietary fibre, dairy products andgarlic, all of which are related to risk of CRC. Whetherthese dietary factors also may have effect on survival anddisease outcomes, remain unclear. With improvement incancer survival, these perspectives are increasingly im-portant. The main objective of the present study is totest if diet will improve survival and cancer-related out-comes, mediated through reduced inflammation and

oxidative stress. To strengthen this assumption, we havechosen to select specific foods within the NFBDG whichhave been identified as anti-inflammatory or antioxidant-rich in previous preclinical and clinical studies [23–29,37–39, 41–48, 53, 55, 96].Four aspects that may be of importance for achieving

lifestyle changes and facilitate compliance to the inter-vention: 1) timing of intervention, 2) choice of motiv-ational approach to achieve lifestyle changes, 3) durationof intervention, and 4) use of incentives and methods toachieve sustainable changes. Previous studies haveshown that cancer patients in general are particularlymotivated to change dietary habits at the time of diagno-sis, often reported as the teachable moment [97–99]. In-terventions designed to include this teachable momenthave shown to be successful [97–99]. In our trial, weintroduce the dietary intervention within a few monthsfrom surgery, thereby expecting we reach the patientswithin the time frame of this teachable moment. Fur-thermore, principles from MI [61] are implemented ineach of the dietary counselling sessions by trained regis-tered clinical dietitians. Previously published trials thathave succeeded in changing lifestyle behaviours in can-cer patients are based on theoretical frameworks andtheories, including social cognitive behaviour therapyand use of MI. It is emphasized that the patient definesher/his own goals to increase the chances that he or shewill succeed in changing dietary habits. We suggest thatit is important to focus on a few realistic goals at thetime, instead of aiming at changing the whole diet im-mediately. In addition, follow-up by the same registeredclinical dietitian during the entire intensive interventionperiod may be of importance for the commitment to theintervention goals.In order to increase the chances of sustainable lifestyle

changes and compliance to the intervention, our inter-vention consists of a one year intensive period and asubsequent maintenance period which lasts for until14 years. Taking into account that the teachable momentmay vary among the patients and it may take time to es-tablish new sustainable dietary habits, the inclusion of amaintenance period will probably be beneficial with re-gard to an increased long-term adherence to the inter-vention. Previous published trials that have failed incompliance from the patients may have had too shorttime frame of diet intervention.Different strategies are reported to be effective in pro-

moting lifestyle changes in cancer survivors [14, 97, 98,100, 101]. Interventions focusing on individual counsel-ling [14, 98, 100], and also interventions with a mixedstrategy of individual in-person counselling, telephonecounselling and mailed materials [97, 101] have beenshown to be effective in health behaviour change amongCRC survivors. Thus, during the first year, the CRC-

Henriksen et al. BMC Cancer (2017) 17:83 Page 13 of 17

Page 96: Malnutrition and dietary interventions in colorectal

NORDIET study offers individualized counselling, freefoods, a discount card on healthy foods, access to alogin-restricted web page, printed materials, cookingcourses and inspiration day, which may all be effectiveincentives to follow the NFBDG.In placebo-controlled RCTs, an intervention is tested

by comparing one group of individuals who receive theintervention with a control group who receives a pla-cebo. This type of placebo-controlled RCT is most oftennot possible when studying food-, or exercise-based in-terventions, since placebo-foods or placebo-exercise donot exist. In addition, no food based intervention can beanalysed thoroughly without considerations regardingenergy intake and energy expenditure. We have there-fore selected to give the intervention group and the con-trol group the same advice on physical activity. Weinclude careful monitoring of physical activity to controlfor any confounding effects of physical activity. Of eth-ical reasons, we also include standard dietary advice (i.e.standard clinical care) in the control group, as well invi-tations to group meetings and feed-back reports onhealth status. Thus, while the control group in thepresent study is not identical to a placebo group, thisparticular study design was used in order to isolate theeffect of diet intervention on CRC patients, and to re-duce drop-outs from the control group, which is a com-mon concern in long-term intervention trials.Sample size estimation is not straightforward in RCTs

with complex diet intervention and long term hard out-comes. This is especially the case when no similar trialshave been previously published. By using the best avail-able information from scientific literature and NorwayCancer Registry on survival rates in the control groupand expected reduction in mortality rates in the inter-vention group, we have performed power calculations onthe two primary outcomes after 5, 10 and 15 years afterbaseline. We conclude that 250 patients in each groupwould give us a reasonable chance (at 80% power) to de-tect any significant effects (see Methods and Design sec-tion for details) after 5, 10 and 15 years.In a similar study, testing the effects of two different

6 months adjuvant cytostatic protocols (i.e. the MOSAICstudy [102]), 2246 patients who had undergone curative re-section for stage II and III colon cancer, were recruited.After a median follow-up for 38 months, fewer cancer-related events was observed in the alternative treatmentgroup compared to the standard treatment group (HR 0.77,p = 0.002). The main reason for the lower number of pa-tients required in our CRC-NORDIET study compared tothe MOSAIC study is due to an older population with moreexpected events (50–80 years versus 19–75 years) and alonger follow-up time (10 and 15 years versus 3 years).We have also performed a number of power esti-

mations on secondary outcomes (data not shown). In

general, the CRC-NORDIET study is expected to haveenough statistical power to detect significant effects in amajority of these intermediate outcomes (to be pub-lished in relevant reports). These power calculations onprimary and secondary outcomes are also supportedfrom the RCTs with physical activity intervention inCRC and breast cancer patients; Friedenreich and Cour-neya detected significant effects on intermediate out-comes (e.g. inflammation biomarkers) as well as diseaseoutcomes in RCTs with 200–250 patients per group[103–105].

Conclusion and perspectivesThe CRC-NORDIET study investigates whether a dietaimed at dampening inflammation and oxidative stressand in full accordance with the NFBDG will improvesurvival and disease outcomes in CRC patients. ThisRCT is unique in several aspects related to the interven-tions as well as outcomes. Since previous research onthe role of diet for CRC survivors is limited, the study isimportant in order to improve health outcomes and sur-vival in this population.

Additional files

Additional file 1: The CRC-NORDIET intervention program (DOC 42 kb)

Additional file 2: Summary of the 13 recommendations of theNorwegian food-based dietary guidelines (NFBDG) (directly translatedfrom the NFBDG) (DOCX 23 kb)

Additional file 3: Detailed list of foods and drinks with high contents ofredox-active compounds and/or antioxidative effects (DOCX 22 kb)

Additional file 4: Questionnaires, biological samplings andmeasurements (DOC 43 kb)

Abbreviations6MWT: 6-min walking test; ACS: American Cancer Society; AICR: AmericanInstitute of Cancer Research; BIA: Bioelectrical impedance analysis; BMI: Bodymass index; BP: Blood pressure; CIMP: CpG island methylator phenotype;CRC: Colorectal cancer; CRP: C-reactive protein; CT: Computerizedtomography; CTCAE: Common terminology criteria for adverse events;CVD: Cardiovascular diseases; DBS: Dried blood spots; DFS: Disease-freesurvival; DNA: Deoxyribonucleic acid; DXA: Dual-energy x-ray absorptiometry;EDTA: Ethylene diamine tetraacetic acid; FFQ: Food frequency questionnaire;FQ: Fatigue questionnaire; HbA1c: Glycated hemoglobin A1c; HR: Hazardratio; HRQOL: Health related quality of life; HUNT: Helseundersøkelsen i Nord-Trøndelag; ICD: International classification of diseases and related healthproblems; IL: Interleukin; L3: Third lumbar vertebra; LDL: Low densitylipoprotein; METs: Metabolic equivalents; MI: Motivational interview;NFBDG: Norwegian food-based dietary guidelines; OS: Overall survival;PBMC: Peripheral blood mononuclear cell; PG-SGA: Patient-GeneratedSubjective Global Assessment; RCT: Randomized controlled trial; RT-PCR: Real-time quantitative reverse transcription polymerase chain reaction; SF-36: Short form-36; TNFα: Tumor necrosis factor alpha; TNM: Tumor NodeMetastases; WCRF: World Cancer Research Fund; WHR: Waist hip-ratio

AcknowledgementsThe authors would like to thank the hospital personnel from Oslo UniversityHospital and Akershus University Hospital, Norway for assistance duringrecruitment of patients. In addition, we would like to thank all collaborativepartners, technicians and students that have, and will contribute to this

Henriksen et al. BMC Cancer (2017) 17:83 Page 14 of 17

Page 97: Malnutrition and dietary interventions in colorectal

study. A specific list of persons will be acknowledged in the relevant reportsthat will be published from the study.We would also like to thank Norgesgruppen, Mills, Stabburet, Haugen-Gruppen Norway, Norsk Kaffeinformasjon, Balholm and TINE for generoussupport of foods. These companies were not involved in the study designand they will not be involved in the collection, analysis, and interpretation ofdata, or in the publications that will result from this study.

FundingThis project has received funding from Research Council of Norway, ThroneHolst Foundation of Nutrition Research, Norwegian Cancer Society, SouthEastern Norway Regional Health Authority.

Availability of data and materialsAnonymized data resulting from this study may be available upon requestby corresponding author.

Authors’ contributionsHBH and HR had a main responsibility for writing the manuscript. SKB, IP,ASK, SÅB, MTE, GW, IE, AF, MBV, MZ, SS and RB contributed to the studydesign and protocol. RB is the principal investigator. All authors contributedto the writing and approval of the final manuscript.

Competing interestsR.B. is a shareholder in the company Vitas AS. The authors declare that theyhave no competing interests.

Consent for publicationNot applicable.

Ethics approval and consent to participateThe study is approved by the Regional Committees for Medical and HealthResearch Ethics (REC Protocol Approval 2011/836) and by the dataprotection officials in Oslo University Hospital and Akershus UniversityHospital. All biological materials are stored in a biobank at University of Oslo.The biobank will expire in 2040 (according to the REC approval). The study isregistered on the National Institutes of Health Clinical Trials(www.ClinicalTrials.gov; Identifier: NCT01570010). Written informed consentto participate has been obtained from the patients enrolled in the CRC-NORDIET study.

Author details1Department of Nutrition, Institute of Basic Medical Sciences, University ofOslo, Oslo, Norway. 2Department of Gastroenterological Surgery, OsloUniversity Hospital, Ullevål, Norway. 3Institute of Clinical Medicine, Universityof Oslo, Oslo, Norway. 4National Institute for Health and Welfare, Helsinki,Finland. 5Department of Digestive and Paediatric Surgery, AkershusUniversity Hospital, Lørenskog, Norway. 6Oslo Centre for Biostatistics andEpidemiology, Department of Biostatistics, Institute of Basic Medical Sciences,University of Oslo, Oslo, Norway. 7Division of Cancer Medicine, OsloUniversity Hospital, Oslo, Norway.

Received: 12 December 2015 Accepted: 18 January 2017

References1. Brenner H, Kloor M, Pox CP. Colorectal cancer. Lancet. 2014;383(9927)

:1490–502.2. Cancer Registry of Norway. http://www.kreftregisteret.no.3. Food, nutrition, physical activity, and the prevention of cancer: a global

perspective. Washington DC: American Institute for Cancer Research; 20074. Continuous Update Project Report. Food, Nutrition, Physical Activity, and

the Prevention of Colorectal Cancer. Washington: World Cancer ResearchFund / American Institute for Cancer Research; 2011.

5. Edwards BK, Noone AM, Mariotto AB, Simard EP, Boscoe FP, Henley SJ, et al.Annual Report to the Nation on the status of cancer, 1975–2010, featuringprevalence of comorbidity and impact on survival among persons withlung, colorectal, breast, or prostate cancer. Cancer. 2014;120(9):1290–314.

6. Simmonds PD, Best L, George S, Baughan C, Buchanan R, Davis C, et al.Surgery for colorectal cancer in elderly patients: a systematic review.Colorectal Cancer Collaborative Group. Lancet 2000;356(9234):968-74.

7. Jorgensen TL, Hallas J, Friis S, Herrstedt J. Comorbidity in elderly cancerpatients in relation to overall and cancer-specific mortality. Br J Cancer.2012;106(7):1353–60.

8. Shack LG, Rachet B, Williams EM, Northover JM, Coleman MP. Does thetiming of comorbidity affect colorectal cancer survival? A population basedstudy. Postgrad Med J. 2010;86(1012):73–8.

9. van Leersum NJ, Janssen-Heijnen ML, Wouters MW, Rutten HJ, CoeberghJW, Tollenaar RA, et al. Increasing prevalence of comorbidity in patientswith colorectal cancer in the South of the Netherlands 1995-2010. Int JCancer. 2013;132(9):2157–63.

10. Yabroff KR, Lawrence WF, Clauser S, Davis WW, Brown ML. Burden of illnessin cancer survivors: findings from a population-based national sample. JNatl Cancer Inst. 2004;96(17):1322–30.

11. Pedrazzani C, Cerullo G, De Marco G, Marrelli D, Neri A, De Stefano A, et al.Impact of age-related comorbidity on results of colorectal cancer surgery.World J Gastroenterol. 2009;15(45):5706–11.

12. Yancik R, Havlik RJ, Wesley MN, Ries L, Long S, Rossi WK, et al. Cancer andcomorbidity in older patients: a descriptive profile. Ann Epidemiol. 1996;6(5):399–412.

13. Meyerhardt JA, Niedzwiecki D, Hollis D, Saltz LB, Hu FB, Mayer RJ, et al.Association of dietary patterns with cancer recurrence and survival inpatients with stage III colon cancer. Jama. 2007;298(7):754–64.

14. Ravasco P, Monteiro-Grillo I, Camilo M. Individualized nutrition intervention is ofmajor benefit to colorectal cancer patients: long-term follow-up of a randomizedcontrolled trial of nutritional therapy. Am J Clin Nutr. 2012;96(6):1346–53.

15. Zhu Y, Wu H, Wang PP, Savas S, Woodrow J, Wish T, et al: Dietary patternsand colorectal cancer recurrence and survival: a cohort study. BMJ Open2013;3(2).

16. Kinzler KW, Vogelstein B. Landscaping the cancer terrain. Science. 1998;280(5366):1036–7.

17. Leggett B, Whitehall V. Role of the serrated pathway in colorectal cancerpathogenesis. Gastroenterology. 2010;138(6):2088–100.

18. Weisenberger DJ, Siegmund KD, Campan M, Young J, Long TI, Faasse MA,et al. CpG island methylator phenotype underlies sporadic microsatelliteinstability and is tightly associated with BRAF mutation in colorectal cancer.Nat Genet. 2006;38(7):787–93.

19. Lindblom A. Different mechanisms in the tumorigenesis of proximal anddistal colon cancers. Curr Opin Oncol. 2001;13(1):63–9.

20. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell.2011;144(5):646–74.

21. Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz Jr LA, Kinzler KW.Cancer genome landscapes. Science. 2013;339(6127):1546–58.

22. Torres S, Bartolome RA, Mendes M, Barderas R, Fernandez-Acenero MJ,Pelaez-Garcia A, et al. Proteome profiling of cancer-associated fibroblastsidentifies novel proinflammatory signatures and prognostic markers forcolorectal cancer. Clin Cancer Res. 2013;19(21):6006–19.

23. Andersen LF, Jacobs Jr DR, Carlsen MH, Blomhoff R. Consumption of coffeeis associated with reduced risk of death attributed to inflammatory andcardiovascular diseases in the Iowa Women’s Health Study. Am J Clin Nutr.2006;83(5):1039–46.

24. Jacobs Jr DR, Andersen LF, Blomhoff R. Whole-grain consumption isassociated with a reduced risk of noncardiovascular, noncancer deathattributed to inflammatory diseases in the Iowa Women’s Health Study. AmJ Clin Nutr. 2007;85(6):1606–14.

25. Karlsen A, Paur I, Bohn SK, Sakhi AK, Borge GI, Serafini M, et al. Bilberry juicemodulates plasma concentration of NF-kappaB related inflammatorymarkers in subjects at increased risk of CVD. Eur J Nutr. 2010;49(6):345–55.

26. Kolberg M, Paur I, Balstad TR, Pedersen S, Jacobs Jr DR, Blomhoff R. Plantextracts of spices and coffee synergistically dampen nuclear factor-kappaBin U937 cells. Nutr Res. 2013;33(10):817–30.

27. Kolberg M, Pedersen S, Bastani NE, Carlsen H, Blomhoff R, Paur I. Tomato pastealters NF-kappaB and cancer-related mRNA expression in prostate cancer cells,xenografts, and xenograft microenvironment. Nutr Cancer. 2015;67(2):305–15.

28. Paur I, Austenaa LM, Blomhoff R. Extracts of dietary plants are efficientmodulators of nuclear factor kappa B. Food Chem Toxicol. 2008;46(4):1288–97.

29. Paur I, Balstad TR, Blomhoff R. Degree of roasting is the main determinantof the effects of coffee on NF-kappaB and EpRE. Free Radic Biol Med. 2010;48(9):1218–27.

30. Ostan R, Lanzarini C, Pini E, Scurti M, Vianello D, Bertarelli C, et al.Inflammaging and cancer: a challenge for the Mediterranean diet. Nutrients.2015;7(4):2589–621.

Henriksen et al. BMC Cancer (2017) 17:83 Page 15 of 17

Page 98: Malnutrition and dietary interventions in colorectal

31. Santoro A, Brigidi P, Gonos ES, Bohr VA, Franceschi C. Mediterranean dietand inflammaging in the elderly: the European project NU-AGE. Preface.Mech Ageing Dev. 2014;136-137:1–2.

32. Kostråd for å fremme folkehelsen og forebygge kroniske sykdommer:metodologi og vitenskapelig kunnskapsgrunnlag. Oslo: Nasjonalt råd forernæring, Helsedirektoratet; 2011

33. Bellera CA, Pulido M, Gourgou S, Collette L, Doussau A, Kramar A, et al.Protocol of the Definition for the Assessment of Time-to-event Endpoints inCANcer trials (DATECAN) project: formal consensus method for thedevelopment of guidelines for standardised time-to-event endpoints’definitions in cancer clinical trials. Eur J Cancer. 2013;49(4):769–81.

34. Punt CJ, Buyse M, Kohne CH, Hohenberger P, Labianca R, Schmoll HJ, et al.Endpoints in adjuvant treatment trials: a systematic review of the literaturein colon cancer and proposed definitions for future trials. J Natl Cancer Inst.2007;99(13):998–1003.

35. Schulz KF, Altman DG, Moher D. CONSORT 2010 statement: updatedguidelines for reporting parallel group randomized trials. Ann Intern Med.2010;152(11):726–32.

36. Edge SB. AJCC cancer staging handbook: from the AJCC cancer stagingmanual. New York: Springer; 2009.

37. Balstad TR, Carlsen H, Myhrstad MC, Kolberg M, Reiersen H, Gilen L, et al.Coffee, broccoli and spices are strong inducers of electrophile responseelement-dependent transcription in vitro and in vivo - studies in electrophileresponse element transgenic mice. Mol Nutr Food Res. 2011;55(2):185–97.

38. Blomhoff R, Carlsen MH, Andersen LF, Jacobs Jr DR. Health benefits of nuts:potential role of antioxidants. Br J Nutr. 2006;96 Suppl 2:S52–60.

39. Bohn SK, Myhrstad MC, Thoresen M, Holden M, Karlsen A, Tunheim SH, et al.Blood cell gene expression associated with cellular stress defense ismodulated by antioxidant-rich food in a randomised controlled clinical trialof male smokers. BMC Med. 2010;8:54.

40. Brevik A, Gaivao I, Medin T, Jorgenesen A, Piasek A, Elilasson J, et al.Supplementation of a western diet with golden kiwifruits (Actinidiachinensis var.‘Hort 16A’:) effects on biomarkers of oxidation damage andantioxidant protection. Nutr J. 2011;10:54.

41. Brevik A, Karlsen A, Azqueta A, Tirado AE, Blomhoff R, Collins A. Both baseexcision repair and nucleotide excision repair in humans are influenced bynutritional factors. Cell Biochem Funct. 2011;29(1):36–42.

42. Carlsen H, Myhrstad MC, Thoresen M, Moskaug JO, Blomhoff R. Berry intakeincreases the activity of the gamma-glutamylcysteine synthetase promoterin transgenic reporter mice. J Nutr. 2003;133(7):2137–40.

43. Carlsen MH, Halvorsen BL, Holte K, Bohn SK, Dragland S, Sampson L, et al.The total antioxidant content of more than 3100 foods, beverages, spices,herbs and supplements used worldwide. Nutr J. 2010;9:3.

44. Dragland S, Senoo H, Wake K, Holte K, Blomhoff R. Several culinary and medicinalherbs are important sources of dietary antioxidants. J Nutr. 2003;133(5):1286–90.

45. Halvorsen BL, Carlsen MH, Phillips KM, Bohn SK, Holte K, Jacobs Jr DR, et al.Content of redox-active compounds (ie, antioxidants) in foods consumed inthe United States. Am J Clin Nutr. 2006;84(1):95–135.

46. Halvorsen BL, Holte K, Myhrstad MC, Barikmo I, Hvattum E, Remberg SF,et al. A systematic screening of total antioxidants in dietary plants. J Nutr.2002;132(3):461–71.

47. Karlsen A, Retterstol L, Laake P, Paur I, Bohn SK, Sandvik L, et al.Anthocyanins inhibit nuclear factor-kappaB activation in monocytes andreduce plasma concentrations of pro-inflammatory mediators in healthyadults. J Nutr. 2007;137(8):1951–4.

48. Karlsen A, Svendsen M, Seljeflot I, Laake P, Duttaroy AK, Drevon CA, et al.Kiwifruit decreases blood pressure and whole-blood platelet aggregation inmale smokers. J Hum Hypertens. 2013;27(2):126–30.

49. Karlsen A, Svendsen M, Seljeflot I, Sommernes MA, Sexton J, Brevik A, et al.Compliance, tolerability and safety of two antioxidant-rich diets: arandomised controlled trial in male smokers. Br J Nutr. 2011;106(4):557–71.

50. Kolberg M, Pedersen S, Mitake M, Holm KL, Bohn SK, Blomhoff HK, et al:Coffee inhibits nuclear factor-kappa B in prostate cancer cells andxenografts. J Nutr Biochem. 2016;27:153–63.

51. Myhrstad MC, Carlsen H, Dahl LI, Ebihara K, Glemmestad L, Haffner K, et al.Bilberry extracts induce gene expression through the electrophile responseelement. Nutr Cancer. 2006;54(1):94–101.

52. Myhrstad MC, Carlsen H, Nordstrom O, Blomhoff R, Moskaug JO. Flavonoidsincrease the intracellular glutathione level by transactivation of the gamma-glutamylcysteine synthetase catalytical subunit promoter. Free Radic BiolMed. 2002;32(5):386–93.

53. Paur I, Balstad TR, Kolberg M, Pedersen MK, Austenaa LM, Jacobs Jr DR, et al.Extract of oregano, coffee, thyme, clove, and walnuts inhibits NF-kappaB inmonocytes and in transgenic reporter mice. Cancer Prev Res (Phila). 2010;3(5):653–63.

54. Qureshi SA, Lund AC, Veierod MB, Carlsen MH, Blomhoff R, Andersen LF, et al.Food items contributing most to variation in antioxidant intake; a cross-sectional study among Norwegian women. BMC Public Health. 2014;14:45.

55. Russnes KM, Wilson KM, Epstein MM, Kasperzyk JL, Stampfer MJ, Kenfield SA,et al. Total antioxidant intake in relation to prostate cancer incidence in theHealth Professionals Follow-Up Study. Int J Cancer. 2014;134(5):1156–65.

56. Svilaas A, Sakhi AK, Andersen LF, Svilaas T, Strom EC, Jacobs Jr DR, et al.Intakes of antioxidants in coffee, wine, and vegetables are correlated withplasma carotenoids in humans. J Nutr. 2004;134(3):562–7.

57. Bjelakovic G, Nikolova D, Gluud C. Antioxidant supplements and mortality.Curr Opin Clin Nutr Metab Care. 2014;17(1):40–4.

58. Ottery FD. Definition of standardized nutritional assessment andinterventional pathways in oncology. Nutrition. 1996;12(1 Suppl):S15–9.

59. Beck AM, Ovesen L, Schroll M. A six months’ prospective follow-up of 65+ -y-old patients from general practice classified according to nutritional riskby the Mini Nutritional Assessment. Eur J Clin Nutr. 2001;55(11):1028–33.

60. Flicker L, McCaul KA, Hankey GJ, Jamrozik K, Brown WJ, Byles JE, et al. Bodymass index and survival in men and women aged 70 to 75. J Am GeriatrSoc. 2010;58(2):234–41.

61. Miller WR, Rollnick S: Motivational interviewing : helping people change, 3rded. edn. New York: Guilford Publication; 2013

62. Nøkkelhullsmerket. http://www.nokkelhullsmerket.no/. Accessed 1 Dec 201563. Activ against cancer. http://aktivagainstcancer.org/. Accessed 1 Dec 201564. Carlsen MH, Karlsen A, Lillegaard IT, Gran JM, Drevon CA, Blomhoff R, et al.

Relative validity of fruit and vegetable intake estimated from an FFQ, usingcarotenoid and flavonoid biomarkers and the method of triads. Br J Nutr.2011;105(10):1530–8.

65. Carlsen MH, Lillegaard IT, Karlsen A, Blomhoff R, Drevon CA, Andersen LF.Evaluation of energy and dietary intake estimates from a food frequencyquestionnaire using independent energy expenditure measurement andweighed food records. Nutr J. 2010;9:37.

66. SenseWear Body Media. https://www.manualslib.com/manual/895732/Bodymedia-Sensewear.html.

67. Helseundersøkelsen i Nord-Trøndelag (HUNT). http://www.ntnu.edu/hunt/databank. Accessed 1 Dec 2015

68. Kern & Sohn. http://www.kern-sohn.com. Accessed 1 Dec 201569. Seca 285. http://www.meddevicedepot.com/PDFs/seca285.pdf. Accessed 1

Dec 201570. Nishida C, Ko GT, Kumanyika S. Body fat distribution and noncommunicable

diseases in populations: overview of the 2008 WHO Expert Consultation onWaist Circumference and Waist-Hip Ratio. Eur J Clin Nutr. 2010;64(1):2–5.

71. Seca mBCA515. http://www.seca.com/fileadmin/documents/product_sheet/seca_pst_515_en.pdf. Accessed 1 Dec 2015

72. Mourtzakis M, Prado CM, Lieffers JR, Reiman T, McCargar LJ, Baracos VE. Apractical and precise approach to quantification of body composition incancer patients using computed tomography images acquired duringroutine care. Appl Physiol Nutr Metab. 2008;33(5):997–1006.

73. GE Healtcare http://www3.gehealthcare.com. Accessed 1 Dec 201574. Bastani NE, Gundersen TE, Blomhoff R. Dried blood spot (DBS) sample

collection for determination of the oxidative stress biomarker 8-epi-PGF(2alpha) in humans using liquid chromatography/tandem massspectrometry. Rapid Commun Mass Spectrom. 2012;26(6):645–52.

75. Bingham S, Cummings JH. The use of 4-aminobenzoic acid as a marker tovalidate the completeness of 24 h urine collections in man. Clin Sci (Lond).1983;64(6):629–35.

76. Jakobsen J, Pedersen AN, Ovesen L. Para-aminobenzoic acid (PABA) used asa marker for completeness of 24 h urine: effects of age and dosagescheduling. Eur J Clin Nutr. 2003;57(1):138–42.

77. Nielsen SE, Freese R, Kleemola P, Mutanen M. Flavonoids in human urine asbiomarkers for intake of fruits and vegetables. Cancer Epidemiol BiomarkersPrev. 2002;11(5):459–66.

78. Naseribafrouei A, Hestad K, Avershina E, Sekelja M, Linlokken A, Wilson R,et al. Correlation between the human fecal microbiota and depression.Neurogastroenterol Motil. 2014;26(8):1155–62.

79. Bernstein R, Parkes JL, Goldy A, Brown D, Harrison B, Chu A, et al. A newtest strip technology platforn for self-monitoring of blood glucose. JDiabetes Sci Technol. 2013;7(5):1386-99.

Henriksen et al. BMC Cancer (2017) 17:83 Page 16 of 17

Page 99: Malnutrition and dietary interventions in colorectal

80. Ware JE SK KM, Gandek B. SF-36 Health Survey Manual & InterpretationGuide. Boston: New England Medical Center, The Health Institute; 1993.

81. Loge JH, Kaasa S. Short form 36 (SF-36) health survey: normative data fromthe general Norwegian population. Scand J Soc Med. 1998;26(4):250–8.

82. Chalder T, Berelowitz G, Pawlikowska T, Watts L, Wessely S, Wright D, et al.Development of a fatigue scale. J Psychosom Res. 1993;37(2):147–53.

83. Butler S, Chalder T, Ron M, Wessely S. Cognitive behaviour therapy inchronic fatigue syndrome. J Neurol Neurosurg Psychiatry. 1991;54(2):153–8.

84. Pawlikowska T, Chalder T, Hirsch SR, Wallace P, Wright DJ, Wessely SC.Population based study of fatigue and psychological distress. BMJ. 1994;308(6931):763–6.

85. Wessely S, Powell R. Fatigue syndromes: a comparison of chronic “postviral”fatigue with neuromuscular and affective disorders. J Neurol NeurosurgPsychiatry. 1989;52(8):940–8.

86. Common Terminology Criteria for Adverse Events (CTCAE). https://ctep.cancer.gov/protocolDevelopment/electrinic_applications/ctc.htm.

87. Birgisson H, Wallin U, Holmberg L, Glimelius B. Survival endpoints incolorectal cancer and the effect of second primary other cancer on diseasefree survival. BMC Cancer. 2011;11:438.

88. Fung TT, Kashambwa R, Sato K, Chiuve SE, Fuchs CS, Wu K, et al. Postdiagnosis diet quality and colorectal cancer survival in women. PLoS One.2014;9(12):e115377.

89. Kabat GC, Matthews CE, Kamensky V, Hollenbeck AR, Rohan TE. Adherence tocancer prevention guidelines and cancer incidence, cancer mortality, and totalmortality: a prospective cohort study. Am J Clin Nutr. 2015;101(3):558–69.

90. King DE, Mainous AG. 3rd, Geesey ME: Turning back the clock: adopting ahealthy lifestyle in middle age. Am J Med. 2007;120(7):598–603.

91. Olsen A, Egeberg R, Halkjaer J, Christensen J, Overvad K, Tjonneland A.Healthy aspects of the Nordic diet are related to lower total mortality. JNutr. 2011;141(4):639–44.

92. Trichopoulou A, Costacou T, Bamia C, Trichopoulos D. Adherence to aMediterranean diet and survival in a Greek population. N Engl J Med. 2003;348(26):2599–608.

93. Vergnaud AC, Romaguera D, Peeters PH, van Gils CH, Chan DS, Romieu I,et al. Adherence to the World Cancer Research Fund/American Institute forCancer Research guidelines and risk of death in Europe: results from theEuropean Prospective Investigation into Nutrition and Cancer cohortstudy1,4. Am J Clin Nutr. 2013;97(5):1107–20.

94. Schmid D, Leitzmann MF. Association between physical activity andmortality among breast cancer and colorectal cancer survivors: a systematicreview and meta-analysis. Ann Oncol. 2014;25(7):1293–311.

95. Hastert TA, Beresford SA, Sheppard L, White E. Adherence to the WCRF/AICRcancer prevention recommendations and cancer-specific mortality: resultsfrom the Vitamins and Lifestyle (VITAL) Study. Cancer Causes Control. 2014;25(5):541–52.

96. Balstad TR, Paur I, Poulsen M, Markowski J, Kolodziejczyk K, Dragsted LO,et al. Apple, cherry, and blackcurrant increases nuclear factor kappa Bactivation in liver of transgenic mice. Nutr Cancer. 2010;62(6):841–8.

97. Demark-Wahnefried W, Aziz NM, Rowland JH, Pinto BM. Riding the crest ofthe teachable moment: promoting long-term health after the diagnosis ofcancer. J Clin Oncol. 2005;23(24):5814–30.

98. Stull VB, Snyder DC, Demark-Wahnefried W. Lifestyle interventions in cancersurvivors: designing programs that meet the needs of this vulnerable andgrowing population. J Nutr. 2007;137(1 Suppl):243s–8s.

99. Davies NJ, Batehup L, Thomas R. The role of diet and physical activity inbreast, colorectal, and prostate cancer survivorship: a review of theliterature. Br J Cancer. 2011;105 Suppl 1:S52–73.

100. Campbell MK, Carr C, Devellis B, Switzer B, Biddle A, Amamoo MA, et al. Arandomized trial of tailoring and motivational interviewing to promote fruitand vegetable consumption for cancer prevention and control. Ann BehavMed. 2009;38(2):71–85.

101. Morey MC, Snyder DC, Sloane R, Cohen HJ, Peterson B, Hartman TJ, et al.Effects of home-based diet and exercise on functional outcomes amongolder, overweight long-term cancer survivors: RENEW: a randomizedcontrolled trial. Jama. 2009;301(18):1883–91.

102. Andre T, Boni C, Mounedji-Boudiaf L, Navarro M, Tabernero J, Hickish T,et al. Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment forcolon cancer. N Engl J Med. 2004;350(23):2343–51.

103. Courneya KS, McKenzie DC, Mackey JR, Gelmon K, Friedenreich CM, Yasui Y,et al. Effects of exercise dose and type during breast cancer chemotherapy:multicenter randomized trial. J Natl Cancer Inst. 2013;105(23):1821–32.

104. Friedenreich CM, Neilson HK, Woolcott CG, Wang Q, Stanczyk FZ, McTiernanA, et al. Inflammatory marker changes in a yearlong randomized exerciseintervention trial among postmenopausal women. Cancer Prev Res (Phila).2012;5(1):98–108.

105. Lynch BM, Courneya KS, Sethi P, Patrao TA, Hawkes AL. A randomizedcontrolled trial of a multiple health behavior change intervention deliveredto colorectal cancer survivors: effects on sedentary behavior. Cancer. 2014;120(17):2665–72.

• We accept pre-submission inquiries

• Our selector tool helps you to find the most relevant journal

• We provide round the clock customer support

• Convenient online submission

• Thorough peer review

• Inclusion in PubMed and all major indexing services

• Maximum visibility for your research

Submit your manuscript atwww.biomedcentral.com/submit

Submit your next manuscript to BioMed Central and we will help you at every step:

Henriksen et al. BMC Cancer (2017) 17:83 Page 17 of 17

Page 100: Malnutrition and dietary interventions in colorectal
Page 101: Malnutrition and dietary interventions in colorectal
Page 102: Malnutrition and dietary interventions in colorectal
Page 103: Malnutrition and dietary interventions in colorectal
Page 104: Malnutrition and dietary interventions in colorectal
Page 105: Malnutrition and dietary interventions in colorectal
Page 106: Malnutrition and dietary interventions in colorectal
Page 107: Malnutrition and dietary interventions in colorectal
Page 108: Malnutrition and dietary interventions in colorectal
Page 109: Malnutrition and dietary interventions in colorectal

Demographic information Assessment of dietary intake

Assessment of physical activity and function

Assessment of nutritional status

Anthropometric measurements

Body composition analysis

Blood pressure Biological samples

Oral glucose tolerance test Health status

Page 110: Malnutrition and dietary interventions in colorectal
Page 111: Malnutrition and dietary interventions in colorectal

II

Page 112: Malnutrition and dietary interventions in colorectal
Page 113: Malnutrition and dietary interventions in colorectal

The ability of PG-SGA to detect low fat-free mass in colorectal cancer patients

Hanna Ræderab, Christine Henriksena, Siv Kjølsrud Bøhna, Anne-Rikke O`de Fey

Vilboa, Hege Berg Henriksena, Ane Sørlie Kværnerab, Katrine Rolidab, Ingvild Paura,

Sigbjørn Smelandbc and Rune Blomhoffab

aDepartment of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norway bDivision of Cancer Medicine, Oslo University Hospital, Oslo, Norway cInstitute of Clinical Medicine, University of Oslo, Oslo, Norway

Corresponding author: Rune Blomhoff, Department of Nutrition, Faculty of Medicine, University

of Oslo, P.O. Box 1049 Blindern, 0316 Oslo, Norway. Telephone; +47 22851395

E-mail: [email protected]

Shortened version of the title: The ability of PG-SGA to detect low FFM

Keywords: Patient-Generated Subjective Global Assessment, fat-free mass, fat-free mass index,

colorectal cancer, sarcopenia

Page 114: Malnutrition and dietary interventions in colorectal

Abstract

Low fat-free mass (FFM) is associated with adverse outcomes in colorectal cancer (CRC) patients.

Patient-generated subjective global assessment (PG-SGA) is a widely used assessment tool

developed to detect patients at risk of malnutrition. The aim of this

study was to investigate the ability of PG-SGA to detect low FFM in patients with non-metastatic

CRC. Ninety-seven patients were included and categorized as well nourished (PG-SGA A, n=67) or

malnourished (PG-SGA B, n=30). Bioelectrical impedance analysis (BIA) was used to assess FFM.

Low FFM was defined as low fat-free mass index (FFMI) according to cut-off values recently

proposed by The European Society for Clinical Nutrition and Metabolism (ESPEN). Twenty-nine

percent of the patients were identified with low FFMI. The proportion with low FFMI was

significantly higher among patients classified as malnourished by PG-SGA compared to well

nourished (p=0.015). The sensitivity was however low, as the PG-SGA categorization classified

only 50.0 % of the patients with low FFMI as malnourished (PG-SGA B). Using the PG-SGA scores

(cut-off point > 4), the sensitivity increased to 60.7 %. Physical examination in the PG-SGA

identified only 64.3 % of the patients with low FFMI as muscle depleted. In conclusion, our results

indicate that the PG-SGA does not identify with sufficient sensitivity patients with low FFMI among

patients with non-metastatic CRC. In clinical practice, PG-SGA should be accompanied by muscle

mass assessments by BIA or other methods in order to detect low FFM in this patient group.

Page 115: Malnutrition and dietary interventions in colorectal

Introduction

Malnutrition and weight loss in cancer occurs due to a negative energy balance caused by a reduced

food intake in combination with metabolic alterations induced by the tumor, such as elevated

resting metabolic rate, lipolysis, and proteolysis driven by systemic inflammation and catabolic

factors1.

It is now recognized that in particular the loss of fat-free mass (FFM) is linked to adverse outcomes

in cancer patients. Progressive loss of FFM and skeletal muscle, the major constituent of FFM, is

shown to be an independent predictor of chemotherapy toxicity2, post-operative complications3 and

mortality4,5 in cancer patients. Depletion of FFM may occur with or without loss of fat mass, and

may therefore be masked by a stable body weight6. Furthermore, weight gain during recovery may

be characterized by an increase in fat mass rather than FFM7.

Loss of skeletal muscle mass may subsequently lead to sarcopenia, defined by the European

Working Group on Sarcopenia in Older People (EWGSOP) as “a syndrome characterized by

progressive and generalized loss of skeletal muscle mass and strength, which is associated with

adverse outcomes such as physical disability, poorer quality of life and death”8.

We have recently that low FFM is common in patients with non-metastatic CRC9. Low

FFM is shown to be associated with reduced survival in patients with non-metastatic primary

CRC10. For CRC patients, identification of low FFM and sarcopenia is therefore of clinical

importance since appropriate interventions may improve prognosis. Interventions focusing on

optimizing food intake and reducing nutritional impact symptoms (i.e. symptoms affecting food

intake) may decrease weight loss or facilitate weight gain in cancer patients11,12. Ravasco and

coworkers demonstrated that early individualized nutritional counselling reduced radiotherapy

toxicity and improved nutritional status, quality of life and survival in colorectal cancer (CRC)

patients receiving radiotherapy13. According to the European Society for Clinical Nutrition and

Page 116: Malnutrition and dietary interventions in colorectal

Metabolism (ESPEN) guidelines on nutrition in cancer patients, nutritional therapy should be

combined with physical therapy, i.e. counseling regarding physical activities of daily life, resistance

and aerobic exercise training, to maintain or increase muscle mass1.

ESPEN recently defined low FFM as FFM index (FFMI) below 15 kg/m2 and 17 kg/m2, in females

and males, respectively14. FFMI can be estimated by the use of different modalities, including air

displacement plethysmography, labeled water-isotope dilution techniques, dual energy x-ray

absorptiometry (DXA) and bioelectrical impedance analysis (BIA)15. In clinical practice, access to

these methods is limited. The Scored Patient-generated subjective global assessment (PG-SGA)16,17

is one of few comprehensive nutritional assessment tools used to identify malnutrition in cancer

patients and the method also includes an evaluation of body composition changes, i.e. loss of

subcutaneous fat and muscle mass. The examination consists of visual inspection and palpation of

muscles, subcutaneous fat and edema. The PG-SGA includes four patient-generated components

(weight history, food intake, nutritional impact symptoms and activities and function) and three

professional components (age and diagnosis, metabolic stress and physical examination). Based on

an evaluation of these components, patients are categorized as well-nourished (PG-SGA A),

moderately malnourished (PG-SGA B) or severely malnourished (PG-SGA C). The scored version

also includes numerical scores for each of the components as well as a total numerical score. PG-

SGA is recommended by the Academy of Nutrition and Dietetics as one of the nutritional

assessment tools to use in clinical oncology practice18. However, although PG-SGA includes an

evaluation of muscle and fat depletion, it is not known whether PG-SGA is suitable to detect low

FFM in cancer patients. The aim of this study was therefore to investigate the ability of PG-SGA to

detect low FFM in patients with non-metastatic CRC.

Page 117: Malnutrition and dietary interventions in colorectal

Subjects and methods

Patients

Patients were enrolled between August 2013 and March 2015. Eligible patients were women and

men aged 50 to 80 years with a confirmed primary CRC (ICD-10 18-20), and staged I-III according

to the tumor node staging (TNM) system19. Patients with distant metastases were not included. All

patients had undergone surgery at two Hospitals in Norway.

The patients included in this cross-sectional study were recruited from the ongoing randomized

clinical trial (RCT), The Norwegian Dietary Guidelines and Colorectal Cancer Survival (CRC-

NORDIET) study20. All measurements were performed prior to the diet intervention. The CRC-

NORDIET study was carried out in accordance to the Helsinki Declaration and informed consent

was obtained from all participants. The study was approved by the Regional Committees for

Medical and Health Research Ethics (REC Protocol Approval 2011/836) and by the data protection

officials in the hospitals, and registered on the National Institutes of Health Clinical Trials

(www.ClinicalTrials.gov; Identifier: NCT01570010).

Measurements

All measurements were conducted at start of the clinical trial (2-9 months post-surgery) and were

performed by trained personnel. The patients were instructed to fast overnight and until all

measurements were completed.

Nutritional assessment by the scored PG-SGA

The orwegian version of the scored PG-SGA (15-004 v10.13.16) was used in the present study,

and permission for use was given by the copyright holder of the instrument. The assessment was

carried out by trained registered clinical dietitians, and the scoring was controlled by one researcher

Page 118: Malnutrition and dietary interventions in colorectal

(H.R). Patients were classified as well-nourished (PG-SGA A), moderately malnourished (PG-SGA

B) or severely malnourished (PG-SGA C). In order to classify patients, emphasis was being placed

on weight loss, reduced food intake and loss of muscle mass detected by the physical examination.

If there was uncertainty regarding which category the patient should be classified within, the least

severe category was chosen. Each section of the PG-SGA was scored according to the manual16 and

a total PG-SGA score was calculated for each patient.

Total PG-SGA score in the range of 4-8 indicates need of an intervention supervised by a dietitian

targeting the reported symptoms, and total PG-SGA score 9 indicates a critical need of a

nutritional intervention16. The number of patients with scores

9 w therefore identified. The PG-SGA includes registration of current

body weight as well as body weight one month and six months prior to assessment. According to

the PG-SGA manual16, scoring of weight loss should preferably be based on weight history the last

month instead of the last six months. Weight loss was therefore calculated by subtracting the

current weight from the one-month weight.

The physical examination was performed according to the manual 16. Muscle wasting was

investigated by visual inspection and palpation of muscles with loss of bulk and tone in temporal

areas, deltoids and quadriceps indicating muscle depletion. The triceps and midaxillary line at the

level of the lower ribs were investigated with regard to depletion of subcutaneous fat. Ankles were

examined for the presence of edema. The degree of muscle and fat depletion was evaluated and

rated as “normal”, mild to moderate” or “severe deficit”.

Questions within the PG-SGA with several categories were merged into two or three categories.

The three food intake categories “normal”, “increased” and “reduced” were re-categorized into two

categories “normal/increased” and “reduced”. The 14 nutrition impact symptoms, were grouped

into “no symptom”,”1 symptom” and “2 or more symptoms”. The five categories within “Activity

Page 119: Malnutrition and dietary interventions in colorectal

and function” were collapsed into two categories, “normal” and “reduced”, with “normal”

corresponding to the first category; “normal with no limitations” and “reduced” corresponding to

the remaining four categories.

Body weight, height and body mass index (BMI)

Body weight was measured by the use of a non-slip Marsden M-420 Digital Portable Floor Scale

(Marshden, Rotherham, South Yorkshire, United Kingdom) or a digital wireless measuring station

for height and weight, Seca 285 (Seca, Birmingham, United Kingdom). Measurements were

performed with patients wearing light clothes and no shoes. Body weight was subtracted by 0.5 kg

to adjust for clothing. Height (cm) was measured post-surgery by the use of either a mechanical

height rod (Kern MSF- 200) or a digital wireless stadiometer (Seca 285). BMI was calculated based

on recorded weight and height.

BIA

To obtain FFM estimates, a single frequency whole-body BIA, BIA 101 (SMT Medical, Würzburg,

Germany) was used. BIA measures body composition indirectly by measuring the impedance (i.e

the resistance and reactance) of a low-voltage current passing through the body. FFM is then

calculated by the BIA software, which utilizes the impedance data in empiric regression equations

incorporated in the software. We have previously validated BIA against DXA in a subgroup of

CRC patients included in the CRC-NORDIET study9.

BIA was performed under standardized conditions according to the manufacturer`s protocol.

Measurements were performed by placing two skin electrodes on the right hand and two electrodes

Page 120: Malnutrition and dietary interventions in colorectal

on the right foot of the patient when lying in supine position. The device applies current of 400 μA

at a constant frequency of 50 kHz.

Determination of low FFMI and sarcopenia

FFM values from BIA were used to calculate FFMI (FFM (kg)/height (m2). FFMI was grouped into

“low FFMI” (<15 kg/m2 for women and < 17 kg/m2 for men) and “normal FFMI” ( 15 kg/m2 for

women and 17 kg/m2 for men) according to cut-off values for FFMI proposed as part of the new

diagnostic criteria for malnutrition by the ESPEN14.

Patients with sarcopenia were identified by the use of the diagnostic criteria for age-related

sarcopenia as proposed by EWGSOP8; presence of low muscle mass (criterion 1) and low muscle

function (strength (criterion 2) or performance (criterion 3)). Criterion 1, and either 2 or 3 must be

present to diagnose sarcopenia. In the current study, we defined low muscle mass as low FFMI.

Low muscle strength was defined as low hand grip strength according to the cut-off values

published by Fried21. Grip strength was assessed with a hand grip dynamometer (KERN & SOHN

GmbH, Balingen, Germany) as described in the manufacturer’s protocol. We defined low physical

performance as low gait speed and/or low number of sit to stands. Gait speed was measured with a

6-min walk test according to the guidelines from the American Thoracic Society22 and gait speed <

1 m/s was defined as “low”23. The sit-to-stand test was performed by instructing the participants to

sit on a chair with arms folded across their chest, and then to stand up and sit down as frequently as

possible within 30 s, keeping both arms folded across the chest. The number of full stands was

counted, and stands < 18 and < 22, i.e. the lower cut-off values for the 95 % CI for a reference

population of healthy Norwegian women and men in the age of 60 years, were defined as “low”24.

Page 121: Malnutrition and dietary interventions in colorectal

Statistical analyses

Determination of sample size was performed in accordance to a guide for sample size for sensitivity

and specificity analysis published by Bujang and Adnan in 201625. According to this guide, the

sensitivity for a screening study must be pre-determined to be at least 0.5025. We estimated the

prevalence of low FFM in CRC patients to be 33 %, based on our previous findings9 . Hence, a

minimum sample size of 67 patients would be needed to achieve a minimum power of 80 % in

order to detect a change in sensitivity from 0.50 to 0.80, based on a significance level of 0.05. Data

were checked for normality using the Kolmogorov-Smirnov test and visual inspection of the

histograms. Normally distributed data were presented as means and standard deviations, and non-

normally distributed data as medians and range (minimum-maximum). Pearson chi-square test for

independence or Fisher`s exact test was performed to investigate differences in proportions between

groups. Mann-Whitney test was used to test differences in medians for non-normally distributed

continuous variables. Independent samples t-test was used to explore differences in means for

normally distributed variables. P-values (2-sided) 0.05 were considered significant. Sensitivity

and specificity were calculated to evaluate PG-SGA as a screening tool with FFMI assessed by BIA

as reference method. All statistical analyses were performed using SPSS (IBM SPSS Statistic 22).

Page 122: Malnutrition and dietary interventions in colorectal

Results

Subject characteristics

One hundred and six patients were included in the study and assessed with the PG-SGA tool. Of

these, nine patients were excluded from the analyses due to lack of data needed to determine FFMI.

Of the 97 eligible patients, 28 patients (29 %) were identified with low FFMI. Subject

characteristics are shown for patients with low and normal FFMI, respectively (Table 1).

Fifty-nine percent of the patients had colon cancer, 35 % had rectum cancer and 7 % patients had

rectosigmoid cancer. The median time from CRC surgery to assessments was 4 months.

Patients with normal and low FFMI were compared with regard to clinical characteristics. In

general, there were few differences between the groups. There were no significant differences in

gender, cancer localization, TNM stage or proportions receiving neoadjuvant or adjuvant treatment

between the groups. Mean BMI was found to be significantly lower in patients with low FFMI, and

the proportion of underweight patients was significantly higher among patients with low FFMI

compared to patients with normal FFMI (p<0.001). Patients with low FFMI were significantly older

than patients with normal FFMI (p=0.027). This finding was expected since loss of FFM is

associated with increased age.

Page 123: Malnutrition and dietary interventions in colorectal

The ability of PG-SGA to detect patients with low FFMI

Based on the PG-SGA global assessment, 67 (69.1 %) and 30 (30.9 %) of 97 eligible patients were

categorized as well-nourished (PG-SGA A) and moderately malnourished (PG-SGA B),

respectively (Table 2). No patients were categorized as severely malnourished (PG-SGA C). The

proportion of patients with low FFMI estimated by BIA was significantly higher among patients

classified by PG-SGA as malnourished compared to well nourished (46.7 vs 20.9 %, p=0.015)

(Table 2). Furthermore, median PG-SGA total score was found to be significantly higher among

patients with low FFMI compared to patients with normal FFMI (5 vs 3, p=0.036). However, the

sensitivity, i.e. the proportion of patients with low FFMI classified as malnourished by PG-SGA

categories, was calculated to only 50.0 %. The specificity, i.e. the proportion of patients with

normal FFMI classified as well nourished by PG-SGA, was found to be 76.8 %. Using the PG-SGA

numerical score, 60.7 % of the patients with low FFMI were identified with score > 4, i.e. the

lowest cut-off for a nutritional intervention. These results indicate that the PG-SGA global rating

does not have sufficient sensitivity and specificity to detect low FFMI, however, using the PG-SGA

score increase the sensitivity.

The individual components of the PG-SGA in patients with low FFMI

In order to elucidate why a significant proportion of the patients with low FFMI (estimated by BIA)

was evaluated as well nourished by the PG-SGA, we investigated the individual components of the

assessment tool (Table 3). Regarding all patients with low FFMI independently of PG-SGA

categorization, 66.7 % of the patients reported weight loss within the last 6 months, whereas only

16.7 % reported weight loss the last month, indicating that the patients experienced their weight loss

earlier in the trajectory of the disease, and that the majority of the patients were weight increasing at

the time of assessment. Furthermore, 60.7 % reported a normal food intake (i.e. unchanged or

Page 124: Malnutrition and dietary interventions in colorectal

increased) the last month and 28.6 % had symptoms affecting food intake. Furthermore, 53.6 % of

the patients reported having reduced activity and function level (Table 3). The sensitivity of PG-

SGA examination to detect muscle mass depletion (i.e. visual inspection and palpation of muscles

in temporal areas, deltoids and quadriceps) was calculated. Only 64.3 % of the patients assessed

with low FFMI by BIA were evaluated as muscle depleted by PG-SGA. The specificity (i.e.

proportion of patients with normal FFMI correctly classified with “no deficit”) was 78 %. Taken

together, these findings suggest that when investigating the various components of the PG-SGA in

patients with low FFMI, the majority of these patients experienced weight increaseat the time of

assessments, accompanied by a normal food intake (i.e. stable or increased) and no symptoms

affecting food intake. Furthermore, the results indicate that the physical examination does not have

sufficient sensitivity and specificity to detect low FFMI.

Comparison of well nourished (PG-SGA A) and malnourished (PG-SGA B) patients among patients

with low FFMI

To further elucidate why a significant proportion of the patients with low FFMI was evaluated as

well nourished by the PG-SGA, we selected the patients with low FFMI and compared well

nourished and malnourished patients with regard to the individual components of the PG-SGA.

Patients categorized as PG-SGA A had significantly lower median total PG-SGA score compared to

patients categorized as PG-SGA B (3 vs 6, p<0.001) (Table 3). This finding was expected since

PG-SGA category is related to the PG-SGA score. Furthermore, none of the patients with PG-SGA

A reported a reduced food intake, whereas the majority of the patients categorized as PG-SGA B

reported reduced food intake (p<0.001). We found no differences between the groups with regard to

weight loss the last 6 months, weight loss the last month, presence of anorexia, presence of

nutritional impact symptoms or physical function and activity. Among patients identified with

Page 125: Malnutrition and dietary interventions in colorectal

“mild to moderate deficit” by the PG-SGA physical examination, a significantly lower proportion of

the patients were classified as PG-SGA A compared to PG-SGA B (27.8 vs 72.2 %, p=0.004). As

PG-SGA category was set mainly based on the three components weight loss the last month,

reduced food intake the last month and muscle mass depletion, these findings were quite expected,

except for weight loss that did not differ between the groups. The groups did not differ with regard

to BMI.

BMI according to physical examination status among patients with low FFMI

In order to investigate why a high proportion (i.e. 36 %) of the patients with low FFMI were not

detected by the physical exam in the PG-SGA, we investigated if there was a difference in BMI

between patients detected and patients not detected by the PG-SGA within patients with low FFMI

(Table 4). Mean BMI was significantly higher in patients not detected by the PG-SGA (24.6 vs

21.5, p=0.006). Furthermore, we found a significantly higher proportion of patients with overweight

among these patients compared to those that were found to be muscle depleted (66.7 vs 33.3 %,

p=0.025). A possible explanation for this finding may be that high BMI camouflages low muscle

mass in patients with low FFMI.

The ability of PG-SGA to detect patients with sarcopenia

In the current study, we also investigated the ability of PG-SGA to detect patients with sarcopenia.

Of 97 patients included in this study, 95 patients were eligible for the diagnosis of sarcopenia.

About twenty-two % (n= 21) of the patients were diagnosed with sarcopenia (Table 5). The

proportion of patients with sarcopenia did not significantly differ between patients classified by PG-

SGA as well nourished and malnourished, respectively. PG-SGA classified 42.9 % of the patients

Page 126: Malnutrition and dietary interventions in colorectal

with sarcopenia as malnourished. With regard to the PG-SGA numerical score, we found no

difference in median total score when we compared sarcopenic patients with non-sarcopenic

patients. Furthermore, 61.9 % of the patients with sarcopenia were identified with total PG-SGA

score > 4, i.e. the lowest cut-off for a nutritional intervention. These results were similar to the

results from the analysis of patients with low and normal FFMI. The sensitivity of PG-SGA to

detect patients with sarcopenia was low, however, we observed increased sensitivity by the use of

the PG-SGA scoring.

Page 127: Malnutrition and dietary interventions in colorectal

Discussion

In this study we investigated the ability of PG-SGA to identify patients with low FFM among

patients with non-metastatic CRC. About twenty-nine percent of the patients had low FFMI

according to the cut-off values proposed by ESPEN. The PG-SGA categorization classified only 50 %

of these patients as malnourished (PG-SGA B). Use of the PG-SGA total scores improved

sensitivity (61 %). However, only 64 % of the patients with low FFMI assessed by BIA were

evaluated as muscle depleted in the physical examination in the PG-SGA. Our results indicate that

the PG-SGA do not have sufficient sensitivity to detect low FFM.

Few previous studies have examined the ability of PG-SGA to detect low FFM, and to the best of

our knowledge, no studies are performed in non-metastatic patients with CRC. The clinical

implications of muscle depletion and sarcopenia is mainly studied in patients with metastatic cancer,

however, the high percentage of patients with low FFMI in our population suggests that it may have

a broader relevance9. Vigano and coworkers examined associations between PG-SGA scores and

features of cancer cachexia in a mixed population of patients with advanced lung and

gastrointestinal cancers. Although they observed that the PG-SGA score was able to predict several

features of cancer cachexia, including decrease of muscle strength and loss of fat mass, PG-SGA

was not able to detect differences in lean body mass26, in agreement with our results. In patients

with gynecologic cancers, FFM was not found to differ between PG-SGA categories27. In the study

performed by Guerra and colleges, FFMI was significantly lower among malnourished patients

according to the PG-SGA in a sample consisting of 455 inpatients with a broad spectrum of

diagnoses28.

Our study demonstrated poor specificity and sensitivity for the PG-SGA categories to detect low

FFMI. Only half of the patients with low FFMI were classified as malnourished. Consequently, half

Page 128: Malnutrition and dietary interventions in colorectal

of the patients were missed by the use of these categories. The literature on sensitivity and

specificity of PG-SGA to detect low FFM or muscle mass is scarce, however, our findings are in

line with the results reported by Elkan et al, who observed that SGA, the earlier version of PG-SGA,

showed poor sensitivity (46 %) in detection of low FFMI in patients with rheumatoid arthritis

assessed with DXA29. Similar to our data, they observed a higher specificity than sensitivity for

SGA.

In order to investigate why a significant proportion of the patients with low FFMI was categorized

as well nourished, we investigated the individual components of the PG-SGA. We observed that the

majority of the patients with low FFMI were anabolic at the time of assessments, reporting a normal

food intake and no symptoms affecting food intake. Since PG-SGA is developed to detect patients

with malnutrition or patients at risk of malnutrition with main focus on recent weight loss,

nutritional impact symptoms and reduction in food intake, the implication of this is that patients

with prior muscle mass depletion, but a stable or increasing body weight may be categorized as PG-

SGA A.

Moreover, when we analyzed differences between those who were categorized as well nourished

(PG-SGA A) and those who were categorized as malnourished (PG-SGA B) among patients with

low FFMI, we observed differences with regard to 1) food intake and 2) proportions detected by the

physical examination. A significantly higher proportion of the patients categorized as PG-SGA B

reported reduced food intake, and a significantly higher proportion of these patients were detected

with muscle mass depletion, compared to the patients categorized as PG-SGA A. Since reduced

food intake and muscle mass depletion constitute two of the three components that were

emphasized in the PG-SGA categorization, it provides a plausible explanation for why these

patients were categorized as PG-SGA B.

Page 129: Malnutrition and dietary interventions in colorectal

We observed that use of the PG-SGA total score improved sensitivity compared to PG-SGA

categories, suggesting that the scoring is better at capturing patients with low FFMI. It should,

however, be mentioned that the physical examination only in minor extent contributes to the total

PG-SGA score, with the maximum score of 3 points indicating severe adipose and muscle deficit.

Hence, patients with low FFMI may hypothetically have a low total score, i.e. a total score below

the lowest cut-off for an intervention.

Although a significantly higher proportion of the patients categorized as malnourished by PG-SGA

were detected as muscle depleted by the physical examination (i.e. visual inspection and palpation

of muscles in temporal areas, deltoids and quadriceps) compared to well nourished patients, the

sensitivity and specificity was found to be low. In order to elucidate why many patients were

missed by the physical examination in the PG-SGA, we hypothesized that muscle mass depletion

could be more difficult to detect in patients with high BMI. In the current study, BMI was found to

be significantly higher in those patients who were not identified as muscle depleted (as indicated by

loss of bulk and tone in selected muscles examined by visual inspection and palpation) by the PG-

SGA physical examination. Furthermore, we observed a higher proportion of overweight patients

among these patients compared to those who were captured as depleted. Based on these findings,

we conclude that PG-SGA is not sensitive enough to detect muscle mass depletion, particularly in

overweight and obese patients. Studies utilizing imaging analyses have confirmed that excessive

muscle wasting can be obscured in patients with excessive fat mass5,30, with computed tomography

(CT) images demonstrating equal low total muscle amounts in obese and underweight patients.

With a growing prevalence of overweight and obesity in several patient populations, including

cancer populations, it is important to be aware of this limitation in the application of PG-SGA.

Page 130: Malnutrition and dietary interventions in colorectal

Monitoring weight loss, nutritional impact symptoms and reduction in food intake are important

aspects of the nutritional assessments of cancer patients. However, since an ongoing loss of muscle

mass may be masked by a stable or increased body weight, particularly in overweight and obese

patients6, assessing and monitoring body weight and food intake is not sufficient. Increase of body

weight in terms of body fat rather than FFM may lead to sarcopenic obesity, a syndrome that entails

the combined health risks of both sarcopenia and obesity. This highlights the importance of

including appropriate tools to identify low FFM as part of the nutritional evaluation. As PG-SGA

seems not be sensitive enough to detect muscle mass depletion, we suggest that the tool should be

accompanied by muscle mass assessments by BIA or other methods.

In the current study we chose to use the FFMI cut-off values recently proposed by ESPEN, to

determine low FFM. Since these cut-offs were published in 2015, validation studies have confirmed

the prognostic impact of the malnutrition criteria on clinical outcomes31 and survival 32.

Our estimates of FFM were generated from BIA. Compared to imaging techniques such as DXA,

CT and magnetic resonance imaging (MRI) that measure lean body mass and muscle mass with

high precision, BIA measures these compartments indirectly by measuring the impedance of the

current applied to the body. The impedance data (i.e. resistance and reactance) is utilized in empiric

equations to calculate FFM. One of the main limitations with BIA is that these empirical equations

are developed in healthy euvolemic adults with a normal body composition, and may therefore

provide less reliable estimates in individuals with disturbances in fluids and alterations in body

composition, such as cancer patients. However, the BIA used in the current study was previously

validated against DXA in a subgroup of CRC patients included in the CRC-NORDIET study, and

use of the equation incorporated in the BIA software for calculation of FFM showed good

agreement with DXA estimates of FFM9.

Page 131: Malnutrition and dietary interventions in colorectal

Similar to the results from the analysis of patients with low FFMI, we observed low sensitivity of

the PG-SGA categories in detection of patients diagnosed with sarcopenia, and furthermore,

increased sensitivity by the use of the PG-SGA scores. Although PG-SGA is primarily developed to

identify patients with malnutrition and increased risk of malnutrition and not sarcopenia, our study

demonstrates that a high proportion of patients diagnosed with sarcopenia who need to be further

evaluated for nutritional therapy, are considered “no need for nutritional intervention” by the PG-

SGA.

Patients identified with low FFMI who have not fully developed sarcopenia, is particularly

interesting as target for nutritional intervention. According to the European Working Group on

Sarcopenia in Older People (EWGSOP), low muscle mass without the presence of reduced strength

or physical performance, corresponds to the stage “presarcopenia”. Identifying these patients and

selecting appropriate treatment, may prevent further loss of muscle mass and inhibit progressive

functional impairment.

Although PG-SGA does not perform sufficient sensitivity to detect low FFM, it covers several

important aspects of malnutrition and sarcopenia. Hence, PG-SGA may be useful to characterize

nutritional problems in patients where low FFM has been documented by the use of BIA or other

methodology. It rapidly provides a detailed overview of the patient`s nutritional status as the

assessment takes only approximately 5 minutes. Furthermore, the PG-SGA scoring may be useful in

the follow up of these patients, by using the scores to monitor changes during and after nutritional

therapy. In addition, PG-SGA score has been shown to predict clinical outcomes26, quality of life33

and survival26,34 in cancer patients.

To our knowledge, this is the first study that has evaluated the ability of the PG-SGA to identify

low FFM in colorectal cancer patients. Since PG-SGA is widely used and accepted as an assessment

tool in oncology it is important to be aware of its strengths and limitations.

Page 132: Malnutrition and dietary interventions in colorectal

Conclusion

The nutritional assessment tool PG-SGA classified only half of the patients with low FFMI as

malnourished. Use of the total PG-SGA score increased the sensitivity. However, only 64.3 % of

the patients with low FFMI were detected by the physical examination which is part of the PG-SGA.

In clinical practice, PG-SGA scores should be accompanied by muscle mass assessments by BIA or

other methods in order to more accurately identify low FFM in this patient group.

List of abbreviations

BIA: Bioelectrical impedance analysis; BMI: Body mass index; CRC: Colorectal cancer; CT:

Computed tomography; DXA: Dual energy x-ray absorptiometry; ESPEN: European Society for

Clinical Nutrition and Metabolism; EWGSOP: European Working Group on Sarcopenia in Older

People; FFM: Fat-free mass; FFMI: Fat-free mass index; ICD: International classification of

diseases and related health problems; MRI: Magnetic resonance imaging; PG-SGA: Patient-

generated subjective global assessment; RCT: Randomized clinical trial; TNM: Tumor node

metastasis.

Acknowledgements

We would like to thank the patients for their valuable contribution to this study. We would also like

to thank Magnhild Håskjold and Siv Åshild Billington for their contribution in data collection.

Finally, we acknowledge Kristine Lillebø Holm for assistance in the Norwegian translation of the

PG-SGA version used in this study.

Conflict of interest statement

R.B is a shareholder in the company Vitas AS. All other authors declare that they have no

competing interests.

Funding sources

Page 133: Malnutrition and dietary interventions in colorectal

This project has received funding from Throne Holst Foundation of Nutrition Research and South

Eastern Norway Regional Health Authority. These funding sources were not involved in the study

design, in the collection, analysis or the interpretation of data.

Page 134: Malnutrition and dietary interventions in colorectal

References

1. Arends J, Bachmann P, Baracos V, et al. ESPEN guidelines on nutrition in cancer patients. Clin Nutr. Feb 2017;36(1):11-48.

2. Prado CM, Baracos VE, McCargar LJ, et al. Sarcopenia as a determinant of chemotherapy toxicity and time to tumor progression in metastatic breast cancer patients receiving capecitabine treatment. Clin Cancer Res. Apr 15 2009;15(8):2920-2926.

3. Lieffers JR, Bathe OF, Fassbender K, Winget M, Baracos VE. Sarcopenia is associated with postoperative infection and delayed recovery from colorectal cancer resection surgery. Br J Cancer. Sep 04 2012;107(6):931-936.

4. Martin L, Birdsell L, Macdonald N, 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. Apr 20 2013;31(12):1539-1547.

5. Prado CM, Lieffers JR, McCargar LJ, 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. Jul 2008;9(7):629-635.

6. Prado CM, Siervo M, Mire E, et al. A population-based approach to define body-composition phenotypes. Am J Clin Nutr. Jun 2014;99(6):1369-1377.

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

8. Cruz-Jentoft AJ, Baeyens JP, Bauer JM, et al. Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People. Age Ageing. Jul 2010;39(4):412-423.

9. Raeder H, Kvaerner AS, Henriksen C, et al. Validity of bioelectrical impedance analysis in estimation of fat-free mass in colorectal cancer patients. Clin Nutr. Jan 12 2017.

10. Black D, Mackay C, Ramsay G, et al. Prognostic Value of Computed Tomography: Measured Parameters of Body Composition in Primary Operable Gastrointestinal Cancers. Ann Surg Oncol. Mar 21 2017.

11. Baldwin C, Spiro A, Ahern R, Emery PW. Oral nutritional interventions in malnourished patients with cancer: a systematic review and meta-analysis. J Natl Cancer Inst. Mar 07 2012;104(5):371-385.

12. Bourdel-Marchasson I, Blanc-Bisson C, Doussau A, et al. Nutritional advice in older patients at risk of malnutrition during treatment for chemotherapy: a two-year randomized controlled trial. PLoS One. 2014;9(9):e108687.

13. Ravasco P, Monteiro-Grillo I, Camilo M. 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. Dec 2012;96(6):1346-1353.

14. Cederholm T, Bosaeus I, Barazzoni R, et al. Diagnostic criteria for malnutrition - An ESPEN Consensus Statement. Clin Nutr. Jun 2015;34(3):335-340.

15. Prado CM, Heymsfield SB. Lean tissue imaging: a new era for nutritional assessment and intervention. JPEN J Parenter Enteral Nutr. Nov 2014;38(8):940-953.

16. Patient-Generated Subjective Global Assessment. . Accessed 23.05.2017. 17. Jager-Wittenaar H, Ottery FD. Assessing nutritional status in cancer: role of the Patient-

Generated Subjective Global Assessment. Current opinion in clinical nutrition and metabolic care. Sep 2017;20(5):322-329.

18. 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. Feb 2017;117(2):297-310.e247.

19. AJCC Cancer Staging. https://cancerstaging.org. Accessed 23 May 2017.

Page 135: Malnutrition and dietary interventions in colorectal

20. Henriksen HB, Raeder H, Bohn SK, et al. The Norwegian dietary guidelines and colorectal cancer survival (CRC-NORDIET) study: a food-based multicentre randomized controlled trial. BMC Cancer. Jan 30 2017;17(1):83.

21. Fried LP, Tangen CM, Walston J, et al. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci. Mar 2001;56(3):M146-156.

22. ATS statement: guidelines for the six-minute walk test. Am.J.Respir.Crit Care Med. 2002;166(1):111-117.

23. Cesari M, Kritchevsky SB, Newman AB, et al. Added value of physical performance measures in predicting adverse health-related events: results from the Health, Aging And Body Composition Study. J Am Geriatr Soc. Feb 2009;57(2):251-259.

24. Tveter AT, Dagfinrud H, Moseng T, Holm I. Health-related physical fitness measures: reference values and reference equations for use in clinical practice. Arch Phys Med Rehabil. Jul 2014;95(7):1366-1373.

25. Bujang MA, Adnan TH. Requirements for Minimum Sample Size for Sensitivity and Specificity Analysis. J Clin Diagn Res. Oct 2016;10(10):Ye01-ye06.

26. Vigano AL, di Tomasso J, Kilgour RD, et al. The abridged patient-generated subjective global assessment is a useful tool for early detection and characterization of cancer cachexia. J Acad Nutr Diet. Jul 2014;114(7):1088-1098.

27. Laky B, Janda M, Cleghorn G, Obermair A. Comparison of different nutritional assessments and body-composition measurements in detecting malnutrition among gynecologic cancer patients. Am J Clin Nutr. Jun 2008;87(6):1678-1685.

28. Guerra RS, Fonseca I, Sousa AS, Jesus A, Pichel F, Amaral TF. ESPEN diagnostic criteria for malnutrition - A validation study in hospitalized patients. Clin Nutr. Sep 08 2016.

29. Elkan AC, Engvall IL, Tengstrand B, Cederholm T, Hafstrom I. Malnutrition in women with rheumatoid arthritis is not revealed by clinical anthropometrical measurements or nutritional evaluation tools. Eur J Clin Nutr. Oct 2008;62(10):1239-1247.

30. Tan BH, Birdsell LA, Martin L, Baracos VE, Fearon KC. Sarcopenia in an overweight or obese patient is an adverse prognostic factor in pancreatic cancer. Clin Cancer Res. Nov 15 2009;15(22):6973-6979.

31. Sanz-Paris A, Gomez-Candela C, Martin-Palmero A, et al. Application of the new ESPEN definition of malnutrition in geriatric diabetic patients during hospitalization: A multicentric study. Clin Nutr. Dec 2016;35(6):1564-1567.

32. Rondel AL, Langius JA, de van der Schueren MA, Kruizenga HM. The new ESPEN diagnostic criteria for malnutrition predict overall survival in hospitalised patients. Clin Nutr. Nov 24 2016.

33. 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. Feb 2003;57(2):305-309.

34. Persson C, Sjoden PO, Glimelius B. The Swedish version of the patient-generated subjective global assessment of nutritional status: gastrointestinal vs urological cancers. Clin Nutr. Apr 1999;18(2):71-77.

Page 136: Malnutrition and dietary interventions in colorectal

Table1. Characteristics of the population

N Normal FFMI (n=69)

Low FFMI* (n=28)

pa

Age Mean (years) (std) 97 64.9 (8.2) 68.1 (5.3) 0.027 Gender, n (%) Women Men

46 51

28 (60.9) 41 (80.4)

18 (39.1) 10 (19.6)

0.058

Cancer localization, n (%) Colon cancer Rectosigmoid cancer Rectum cancer

54 6 32

36 (66.7) 6 (100) 23 (71.9)

18 (33.3) 0 (0) 9 (28.1)

0.264

TNM stage, n (%) Stage 1 Stage 2 Stage 3

10 46 33

8 (80.0) 36 (78.3) 20 (60.6)

2 (20.0) 10 (21.7) 13 (39.4)

0.199

Neoadjuvant treatment, n (%) No Yes Adjuvant treatment, n (%) None Ongoing Completed

81 14

74 16 5

59 (72.8) 9 (64.3)

55 (74.3) 10 (62.5) 3 (60.0)

22 (27.2) 5 (35.7)

19 (25.7) 6 (37.5) 2 (40.0)

0.531

0.512

BMI Mean (kg/m2) (std) BMI categories, n (%) Underweight (BMI < 20) Normal range (BMI 20-24,9) Overweight (BMI 25-29,9) Obese (BMI >30)

97

8 32 43 14

27.2 (4.3)

1 (12.5) 20 (62.5) 34 (79.1) 14 (100)

22.6 (3.0)

7 (87.5) 12 (37.5) 9 (20.9) 0 (0)

<0.001 <0.001

Abbreviations: FFMI, Fat-free mass index; TNM, Tumor node metastasis; BMI, Body mass index aIndependent samples t-test, chi-square test for independence or Fisher`s exact test, significance level p 0.05 *Low FFMI defined as FFMI < 17 kg/m2 for men and < 15 kg/m2 for women14.

Page 137: Malnutrition and dietary interventions in colorectal

Table 2. Global rating and PG-SGA scoring according to normal and low FFMI

N

Normal FFMI (n=69)

Low FFMI* (n=28) Pa

Global rating, n (%) Well nourished (A) Moderately malnourished (B) Severely malnourished (C)

67 30 0

53 (79.1) 16 (53.3) 0 (0)

14 (20.9) 14 (46.7) 0 (0)

0.015

Total PG-SGA score PG-SGA score < 4, n (%) PG-SGA score 4-8, n (%) PG-SGA score 9, (%) Median (range)

53 35 9 97

42 (79.2) 22 (62.9) 5 (55.6) 3 (1-17)

11 (20.8) 13 (37.1) 4 (44.4) 5 (1-20)

0.146

0.036 Abbreviations: PG-SGA, Patient-generated subjective global assessment; FFMI, Fat-free mass index aMann-Whitney test, chi-square test for independence or Fisher`s exact test, significance level p 0.05 *Low FFMI defined as FFMI < 17 kg/m2 for men and < 15 kg/m2 for women14.

Page 138: Malnutrition and dietary interventions in colorectal

Table 3. Comparison of PG-SGA A and PG-SGA B among patients with low FFM with regard to the various components of the PG-SGA and BMI

Patients with low FFMI*

N Patients with PG-SGA A (n=14)

Patients with PG-SGA B (n=14)

Pa

PG-SGA score, median (range) 28 3 (1-8) 6 (3-20) <0.001 Weight loss last 6 months, n (%) Yes No

18 9

8 (44.4) 6 (66.7)

10 (55.6) 3 (33.3)

0.420

Weight loss last month, n (%)Yes No

4 20

1 (25.0) 10 (50.0)

3 (75.0) 10 (50.0)

0.596

Presence of anorexia, n (%) Yes No

2 26

0 (0) 14 (53.8)

2 (100) 12 (46.2)

0.481

Food intake, n (%)Normal Reduced

17 11

14 (82.4) 0 (0)

3 (17.6) 11 (100)

<0.001

Symptoms, n (%) Yes No

8 20

2 (25.0) 12 (60.0)

6 (75.0) 8 (40.0)

0.209

Physical function and activity, n (%) Normal Reduced

13 15

9 (69.2) 5 (33.3)

4 (30.8) 10 (66.7)

0.058

Physical examination, n (%) No deficit Mild to moderate deficit Severe depletion

10 18 0

9 (90.0) 5 (27.8) 0 (0)

1 (10.0) 13 (72.2) 0 (0)

0.004

BMI, mean (std) 28 23.4 (3.0) 21.8 (3.0) 0.184 BMI categories, n (%) Underweight (BMI < 20) Normal range (BMI 20-24,9) Overweight (BMI 25-29,9) Obese (BMI >30)

7 12 9 0

3 (42.9) 5 (41.7) 6 (66.7) 0 (0)

4 (57.1) 7 (58.3) 3 (33.3) 0 (0)

0.623

Abbreviations: FFMI, Fat-free mass index; PG-SGA: Patient-generated subjective global assessment; BMI: Body mass index. aMann-Whitney test (PG-SGA score), independent samples t-test (BMI), chi-square test for independence or Fisher`s exact test significance level p 0.05 *Low FFMI defined as FFMI < 17 kg/m2 for men and < 15 kg/m2 for women14.

Page 139: Malnutrition and dietary interventions in colorectal

Table 4. BMI according to physical examination status in the PG-SGA among patients with low FFMI.

Low FFMI*

N No deficit (n=10) by the PG-SGA

Mild to moderate deficit (n=18) by the PG-SGA pa

BMI, mean (std) 28 24.6 (2.7) 21.5 (2.7) 0.006 BMI categories, n (%) BMI < 20 BMI 20-24,9 BMI 25-29,9 BMI >30

7 12 9 0

0 (0) 4 (33.3) 6 (66.7)) 0 (0)

7 (100) 8 (66.7) 3 (33.3) 0 (0)

0.025

Abbreviations: FFMI, Fat-free mass index; PG-SGA: Patient-generated subjective global assessment; BMI, Body mass index. aIndependent-samples t-test, chi-square test for independence or Fisher`s exact test, significance level p 0.05 *Low FFMI defined as FFMI <17 kg/m2 for men and < 15 kg/m2 for women14.

Table 5. Global rating and PG-SGA scoring according to sarcopenia and no sarcopenia

N

No sarcopenia (n=74)

Sarcopenia* (n=21) pa

Global rating, n (%) Well nourished (A) Moderately malnourished (B) Severely malnourished (C)

67 28 0

55 (82.1) 19 (67.9) 0 (0)

12 (17.9) 9 (32.1) 0 (0)

0.127

Total PG-SGA score PG-SGA score < 4, n (%) PG-SGA score 4-8, n (%) PG-SGA score 9, n (%) Median (range)

52 34 9 95

44 (84.6) 23 (67.6) 7 (77.8) 3 (1-17)

8 (15.4) 11 (32.4) 2 (22.2) 4 (1-20)

0.162

0.092 Abbreviations: PG-SGA: Patient-generated subjective global assessment. aMann-Whitney test, chi-square test for independence or Fisher`s exact test, significance level p 0.05 *Sarcopenia was diagnosed based on EWGSOP diagnostic criteria8.

Page 140: Malnutrition and dietary interventions in colorectal
Page 141: Malnutrition and dietary interventions in colorectal

III

Page 142: Malnutrition and dietary interventions in colorectal
Page 143: Malnutrition and dietary interventions in colorectal

Contents lists available at ScienceDirect

Clinical Nutrition 37 (2018) 292e300

Original article

Clinical Nutrition

journal homepage: http: / /www.elsevier .com/locate/c lnu

Validity of bioelectrical impedance analysis in estimation of fat-free

mass in colorectal cancer patients

Hanna Ræder a, b, 1, Ane Sørlie Kværner a, b, 1, Christine Henriksen a, Geir Florholmen a,Hege Berg Henriksen a, Siv Kjølsrud Bøhn a, Ingvild Paur a, Sigbjørn Smeland b, c,Rune Blomhoff a, b, *

a Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norwayb Division of Cancer Medicine, Oslo University Hospital, Oslo, Norwayc Institute of Clinical Medicine, University of Oslo, Oslo, Norway

a r t i c l e i n f o s u m m a r y

Article history:Received 13 July 2016Accepted 26 December 2016

Keywords:Bioelectrical impedance analysisDual-energy X-ray absorptiometryBody compositionFat-free massColorectal cancer

* Corresponding author. Department of Nutrition,of Oslo, P.O. Box 1049 Blindern, 0316 Oslo, Norway.

E-mail address: [email protected] (1 These authors contributed equally.

http://dx.doi.org/10.1016/j.clnu.2016.12.0280261-5614/© 2017 The Author(s). Published by Elsevi

Background & aims: Bioelectrical impedance analysis (BIA) is an accessible and cheap method to measurefat-free mass (FFM). However, BIA estimates are subject to uncertainty in patient populations withaltered body composition and hydration. The aim of the current study was to validate a whole-body anda segmental BIA device against dual-energy X-ray absorptiometry (DXA) in colorectal cancer (CRC) pa-tients, and to investigate the ability of different empiric equations for BIA to predict DXA FFM (FFMDXA).Methods: Forty-three non-metastatic CRC patients (aged 50e80 years) were enrolled in this study.Whole-body and segmental BIA FFM estimates (FFMwhole-bodyBIA, FFMsegmentalBIA) were calculated using14 empiric equations, including the equations from the manufacturers, before comparison to FFMDXA

estimates.Results: Strong linear relationships were observed between FFMBIA and FFMDXA estimates for all equa-tions (R2 ¼ 0.94e0.98 for both devices). However, there were large discrepancies in FFM estimatesdepending on the equations used with mean differences in the ranges �6.5e6.8 kg and �11.0e3.4 kg forwhole-body and segmental BIA, respectively. For whole-body BIA, 77% of BIA derived FFM estimateswere significantly different from FFMDXA, whereas for segmental BIA, 85% were significantly different. Forwhole-body BIA, the Schols* equation gave the highest agreement with FFMDXA with mean difference±SD of �0.16 ± 1.94 kg (p ¼ 0.582). The manufacturer's equation gave a small overestimation of FFM with1.46 ± 2.16 kg (p < 0.001) with a tendency towards proportional bias (r ¼ 0.28, p ¼ 0.066). For segmentalBIA, the Heitmann* equation gave the highest agreement with FFMDXA (0.17 ± 1.83 kg (p ¼ 0.546)). Usingthe manufacturer's equation, no difference in FFM estimates was observed (�0.34 ± 2.06 kg (p ¼ 0.292)),however, a clear proportional bias was detected (r ¼ 0.69, p < 0.001). Both devices demonstratedacceptable ability to detect low FFM compared to DXA using the optimal equation.Conclusion: In a population of non-metastatic CRC patients, mostly consisting of Caucasian adults andwith a wide range of body composition measures, both the whole-body BIA and segmental BIA deviceprovide FFM estimates that are comparable to FFMDXA on a group level when the appropriate equationsare applied. At the individual level (i.e. in clinical practice) BIA may be a valuable tool to identify patientswith low FFM as part of a malnutrition diagnosis.© 2017 The Author(s). Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND

license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

1. Introduction

Rapid loss of fat-free mass (FFM) and skeletal muscle, the major

Faculty of Medicine, University

R. Blomhoff).

er Ltd. This is an open access article u

constituent of FFM, has been shown to be an independent predictorof severe toxicity following cancer treatment [1], and to negativelyaffect efficacy of treatment [2] and survival [3,4]. Depletion of FFMmay be masked by a stable body weight or weight gain [5]. In

nder the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Page 144: Malnutrition and dietary interventions in colorectal

cancer patients experiencing treatment-related weight loss, weight 2. Methods

H. Ræder et al. / Clinical Nutrition 37 (2018) 292e300 293

gain during or after recoverymay be characterized by an increase infat mass (FM) rather than FFM [6]. Easy available instruments thatcan be used in the clinic to monitor loss of FFM are thereforeneeded.

Dual-energy X-ray absorptiometry (DXA) is an instrument thatallows for precise whole-body and regional determination of FFMby a low X-ray exposure [7]. Therefore DXA is considered one of thereference methods for measurement of body composition. Accessto DXA may, however be limited in clinical practice. Bioelectricalimpedance analysis (BIA) is a more easily available method for bodycomposition analysis as it is relatively cheap, provides rapid resultsand requires minimal operator training. BIA may therefore be auseful tool in clinical practice to identify patients with low FFM as apart of the diagnostic criteria for malnutrition [8]. BIA estimatesbody composition indirectly. A low-voltage current is passedthrough the body, whereby impedance (i.e. tissue resistance andreactance) is measured. Impedance data is then utilized in empiricequations to estimate body composition. Such equations have beendeveloped for different populations and incorporate impedancedata with variables such as height, weight, age and gender tocalculate FFM [9].

There are several types of BIA devices commercially available.Single-frequency BIA measures impedance at one frequency,usually 50 kHz, whereas multi-frequency BIA measures imped-ance at several frequencies. Moreover, BIA devices can be basedon a whole-body or a segmental approach. With the whole-bodyapproach the body is viewed as a cylindrical conductor with auniform cross-sectional area. This model is demonstrated to bevalid in healthy individuals with BMI within the range16.0e34 kg/m2, provided that hydration is normal and the BIAequation used is applicable to the population studied [10]. How-ever, since it does not take into account the differences inimpedance represented by the various body segments, e.g. thetrunk consisting of ~50% of the body weight and only contributingto 5e12% of the whole-body resistance, it has limited validity inpopulations with abnormal body composition [11]. Segmental BIAhas more recently been developed to overcome the in-consistencies between the resistance and body mass of the trunk.Additional research is however required to determine whetherthis model is better adapted at measuring body composition un-der these circumstances.

There are a large number of equations available in the literaturefor estimation of FFM. These equations havemostly been developedin healthy euvolemic adults with a normal body composition.Therefore, the equations may yield less reliable estimates in in-dividuals where these conditions are not met [10]. Patients withcolorectal cancer (CRC) are particularly interesting as they arevulnerable to fluid imbalance and alterations in body compositionpost-operatively and during chemotherapy and/or radiotherapy.CRC patients often experience symptoms such as anorexia, vomit-ing, diarrhoea and obstipation as a result of treatment. This mayadversely affect weight status as well as influence water and elec-trolyte balance. Furthermore, obesity and abdominal obesity aremain risk factors for CRC [12] and thus, many CRC patients will haveexcess body weight at the time of diagnosis.

Few studies have tested the ability of BIA to estimate FFM incancer patients using DXA as a reference method, none of whichhas simultaneously compared two different BIA devices with DXA.Hence, the aim of this study was to validate two different BIA de-vices, a whole body BIA and a segmental BIA device, against DXA inCRC patients, and to investigate the ability of 14 different empiricequations, including the equations from the manufacturers, topredict DXA FFM (FFMDXA).

2.1. Patients

All patients in this validation study were recruited from anongoing randomized clinical trial, the Norwegian Dietary Guide-lines and Colorectal Cancer Survival (CRC-NORDIET) study. TheCRC-NORDIET study is carried out in accordance to the HelsinkiDeclaration and informed consent was obtained from all partici-pants. The study was approved by the Regional Committees forMedical and Health Research Ethics (REC Protocol Approval 2011/836) and by the data protection officials at Oslo University Hospitaland Akershus University Hospital. The study is registered on theNational Institutes of Health Clinical Trials (www.ClinicalTrials.gov;Identifier: NCT01570010).

Eligible patients were women and men aged 50e80, with aconfirmed CRC (ICD-10 18-20), and staged I-III according to theTNM staging system [13] when they entered the CRC-NORDIETstudy. Patients with pacemakers were excluded since currentfrom the BIA device could possibly alter the pacemaker activity. Toincrease generalizability, we chose to include patients with ab-normalities in body shape (for example amputations), obesity, or-thopedic prosthesis/implants, chronic diseases and fluiddisturbances (presence of oedema). All patients had undergonesurgery for CRC within the last 4 years.

2.2. Measurements

All measurements took place between December 1st 2015 andFebruary 1st 2016 at the Department of Nutrition, University ofOslo. The patients were instructed to fast overnight and until allmeasurements were completed. They were also encouraged to voidtheir bladders before measurements. For each patient, all mea-surements were conducted in the morning in a sequential mannerwithin a timeframe of 2 h.

2.3. BIA

BIA measures body composition indirectly by measuring theimpedance of a low-voltage current passing through the body. Theimpedance consists of two components, resistance (R), the oppo-sition of an ionic solution in both intra- and extracellular spaces andreactance (Xc), representing the capacitance from cell membranes[9]. Estimates of various body compartments, including FFM, arecalculated from R or R and Xc values, based on equations, eitherincorporated in the software or reported in the literature.

Two different BIA devices were used, one whole-body single-frequency (50 KHz) BIA, BIA-101 (SMT Medical, Würzburg, Ger-many), hereby referred to as whole-body BIA, and a multi-frequency segmental BIA, Seca mBCA515 (Seca, Birmingham,United Kingdom), hereby referred to as segmental BIA. For bothinstruments, BIA was performed under standardized conditionsaccording to the manufacturer's protocol. All measurements wereperformed with light clothing and with metal objects (e.g. jewelry,keys) being removed.

The whole-body BIA measurements were performed by placingtwo adhesive single-use skin electrodes (purchased from MaltronInternational Ltd, UK) on the right hand and foot, respectively, onthe patient when lying in supine position. The device applies acurrent of 400 mA at constant frequency of 50 kHz.

The segmental BIA measurements were performed on patientsstanding barefoot on the instrument platform. The device has anintegrated scale and uses four pair of electrodes of stainless steelthat are positioned at each hand and foot, through which the

Page 145: Malnutrition and dietary interventions in colorectal

current enters the limbs. The device enables segmental impedance Differences in raw data (R, Xc and phase angle (PhA)) and FFM

H. Ræder et al. / Clinical Nutrition 37 (2018) 292e300294

measurements of the right arm, left arm, trunk, right leg, left legand the right and left body side. A current of 100 mA is applied atfrequencies of 1, 1.5, 2, 3, 5, 7.5, 10, 15, 20, 30, 50, 75, 100, 200, 300,500, 750 and 1000 kHz. In the current study, measurements at50 kHz were utilized.

For estimation of FFM, a selection of equations was used(Tables 3A and B). These included the equations incorporated intothe manufacturers software, The Kyle (“Geneva”) equation [14]recommended by the European Society for Clinical Nutrition andMetabolism (ESPEN) and equations previously tested in cancerpopulations, see Appendix 1 [14e23]. Of the 14 equations used, 11were originally developed in healthy volunteers [14e18,20e22,24].The equations varied with regards to the predictor variablesincluded (e.g. R, Xc, weight, height, age and gender) and whetherthey were gender-specific (indicated by “*”) or not in addition tothe empirically obtained parameter estimates (intercept andregression coefficients).

2.4. DXA

Total body DXA scans were performed using GE Lunar iDXAenCORE version 16 (GE Healthcare). All scans were performed usingautomated mode by a trained GE Lunar iDXA operator. Patientswere instructed to wear a hospital gown and all jewelry wasremoved. Regions of interest were automatically detected by thesoftware and verified by the operator according to instructionsprovided by the Lunar enCORE operator manual.

2.5. Body weight, height, body mass index (BMI) and fat-free massindex (FFMI)

Bodyweight wasmeasuredwith light clothes andwithout shoesby the use of the incorporated scale in the body compositionanalyzer Seca mBCA515. This measured body weight was applied inboth BIA devices. DXA assessment was based on the weightrecorded by the DXA software according to standard procedure inthe manufacturer's manual. Height was measured by the use of adigital wireless stadiometer, Seca 285 (Seca, Birmingham, UnitedKingdom), and recorded in cm.

BMI (kg/m2) was calculated based on the recorded weight andheight. FFM adjusted for height, FFMI (kg/m2), was calculated bothfor BIA (FFMBIA) and DXA (FFMDXA). To detect low FFMI, thefollowing cut-off values were used: 15 kg/m2 for women and 17 kg/m2 for men [8].

2.6. Nutritional status

Nutritional status was assessed with the Patient-GeneratedSubjective Global Assessment (PG-SGA). This is a nutritionalassessment tool, specifically designed to identify malnutrition orrisk of malnutrition in cancer patients [25]. The assessment wascarried out by trained registered clinical dietitians, and the scoringwas controlled by one researcher (H.R). Patients were classified aseither well-nourished, moderately malnourished or severelymalnourished [26].

The PG-SGA form also provides information on status withregards to problems related to ascites, ankle oedema, vomiting anddiarrhoea, all of which can affect the BIA raw data.

2.7. Statistical analyses

Descriptive statistics are given as median (interquartile range(Q1eQ3)) for continuous variables and number (n) (%) for cate-gorical variables.

estimates between the various devices were tested for normality bythe ShapiroeWilk normality test and visual inspection ofhistograms.

To compare raw data assessed by the two BIA devices, pairedsample t-tests and bivariate (Pearson's) correlation analysis wasperformed.

FFM estimates derived from equations in the BIA softwares(hereby referred to as manufacturer's equations) as well as selectedpreviously published equations (see Appendix 1) were comparedwith DXA estimates using paired sample t-test and linear regres-sion. To determine the most suitable equations, the following as-pects of validity were considered: 1) Equality of means, determinedby non-significant difference between mean FFMBIA and FFMDXAusing paired sample t-tests, and 2) Ability of FFMBIA to predictFFMDXA, determined by high coefficients of determination (R2) andlow prediction error (SEE) using linear regression models. Theseequations, as well as the manufacturer's equations were testedfurther for validity by constructing scatter plots, BlandeAltmanplots and by calculating Lin's concordance correlation coefficient(CCC). BlandeAltman plots were constructed to diagnose eventualbias (estimated by mean differences), limits of agreement (meandifference ± 1.96 SD) and presence of outliers in the data. Propor-tional bias (e.g. variation in the vertical spread of scatter points withincreasing value of FFM) was assessed by analysing the Pearson'scorrelation coefficient between mean of FFMBIA and FFMDXA anddifferences in FFMBIA and FFMDXA. CCC, which takes both equality ofmeans and strength of linear relationship into consideration, was

calculated using the following formula: CCC ¼ 2rsxsy

s2xþs2

yþðmx� my Þ2 ,

where r is the correlation coefficient r between the twomethods, mxand my are the means for the two methods and s2x and s2yare thecorresponding variances.

For the most suitable BIA equations, as well as the manufac-turer's equations, sensitivity and specificity for identification of lowand normal FFMI was calculated using DXA as reference method[27]. Low FFMI was defined as <17 kg/m2 for men and <15 kg/m2

for women.SPSS 21.0 for Windows (SPSS, Chicago, IL, USA) was used for all

statistical analyses. Statistical significance was defined as p < 0.05.

3. Results

3.1. Characteristics of the study population

All the 45 eligible patients completed the DXA scan, while 43 and41 completed the whole-body BIA and segmental BIA assessment,respectively. Two participants were excluded from BIA measure-ments due to pacemakers. Furthermore, 2 patients were not able toperform the segmental BIA measurement due to upper extremityamputation and technical issues during the assessment, respectively.

Subject characteristics are shown in Table 1. Median (inter-quartile range (Q1eQ3)) time from surgery to assessment was 293(190, 500) days. The participants had a wide range of BMI (18e43)kg/m2, FFM (33e72) kg, FM (11e63) kg and visceral adipose tissue(72e2734) g assessed by DXA. Thirty-nine percent of the womenand 41% of the men were classified as abdominally obese (�88 cmfor women and �102 cm for men). Seventeen percent were clas-sified as moderately malnourished according to the PG-SGAassessment tool. None of the patients were classified as severelymalnourished. Regarding factors possibly contributing to alteredwater- and electrolyte balance, 12% had clinically visible ankleoedema, 18% had an ileostomy or a colonostomy and 29% haddiarrhoea. None of the patients had ascites.

Page 146: Malnutrition and dietary interventions in colorectal

Mean difference ± SD between body weight assessed by DXAand by the weight scale used for BIA assessments (BIAeDXA) were

indicator of functional and nutritional status, was significantlyhigher for whole-body BIA compared to segmental BIA, with mean

Table 1Characteristics of the study population. Numbers are median (interquartile range (Q1eQ3)).

Variables All subjects (n ¼ 43)a Women (n ¼ 26)a Men (n ¼ 17)a

Age, years 67.0 (62.0, 71.5) 63.5 (58.0, 71.0) 69.0 (65.0, 72.0)RaceWhite, n (%) 41 (97.6) 25 (100.0) 16 (94.1)Others, n (%) 1 (2.4) 0 (0.0) 1 (5.9)

OstomyYes, % 7 (18.4) 2 (8.7) 5 (33.3)

Time since surgery, days 293 (189.5, 499.5) 316.0 (229.0, 550.0) 278.0 (152.0, 441.0)Nutritional statusAnthropometric measuresHeight, cm 169.3 (164.5, 175.8) 165.2 (162.4, 169.3) 175.9 (170.6, 180.7)Body weight, kgb 72.0 (60.3, 84.8) 61.6 (54.0, 80.5) 84.5 (71.4, 89.8)Body weight (DXA), kg 71.7 (60.3, 84.8)) 61.7 (54.1, 80.7) 83.9 (71.7, 89.9)BMI, kg/m2b 25.8 (22.0, 28.9) 24.8 (20.7, 28.4) 27.5 (23.1, 29.5)Waist circumference, cm 93.3 (80.6, 101.7) 83.7 (74.9, 95.6) 101.0 (93.3, 104.1)Hip circumference, cm 97.3 (90.0, 104.0) 97.1 (89.2, 104.9) 98.6 (94.0, 103.0)Waist/hip-ratio 0.90 (0.84, 0.99) 0.87 (0.82, 0.90) 1.01 (0.98, 1.03)PG-SGAGlobal ratingWell nourished, n (%) 35 (83.3) 20 (80.0) 15 (88.2)Moderately malnourished, n (%) 7 (16.7) 5 (20.0) 2 (11.8)

Total scoring, sum 3.0 (2.0, 4.0) 3.0 (2.0, 4.0) 3.0 (2.0, 4.0)AscitesYes, n (%) 0 (0.0) 0 (0.0) 0 (0.0)

Ankle oedemaYes, n (%) 5 (11.9) 4 (16.0) 1 (5.9)

VomitingYes, n (%) 0 (0.0) 0 (0.0) 0 (0.0)

DiarrhoeaYes, n (%) 12 (28.6) 8 (32.0) 4 (23.5)

Body composition (DXA)FFM, kg 47.6 (38.9, 55.1) 40.5 (37.8, 45.4) 60.1 (52.9, 62.2)FM, kg 23.7 (17.1, 33.0) 22.7 (15.8, 35.3) 25.9 (18.5, 29.5)BM, kg 2.5 (2.0, 3.0) 2.2 (1.9, 2.4) 3.1 (2.9, 3.3)VATM, g 988.0 (477.0, 2003.0) 695.0 (215.0, 1298.0) 1949 (906.0, 2381.0)

Abbreviations: Q, Quartile; BMI, Body mass index; PG-SGA, Patient-generated Subjective global assessment; DXA, Dual energy X-ray absorptiometry; FFM, Fat-free mass; FM,Fat mass; BM, bone mass; VATM, Visceral adipose tissue mass.

a Numbers may vary due to missing information.b Body weight assessed by Seca mBCA515.

H. Ræder et al. / Clinical Nutrition 37 (2018) 292e300 295

0.3 kg ± 1.0 (p ¼ 0.046), indicating a slight overestimation of bodyweight using the body composition analyser Seca mBCA515compared to DXA.

3.2. Comparison of raw data between the two BIA devices

As raw data, i.e. the resistance and the reactance values (R andXc, respectively), are important determinants of FFM estimates, wecompared R and Xc values generated by the two BIA-devices at50 kHz. We observed significant differences in both R and Xc valuesbetweenwhole-body BIA and segmental BIA. Whole-body BIA gavelower mean R values compared to segmental BIA, 547 and622 Ohm, respectively, and higher mean Xc values, 61 and 49 Ohm,respectively (Table 2). Phase angle ((PhA), i.e. (Xc/R)� (180�/p)), an

Table 2Comparison of resistance (R), reactance (Xc) and phase angle (PA) assessed bywhole-body BIA and segmental BIA.

Variable Whole-body BIA Segmental BIA Correlations

Mean (SD) Mean (SD) P1 r P2

R, Ohm 546.9 (91.3) 622.2 (102.1) <0.001 0.99 <0.001Xc, Ohm 60.6 (13.3) 49.3 (6.9) <0.001 0.58 <0.001PhA 6.4 (1.3) 4.6 (0.6) <0.001 0.57 <0.001

Abbreviations: SD, Standard deviation; r, Correlation coefficient (Spearman's); R,Resistance; Xc, Reactance; PhA, Phase angle.P-value: p1; Paired sample t-tests, p2; test for bivariate correlations (Pearson's).

PhA values of 6.4 and 4.6, respectively.

3.3. Validity of whole-body BIA to assess FFM

Whole-body BIA FFM estimates (FFMBIA-whole-body) were calcu-lated using different empiric equations including the equationderived from the manufacturer's software. These estimates werethen compared with DXA estimates.

There was a high degree of linear relationship between BIA andDXA estimates for all equations tested with R2 ranging from 0.94 to0.97 (Table 3A). However, large discrepancies in FFM estimateswere observed depending on the equations used, with mean dif-ferences ranging from �6.5 to 6.8 kg (Table 3A, Fig. 1A). Only threeequations produced similar mean FFM values compared to DXA (i.e.not significantly different by paired t-tests). Those were the Gray*,Schols* and Segal* equations (Table 3A). Of these equations, theSchols* and Gray* equations demonstrated lower SEE (2.0 and1.9 kg, respectively) than the Segal* equation (2.4 kg), and werehence considered more suitable.

The manufacturer, Gray* and Schols* equations were testedfurther for validity by constructing scatter plots (Fig. 2AeC),BlandeAltman plots (Fig. 2DeF) and by calculating CCC. Visual in-spection of the scatter plots clearly revealed how FFM estimates co-varied with the corresponding DXA estimates for all equations.However, the BlandeAltman plots showed that only the Schols*equation gave satisfactory agreement with FFMDXA, indicated by no

Page 147: Malnutrition and dietary interventions in colorectal

constant bias (i.e. fixed bias), no proportional bias and small limits

shown). Taken together, these results suggest that the Heitmann*equation may be the superior equation for estimation of FFM usingsegmental BIA.

Table 3AEvaluation of fat-free mass (FFM) measured by whole-body BIA against DXA asreference method. A comparison of different equations. N ¼ 43a.

Variable Meandifference

SD 95% CI P R2 SEE

Manufacturer, kg 1.46 2.16 0.80, 2.13 <0.001 0.96 2.03Heitmann* [18,28], kg 2.41 1.75 1.87, 2.95 <0.001 0.97 1.76Gray* [17], kg 0.36 2.99 �0.56, 1.28 0.437 0.97 1.87Heitmann [18,28], kg 1.38 1.91 0.79, 1.97 <0.001 0.97 1.92Schols* [23], kg �0.16 1.94 �0.76, 0.43 0.582 0.96 1.96Geneva [14], kg 2.66 2.42 1.92, 3.41 <0.001 0.96 2.18Deurenberg (1991) [9,16], kg �2.38 2.59 �3.18, �1.59 <0.001 0.95 2.29Kyle [19], kg �2.54 2.76 �3.39, �1.69 <0.001 0.95 2.30Lukaski [21,28], kg 2.37 2.42 1.62, 3.11 <0.001 0.95 2.33Lukaski* [21], kg 1.78 2.61 0.97, 2.58 <0.001 0.95 2.34Lohman* [20,32], kg 6.81 3.27 5.80, 7.82 <0.001 0.95 2.37Segal* [22], kg 0.40 2.37 �0.33, 1.13 0.272 0.95 2.39Deurenberg (1990) [15], kg �6.54 2.47 �7.29, �5.78 <0.001 0.94 2.49

Abbreviations: SD, Standard Deviation; 95% CI, 95% Confidence interval; R2, Rsquared; SEE, Standard Error of the Estimate.P-values: Paired sample T-test.

a Equations included: 1) Manufacturer's equation, 2) The Geneva equation (rec-ommended by ESPEN) and 3) Equations previously tested for validity in cancerpatients. * Indicate sex-specific equations.

Fig. 1. Error bars showing mean difference ±1.96 standard deviations (SD) betweenfat-free mass (FFM) measured by whole-body BIA and segmental BIA using variousequations and FFM measured by DXA.

H. Ræder et al. / Clinical Nutrition 37 (2018) 292e300296

of agreement (�3.96, 3.64 kg). For the manufacturer's equation,there was a small fixed bias of 1.46 (�2.77, 5.69 kg) with a tendencytowards proportional bias (r ¼ 0.28, p ¼ 0.066). For the Gray*equation, wide limits of agreement was observed (�5.50, 6.22 kg),as well as a clear proportional bias (r ¼ 0.73, p < 0.001), with un-derestimation and overestimation for low and high values of FFM,respectively. CCC values (i.e. a combined measure of equality ofmeans and strength of linear relationship), were within the range0.95e0.99 for all three equations (data not shown), which isconsidered substantial. The CCC value was higher for the Schols*equation (CCC ¼ 0.98) than the manufacturer's and Gray* equation(CCC of 0.97 for both). Taken together, these results suggest that theSchols* equation may be superior to the other equations for esti-mation of FFM using whole-body BIA.

3.4. Validity of segmental BIA to assess FFM

Similar to whole-body BIA, segmental BIA FFM estimates(FFMBIA-segmental) were calculated using the different empiricequations including the equation derived from the manufacturer'ssoftware, and compared with DXA estimates.

As for whole-body BIA, all equations showed strong linearagreement with DXA estimates with R2 ranging from 0.94 to 0.98(Table 3B) but large variations in FFM estimates depending on theequation used with mean differences ranging from �11.0 to 3.4 kg(Table 3B, Fig. 1B). Only two equations gave similar FFM estimatesas FFMDXA, the manufacturer's and the Heitmann* equations(Table 3B).

The manufacturer and Heitmann* equations were tested furtherfor validity by constructing scatter plots (Fig. 3AeB), BlandeAltmanplots (Fig. 3CeD) and by calculating CCC. The scatter plotsdemonstrated how FFM estimates co-varied with the correspond-ing FFMDXA estimates for both equations. Furthermore, inspectionof the BlandeAltman plots showed no fixed bias for any of theequations with limits of agreement ranging from �4.38 to 3.70 kgand �3.42 to 3.76 kg for the manufacturer's and Heitmann* equa-tions, respectively. Whereas no proportional bias was seen for theHeitmann* equation, a clear positive association was observed forthe manufacturer's equation (r ¼ 0.69, p < 0.001) with underesti-mation and overestimation for low and high values of FFM,respectively. CCC values were 0.98 for both equations (data not

3.5. Use of BIA-derived FFM estimates to diagnose malnutrition

The type of BIA device and equation used to assess FFM mayhave clinical implication for the diagnosis of malnutrition. Ac-cording to the new consensus statement from ESPEN [8], malnu-trition can be diagnosed by either low BMI alone or a combinationof unintentional weight loss (mandatory) and low BMI or low FFMI.Using the cut-off values for FFMI, 33% of our patients were identi-fied with low FFMI based on FFMDXA (Table 4). Whole-body BIAwith use of the manufacturer's equation resulted in the lowestproportion of patients with low FFMI (26%) compared to segmentalBIA using the manufacturer's equation resulting in the highest

Page 148: Malnutrition and dietary interventions in colorectal

proportion (44%). Considering DXA as a reference method to assessFFMI, the highest sensitivity (i.e. proportion with low FFMIcorrectly identified as such) was seen for whole-body BIA using the

4. Discussion

To our knowledge, this is the first study to test the validity of two

Fig. 2. Fat-free mass (FFM) assessed by DXA and whole-body BIA. AeC: Scatter plots for; A) Manufacturer's (R2 ¼ 0.96, SEE ¼ 2.03), B) Schols* (R2 ¼ 0.96, SEE ¼ 1.96) and C) Gray*(R2 ¼ 0.97, SEE ¼ 1.87) equation. DeE: BlandeAltman plots for D) Manufacturer's; Mean difference (95% limits of agreement (±1.96 SD)) 1.46 (�2.77, 5.69), E) Schols*; �0.16 (�3.96,3.64) and F) Gray*; 0.36 (�5.50, 6.22) equation. The dotted lines represent the regression lines between mean FFMDXA and FFMBIA and differences between FFMBIA and FFMDXA.Pearson's correlation coefficient r (p-value); D: 0.28 (p ¼ 0.066) E: �0.14 (p ¼ 0.356) and F: 0.73 (p < 0.001).

Table 3BEvaluation of fat-free mass (FFM) measured by segmental BIA against DXA as reference method. A comparison of different equations. N ¼ 41a.

Variable Mean difference SD 95% CI P R2 SEE

Manufacturer [24], kg �0.34 2.06 �0.99, 0.31 0.292 0.98 1.42Heitmann* [18,28], kg 0.17 1.83 �0.40, 0.75 0.546 0.97 1.85Gray* [17], kg �3.48 2.47 �4.26, �2.71 <0.001 0.97 1.95Heitmann [18,28], kg �0.69 1.97 �1.31, �0.07 0.031 0.97 1.96Geneva [14], kg �2.40 1.99 �3.03, �1.78 <0.001 0.96 2.00Schols* [23], kg �3.39 2.20 �4.09, �2.70 <0.001 0.96 2.00Kyle [19], kg �4.44 2.58 �5.25, �3.63 <0.001 0.96 2.11Lukaski [21,28], kg �3.01 2.29 �3.73, �2.29 <0.001 0.96 2.19Lukaski* [21], kg �3.70 2.20 �4.40, �3.01 0.001 0.96 2.20Deurenberg (1991) [9,16], kg �4.85 2.33 �5.59, �4.11 <0.001 0.95 2.29Lohman* [20,32], kg 3.36 2.70 2.51, 4.21 <0.001 0.95 2.39Deurenberg (1990) [15], kg �10.96 2.58 �11.77, �10.14 <0.001 0.95 2.40Segal* [22], kg �1.86 2.64 �2.70, �1.03 <0.001 0.94 2.61

Abbreviations: SD, Standard Deviation; 95% CI, 95% Confidence interval; R2, R squared; SEE, Standard Error of the Estimate.P-values: Paired sample T-test.

a Equations included: 1) Manufacturers equation, 2) The Geneva equation (recommended by ESPEN) and 3) Equations previously tested for validity in cancer patients.*Indicate sex-specific equations.

H. Ræder et al. / Clinical Nutrition 37 (2018) 292e300 297

Schols* equation (93%), followed by segmental BIA using themanufacturer's equation (86%). Specificity (i.e. proportion withnormal FFMI correctly identified as such) was highest for whole-body BIA using the manufacturer's equation (100%), followed bysegmental BIA-using the Heitmann* equation (96%) and whole-body BIA using the Schols* equation (93%). The results demon-strate that type of BIA device and equation used have implicationsfor the proportion of patients categorized with low FFMI, andconsequently the ability to correctly classify patients asmalnourished.

different BIA devices for estimation of FFM by using DXA as areference method in CRC patients. Furthermore, our study is thefirst to evaluate various existing equations for estimation of FFM inorder to find the most appropriate BIA equation(s) for estimatingFFM in a cohort consisting solely of CRC patients.

The results of the present study show that estimating bodycomposition from impedance data is dependent on type of BIAdevice and equation used. The two BIA devices tested in the currentstudy, a whole-body BIA and a segmental BIA, resulted in signifi-cantly different R and Xc values, and hence FFM estimates, despite

Page 149: Malnutrition and dietary interventions in colorectal

using the same equation. This suggests that the two BIA-devicesshould not be used interchangeably; the accuracy of the equation

range�6.5 and 6.8 kg, whereas for segmental BIA, estimates variedfrom �11.0 to 3.4 kg. Our results are in accordance with observa-

Fig. 3. Fat-free mass (FFM) assessed by DXA and segmental BIA. AeB: Scatter plots for; A) Manufacturer's (R2 ¼ 0.98, SEE ¼ 1.42) and B) Heitmann* (R2 ¼ 0.97, SEE ¼ 1.85) equation.CeD: BlandeAltman plots for C) Manufacturer's; Mean difference (95% limits of agreement (±1.96 SD)) �0.34 (�4.38, 3.70) and D) Heitmann*; 0.17 (�3.42, 3.76) equation. Thedotted lines represent the regression lines between mean FFMDXA and FFMBIA and differences between FFMBIA and FFMDXA. Pearson's correlation coefficient r (p-value); C: 0.69(p < 0.001) and D: �0.03 (p ¼ 0.870).

for

H. Ræder et al. / Clinical Nutrition 37 (2018) 292e300298

will relate to the type of BIA device used in addition to population-specific factors such as age, gender, ethnicity and body compositioncharacteristics.

We observed a strong linear relationship between BIA and DXAestimates for all equations tested. However, there was a discrep-ancy in FFM estimates depending on the equations used. Forwhole-body BIA, mean difference in FFM estimates were in the

Table 4Performance of whole-body and segmental BIA to detect low FFMI (FFMI < 15 kg/m2

method.

DXA Whole-body BIA

Manufacturer Gray*

Low FFMI, % 32.6 25.6 34.9Sensitivity, % e 78.6 78.6Specificity, % e 100.0 86.2PPV, % e 100.0 73.3NPV, % e 90.6 89.3

Reference values: FFMI derived from DXA measurements (n ¼ 43).Abbreviations: BIA, Bioelectrical impedance analysis; DXA, Dual energy X-ray absorptipredictive value.

tions reported in a newly published review article by Haverkortet al. [28], including surgical and oncological patients, and under-score that selection of BIA equation has significant implications forthe accuracy of the estimates.

For whole-body BIA, the manufacturer's equation resulted in asmall overestimation of FFM by 1.5 kg whereas for segmental BIAno differences were detected. Despite small or no differences at the

women and <17 kg/m2 for men) using selected equations against DXA as reference

Segmental BIA

Schols* Manufacturer Heitmann*

34.9 43.9 29.392.9 85.7 78.693.1 77.8 96.386.7 66.7 91.796.4 91.3 89.7

ometry; FFMI, Fat-free mass index; PPV, Positive predictive values; NPV, Negative

Page 150: Malnutrition and dietary interventions in colorectal

group level, we observed that both devices demonstrated limita- has been suggested that segmental BIA may provide more accurate

This project has received funding from Research Council of

We would like to thank the patients for their valuable contri-

H. Ræder et al. / Clinical Nutrition 37 (2018) 292e300 299

tions with regards to proportional bias when using the manufac-turer's equations. We cannot explore this bias further, as theequations incorporated into the BIA devices software are unknown,a phenomena referred to as black-box equations in the literature.

Most of the BIA equations available are developed in healthyindividuals and consequently may not provide valid estimates inpatient populations due to differences across populations, forinstance with regard to body shape, fat fraction and hydration ofFFM. It is therefore recommended not to use general equationswithout prior validation in the population of interest [9]. In absenceof a CRC-specific equation, we chose to test the validity of BIA byuse of the Geneva equation recommended by ESPEN and a selectionof equations previously tested in cancer patients. Using a broadrange of validity measures, our results suggest that the Schols* andHeitmann* equations are the most suitable equations for estima-tion of FFM using whole-body and segmental BIA, respectively. TheSchols* equation was developed in patients with chronic obstruc-tive pulmonary disease using deuterium dilution as the referencemethod [23], while the Heitmann* equation was developed in ahealthy population using densitometry as the reference method[18]. The validity of the Schols* equation was tested in 51 patientswith locally advanced or metastatic cancer of the lung or color-ectum [29]. In contrast to our results, this study reported poorability of BIA to estimate FFM in cancer patients. The discrepancybetween our results and the results of Mourtzakis et al. [29] may bedue to the older foot-to foot BIA-device (Tanita Body CompositionAnalyzer TBF-300A) used by Mourtzakis et al. The validity of theHeitmann* equation was tested in a group of 26 patients who hadundergone major abdominal surgery. Similar to our results, Jensenet al. found good agreement between FFM estimates derived fromBIA and DXA [30].

In clinical practice, the manufacturer's equations will most oftenbe utilized. On a group level, the use of these equations resulted insmall differences in FFM for the whole-body BIA device and nodifference for the segmental BIA device. On the individual level,FFM estimates were within �2.8e5.7 and �4.4e3.7 kg, usingwhole-body and segmental BIA, respectively, for approximately95% of the patients. Furthermore, both equations gave proportionalbias. Taken together, these findings suggest that both BIA devicesmay be appropriate to determine body composition of groups,however, considering the variation in measurement accuracy at theindividual level, single measurements should be interpreted withcare. Furthermore, onemust be aware that the direction of bias maybe affected by the size of the FFM compartment.

To investigate the clinical implications of using two different BIAdevices, we used FFM estimates from both devices, based on themanufacturer's equation as well as the most suitable equations (i.e.the Schols* and Gray* equation for whole-body BIA and the Heit-mann* equation for segmental BIA) to identify the proportion ofpatients with low FFMI according to the new consensus definitionfor malnutrition proposed by ESPEN [8]. We observed that theproportion of patients identified with low FFMI varied from 26 to44% depending on device and equation used, with sensitivityranging from 79 to 93%. Thus, some patients will not be correctlyidentified with low FFMI if a suboptimal equation is used. On theother hand, by using the optimal equation, whole-body BIA as wellas segmental BIA (to a lower degree) may have acceptable ability todetect low FFM from a single measurement.

In the current study, including a high proportion of patientswith obesity and abdominal obesity, we compared two differentBIA devices relying on different approaches to estimate FFM; awhole-body approach and a segmental approach. For the whole-body approach, the various segments of the body will contributedifferently to resistance values based on conductive mass. Hence, it

estimates of FFM in patients at extremes of BMI ranges [10].However, this has yet to be confirmed in clinical studies [31]. Ourstudy demonstrated that both BIA devices showed good agreementwith DXA when using the appropriate equation. This was demon-strated in the group as a whole, and when looking at the obesesubjects only (data not shown). Hence, we could not confirm thesuperiority of segmental BIA over whole-body BIA in estimation ofFFM in this CRC population.

A strength of our study is the unselected inclusion of patientswithin the study patient cohort, e.g. inclusion of patients withabnormal body shapes, obesity, presence of chronic diseases andorthopedic prosthesis/implants. In previous validation studies,patients with these conditions have often been excluded due topossible interference with the BIA measurements, resulting inhighly selected patient populations. Despite our broad inclusion,we observed high agreement between FFM estimates fromboth BIAdevices and DXA using the appropriate equation, increasing thegeneralizability of our results in this patient population.

5. Conclusion

In a population of non-metastatic CRC patients, mostly con-sisting of Caucasian adults and with a wide range of body compo-sition measures, FFM estimates from both whole-body andsegmental BIA shows good agreement with DXA when using theappropriate equation. For whole-body BIA, the highest agreementwas observed for the Schols* equation, whereas for segmental BIA,the Heitmann* equation was the superior choice. At the individuallevel, both BIA-devices show acceptable ability to detect low FFMwhen using the optimal equation. We recommend using one ofthese combinations of device and equation for measuring FFM inthis population.

Statement of authorship

HR and ASK had the main responsibility for data analysis andwriting the manuscript. HR, ASK, CH, GF, HBH, SKB, IP, SS and RBcontributed to the conception and the design of the study, analysisand interpretation of the data and drafting of the manuscript. ASK,HR, CH, GF, HBH, SKB and IP contributed to acquisition of data. Allauthors contributed to the writing and final approval of themanuscript.

Conflict of interest statement

All other authors declare that they have no competing interests.

Funding sources

Norway (grant number 190329), Throne Holst Foundation ofNutrition Research, Norwegian Cancer Society (grant number 3924/3927) and South Eastern Norway Regional Health Authority. Thesefunding sources were not involved in the study design, in thecollection, analysis or the interpretation of data.

Acknowledgements

bution to this study. We would also like to thank Torgrim Langleite,Jonas Grenne and Toril Melgaard for their contribution in datacollection and study administration.

Page 151: Malnutrition and dietary interventions in colorectal

List of abbreviations [10] Kyle UG, Bosaeus I, De Lorenzo AD, Deurenberg P, Elia M, Manuel Gomez J,

H. Ræder et al. / Clinical Nutrition 37 (2018) 292e300300

BIA Bioelectrical impedance analysisBMI Body mass indexCCC concordance correlation coefficientCRC Colorectal cancerDXA Dual energy X-ray absorptiometryESPEN European Society for Clinical Nutrition and MetabolismFFM Fat-free massFFMI Fat-free mass indexFM Fat massICD International classification of diseases and related health

problemsNPV Negative predictive valueN NumberPG-SGA Patient-generated subjective global assessmentPhA Phase anglePPV Positive predictive valueR ResistanceSD Standard deviationSEE Standard error of the estimateTNM Tumor node metastasisXc Reactance

Appendix A. Supplementary data

Supplementary data related to this article can be found at http://dx.doi.org/10.1016/j.clnu.2016.12.028.

References

[1] Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Fainsinger RL, et al. Defi-nition and classification of cancer cachexia: an international consensus. LancetOncol 2011;12(5):489e95.

[2] Prado CM, Baracos VE, McCargar LJ, Reiman T, Mourtzakis M, Tonkin K, et al.Sarcopenia as a determinant of chemotherapy toxicity and time to tumorprogression in metastatic breast cancer patients receiving capecitabinetreatment. Clin Cancer Res 2009;15(8):2920e6.

[3] Prado CM, Lieffers JR, McCargar LJ, Reiman T, Sawyer MB, Martin L, et al.Prevalence and clinical implications of sarcopenic obesity in patients withsolid tumours of the respiratory and gastrointestinal tracts: a population-based study. Lancet Oncol 2008;9(7):629e35.

[4] Tan BH, Birdsell LA, Martin L, Baracos VE, Fearon KC. Sarcopenia in an over-weight or obese patient is an adverse prognostic factor in pancreatic cancer.Clin Cancer Res 2009;15(22):6973e9.

[5] Prado CM, Siervo M, Mire E, Heymsfield SB, Stephan BC, Broyles S, et al.A population-based approach to define body-composition phenotypes. Am JClin Nutr 2014;99(6):1369e77.

[6] Thibault R, Pichard C. The evaluation of body composition: a useful tool forclinical practice. Ann Nutr Metabolism 2012;60(1):6e16.

[7] Rothney MP, Martin FP, Xia Y, Beaumont M, Davis C, Ergun D, et al. Precision ofGE Lunar iDXA for the measurement of total and regional body composition innonobese adults. J Clin Densitom 2012;15(4):399e404.

[8] Cederholm T, Bosaeus I, Barazzoni R, Bauer J, Van Gossum A, Klek S, et al.Diagnostic criteria for malnutrition e an ESPEN consensus statement. ClinNutr (Edinburgh, Scotland) 2015;34(3):335e40.

[9] Kyle UG, Bosaeus I, De Lorenzo AD, Deurenberg P, Elia M, Gomez JM, et al.Bioelectrical impedance analysisepart I: review of principles and methods.Clin Nutr (Edinburgh, Scotland) 2004;23(5):1226e43.

et al. Bioelectrical impedance analysis-part II: utilization in clinical practice.Clin Nutr (Edinburgh, Scotland) 2004;23(6):1430e53.

[11] Buchholz AC, Bartok C, Schoeller DA. The validity of bioelectrical impedancemodels in clinical populations. Nutr Clin Pract 2004;19(5):433e46.

[12] Colorectal cancer 2011 report: food, nutrition, physical activity, and theprevention of colorectal cancer. 2011.

[13] Edge SB. AJCC cancer staging handbook: from the AJCC cancer staging manual.New York: Springer; 2009. XIX, 718 s.ill. p.

[14] Kyle UG, Genton L, Karsegard L, Slosman DO, Pichard C. Single predictionequation for bioelectrical impedance analysis in adults aged 20e94 years.Nutrition (Burbank, Los Angeles County, Calif) 2001;17(3):248e53.

[15] Deurenberg P, van der Kooij K, Evers P, Hulshof T. Assessment of bodycomposition by bioelectrical impedance in a population aged greater than 60y. Am J Clin Nutr 1990;51(1):3e6.

[16] Deurenberg P, van der Kooy K, Leenen R, Weststrate JA, Seidell JC. Sex and agespecific prediction formulas for estimating body composition from bioelec-trical impedance: a cross-validation study. Int J Obes 1991;15(1):17e25.

[17] Gray DS, Bray GA, Gemayel N, Kaplan K. Effect of obesity on bioelectricalimpedance. Am J Clin Nutr 1989;50(2):255e60.

[18] Heitmann BL. Prediction of body water and fat in adult Danes from mea-surement of electrical impedance. A validation study. Int J Obes 1990;14(9):789e802.

[19] Kyle UG, Pichard C, Rochat T, Slosman DO, Fitting JW, Thiebaud D. Newbioelectrical impedance formula for patients with respiratory insufficiency:comparison to dual-energy X-ray absorptiometry. Eur Respir J 1998;12(4):960e6.

[20] Lohman TG. Preliminary results from the 1986 interlaboratory investigationon bioelectrical impedance. 1988.

[21] Lukaski HC, Bolonchuk WW, Hall CB, Siders WA. Validation of tetrapolarbioelectrical impedance method to assess human body composition. J ApplPhysiol (Bethesda, Md:1985) 1986;60(4):1327e32.

[22] Segal KR, Van Loan M, Fitzgerald PI, Hodgdon JA, Van Itallie TB. Lean bodymass estimation by bioelectrical impedance analysis: a four-site cross-vali-dation study. Am J Clin Nutr 1988;47(1):7e14.

[23] Steiner MC, Barton RL, Singh SJ, Morgan MD. Bedside methods versus dualenergy X-ray absorptiometry for body composition measurement in COPD.Eur Respir J 2002;19(4):626e31.

[24] Bosy-Westphal A, Schautz B, Later W, Kehayias JJ, Gallagher D, Muller MJ.What makes a BIA equation unique? Validity of eight-electrode multifre-quency BIA to estimate body composition in a healthy adult population. Eur JClin Nutr 2013;67(Suppl 1):S14e21.

[25] Ottery F. Patient-generated subjective global assessment. In: The clinicalguide to oncology nutrition. Chigaco: The American Dietetic Association;2000. p. 11e23.

[26] Patient-generated subjective global assessment. Available from: http://pt-global.org/.

[27] Parikh R, Mathai A, Parikh S, Chandra Sekhar G, Thomas R. Understanding andusing sensitivity, specificity and predictive values. Indian J Ophthalmol2008;56(1):45e50.

[28] Haverkort EB, Reijven PL, Binnekade JM, de van der Schueren MA,Earthman CP, Gouma DJ, et al. Bioelectrical impedance analysis to estimatebody composition in surgical and oncological patients: a systematic review.Eur J Clin Nutr 2015;69(1):3e13.

[29] Mourtzakis M, Prado CM, Lieffers JR, Reiman T, McCargar LJ, Baracos VE.A practical and precise approach to quantification of body composition incancer patients using computed tomography images acquired during routinecare. Appl Physiol Nutr Metab 2008;33(5):997e1006.

[30] Jensen MB, Hermann AP, Hessov I, Mosekilde L. Components of variance whenassessing the reproducibility of body composition measurements using bio-impedance and the Hologic QDR-2000 DXA scanner. Clin Nutr (Edinburgh,Scotland) 1997;16(2):61e5.

[31] Ward LC. Segmental bioelectrical impedance analysis: an update. Curr OpinClin Nutr Metab Care 2012;15(5):424e9.

[32] Graves JE, Pollock ML, Colvin AB, Van Loan M, Lohman TG. Comparison ofdifferent bioelectrical impedance analyzers in the prediction of bodycomposition. Am J Hum Biol 1989;1:603e11.

Page 152: Malnutrition and dietary interventions in colorectal

Ap

pen

dix

1.

Exi

stin

g em

piri

c eq

uati

ons

appl

ied

in th

e cu

rren

t stu

dy. T

he ta

ble

prov

ides

info

rmat

ion

abou

t the

pop

ulat

ion

in w

hich

the

equa

tion

s ar

e de

velo

ped,

type

of

BIA

inst

rum

ent a

nd r

efer

ence

met

hod

used

as

wel

l as

com

pone

nts

incl

uded

in th

e va

riou

s eq

uati

ons.

C

ompo

nent

s in

clud

ed in

equ

atio

n A

uth

or (

year

) P

opu

lati

on

dev

elop

ed

BIA

inst

rum

ent

P

has

e-se

nsi

tive

R

efer

ence

m

eth

od

Eq

uat

ion

Sex

Age

Wei

ght

Hei

ght

R

Xc

Bos

y-W

estp

hal

(201

3)

(“M

anuf

actu

rer’

s eq

uati

on”,

se

gmen

tal B

IA)

(1)

Hea

lthy

adu

lts

(18-

65 y

ears

) w

ith

BM

I; 1

8.5-

35.0

kg

/m2 (

n=12

4)

Seg

men

tal M

F-

BIA

(S

eca

mB

CA

515,

Sec

a,

Bir

min

gham

, U

nite

d K

ingd

om)

x F

our-

com

part

men

t m

odel

(A

DP

, D2O

, D

XA

)

Unk

now

n1 x

x x

x x

x

Unk

now

n (“

Man

ufac

ture

r’s

equa

tion

”, w

hole

-bo

dy B

IA)

Unk

now

n W

hole

-bod

y S

F-

BIA

(B

IA-m

odel

10

1, S

MT

M

edic

al,

Wür

zbur

g,

Ger

man

y)

x U

nkno

wn

Unk

now

n1

Kyl

e (2

001)

(“T

he

Gen

eva

equa

tion

”)

(2)

Hea

lthy

adu

lts

(20-

94 y

ears

) w

ith

BM

I; 1

7.0-

33.8

kg

/m2 (

n=34

3)

Who

le-b

ody

SF

-B

IA (

Xit

ron

4000

B, X

itro

n T

echn

olog

ies,

S

an D

iego

, CA

, U

SA)

x D

XA

(H

olog

ic

QD

R-4

500)

F

FM

= -

4.10

4 +

(0.

518

x H

t2 /R50

) +

(0

.231

x

wei

ght)

+

(0.1

30 x

Xc 5

0)

+ (

4.22

9 x

sex)

x

x x

x x

Luk

aski

(19

86)

(3,

4)2

Hea

lthy

adu

lts

(18-

50 y

ears

) w

ith

FF

M; 3

4.4-

96.3

kg

(n=

114)

Who

le-b

ody

SF

-B

IA (

RJL

-mod

el

101,

RJL

Sys

tem

s,

Det

roit

, MI,

US

A)

x D

ensi

tom

etry

F

FM

= (

0.81

0 x

H

t2 /R50

) +

6.3

9

x x

Luk

aski

* (1

986)

(3

)2 H

ealt

hy a

dults

(1

8-50

yea

rs)

wit

h F

FM

; 34.

4-96

.3

kg (

n=11

4)

Who

le-b

ody

SF

-B

IA (

RJL

-mod

el

101,

RJL

Sys

tem

s,

Det

roit

, MI,

US

A)

x D

ensi

tom

etry

F

FM

for

men

=

(0.8

27 x

H

t2 /R50

) +

5.

214

FF

M f

or

x

x

Page 153: Malnutrition and dietary interventions in colorectal

wom

en =

(0

.821

x

Ht2 /R

50)

+

4.91

7 S

egal

* (1

988)

(5)

3H

ealt

hy a

dults

(1

7-62

yea

rs)

wit

h F

M; 3

.0-5

6.0

%

(n=

1567

)

Who

le-b

ody

SF

-B

IA (

Unk

now

n m

odel

, R

JL

Sys

tem

s, D

etro

it,

MI,

US

A)

x D

ensi

tom

etry

F

FM

for

men

=

(0.0

0132

x H

t2 ) –

(0.0

4394

x

R50

) +

(0.

3052

0 x

wei

ght)

(0.1

6760

x a

ge)

+ 2

2.66

827

FF

M f

or

wom

en =

(0

.001

08 x

Ht2 )

– (0

.020

90 x

R

50)

+ (

0.23

199

x w

eigh

t) –

(0

.067

77 x

age

) +

14.

5945

3

x

x x

x

Hei

tman

n (1

990)

(4,

6)

4 H

ealt

hy a

dults

(3

5-65

yea

rs)

(n=

139)

Who

le-b

ody

SF

-B

IA (

RJL

-mod

el

103,

RJL

Sys

tem

s,

Det

roit

, MI,

US

A)

x F

our-

com

part

men

t m

odel

(B

ody

pota

ssiu

m (

who

le

body

cou

ntin

g)

and

tota

l bod

y w

ater

(d

ilut

omet

ry))

FF

M =

(0.

295

x H

t2 /R50

) +

(0

.204

x

wei

ght)

+

(5.0

09 x

sex

) –

(0.0

76 x

age

) +

(0

.227

x H

t) –

17

.04

x x

x x

x

Hei

tman

n* (

1990

) (4

, 6)4

Hea

lthy

adu

lts

(35-

65 y

ears

) (n

=13

9)

Who

le-b

ody

SF

-B

IA (

RJL

-mod

el

103,

RJL

Sys

tem

s,

Det

roit

, MI,

US

A)

x F

our-

com

part

men

t m

odel

(B

ody

pota

ssiu

m (

who

le

body

cou

ntin

g)

and

tota

l bod

y w

ater

(d

ilut

omet

ry))

FF

M f

or m

en =

(0

.244

x

Ht2 /R

50)

+

(0.2

70 x

w

eigh

t) +

(0

.284

x H

t) -

28

.02

x x

x

Page 154: Malnutrition and dietary interventions in colorectal

FF

M f

or

wom

en =

(0

.411

x

Ht2 /R

50)

+

(0.1

41 x

w

eigh

t) +

(0

.267

x H

t) -

28

.61

Deu

renb

erg

(199

0)

(7)

Hea

lthy

adu

lts

(60-

83 y

ears

) (n

=72

)

Who

le-b

ody

SF

-B

IA (

RJL

-mod

el

101,

RJL

Sys

tem

s,

Det

roit

, MI,

US

A)

x D

ensi

tom

etry

F

FM

= (

0.67

1 x

H

t2 /R50

) +

(3.

1 x

sex)

+ 3

.9

x

x

x

Deu

renb

erg

(199

1)

(8, 9

)5 H

ealt

hy a

dults

(>

16 y

ears

) (n

=66

1)

Who

le-b

ody

SF

-B

IA (

RJL

-mod

el

101,

RJL

Sys

tem

s,

Det

roit

, MI,

US

A)

x D

ensi

tom

etry

F

FM

= (

0.34

0 x

Ht2 /R

50)

+

(0.1

534

x H

t) +

(0

.273

x

wei

ght)

(0.1

27 x

age

) +

(4

.56

x se

x) -

12

.44

x x

x x

x

Loh

man

* (1

988)

(1

0, 1

1)6

Hea

lthy

adu

lts

(18-

29 y

ears

) (n

=15

3)

Who

le-b

ody

SF

-B

IA (

Unk

now

n m

odel

, Val

hall

a S

cien

tifi

c, S

an

Die

go, C

A, U

SA

)

Unk

now

n D

ensi

tom

etry

F

FM

for

men

=

(0.4

85 x

H

t2 /R50

) +

(0

.338

x

wei

ght)

+ 5

.32

FF

M f

or

wom

en =

(0

.475

x

Ht2 /R

50)

+

(0.2

95 x

w

eigh

t) +

5.4

9

x x

x

Sch

ols*

(12

)7C

OP

D p

atie

nts

(n=

117)

W

hole

-bod

y S

F-

BIA

(U

nkno

wn

man

ufac

ture

r an

d m

odel

)

Unk

now

n D

ilut

omet

ry

FF

M f

or m

en:

8.38

3 +

(0.

465

x H

t2 /R50

) +

(0

.213

x

x x

x

Page 155: Malnutrition and dietary interventions in colorectal

*Ind

icat

es g

ende

r-sp

ecif

ic e

quat

ions

; wom

en=

0, m

en=

1. N

one

of th

e eq

uati

ons

test

ed in

clud

ed a

nthr

opom

etri

c m

easu

res

(e.g

. thi

gh o

r an

kle

circ

umfe

renc

e).

1 Bot

h th

e eq

uati

ons

inco

rpor

ated

into

the

who

le-b

ody

BIA

and

seg

men

tal B

IA w

ere

unkn

own.

How

ever

, for

seg

men

tal B

IA th

e co

mpo

nent

s w

ere

know

n.

2 T

he e

quat

ion

test

ed w

as d

evel

oped

in 4

7 m

ale

volu

ntee

rs a

nd c

ross

-val

idat

ed a

gain

st a

pre

viou

s li

near

reg

ress

ion

equa

tion

dev

elop

ed in

37

mal

e vo

lunt

eers

(15

). T

he r

esul

ts o

f th

e cr

oss-

vali

dati

on s

how

ed n

o si

gnif

ican

t dif

fere

nce

betw

een

the

slop

es o

r th

e in

terc

epts

of

the

indi

vidu

al r

egre

ssio

n li

nes.

Fur

ther

mor

e, n

o si

gnif

ican

t dif

fere

nce

was

fou

nd b

etw

een

the

slop

es o

r th

e in

terc

epts

of

the

indi

vidu

al r

egre

ssio

n li

nes

for

mal

es (

n=84

) an

d fe

mal

es (

n=67

).

3 The

equ

atio

n te

sted

is th

e be

st-f

itti

ng r

egre

ssio

n li

ne f

rom

4 la

bora

tori

es p

oole

d bu

t wit

h se

para

te e

quat

ions

for

men

and

wom

en. A

qua

drup

le c

ross

-val

idat

ion

was

per

form

ed to

det

erm

ine

the

repr

oduc

ibil

ity

acro

ss la

bora

tori

es o

f th

e re

lati

onsh

ip b

etw

een

dens

itom

etri

cally

det

erm

ined

FFM

and

FFM

pre

dict

ed f

rom

BIA

and

oth

er v

aria

bles

.

4 The

equ

atio

n is

take

n fr

om th

e su

pple

men

tary

mat

eria

l of

the

revi

ew a

rtic

le b

y H

aver

kort

et a

l (20

15)

(4).

The

ori

gina

l art

icle

by

Hei

tman

n et

al (

1990

) (6

) is

not

ava

ilab

le a

t ww

w.p

ubm

ed.g

ov

(abs

trac

t onl

y).

5 The

ori

gina

l art

icle

by

Deu

renb

erg

et a

l (8)

is n

ot a

vaila

ble

at w

ww

.pub

med

.gov

(ab

stra

ct o

nly)

. The

equ

atio

n us

ed in

the

pres

ent s

tudy

is ta

ken

from

the

revi

ew a

rtic

le b

y K

yle

et a

l (20

04)

(9).

wei

ght)

F

FM

for

w

omen

: 7.6

10

+ (

0.47

4 x

Ht2 /R

50)

+

(0.1

84 x

w

eigh

t)

Kyl

e (1

998)

(13

) A

mbu

lato

ry

pati

ents

 wit

h se

vere

chr

onic

re

spir

ator

y in

suff

icie

ncy

(45-

86 y

ears

); B

MI

<

32.0

kg/

m2 (

n=75

)

Who

le-b

ody

SF

-B

IA (

Bio

-Z®

, New

C

ardi

ocor

p,

Fri

bour

g,

Sw

itze

rlan

d)

x D

XA

(H

olog

ic

CD

R-2

000®

) F

FM

= -

6.06

+

(Ht x

0.2

83)

+

(wei

ght x

0.

207)

- (

R50

x

0.02

4) +

(se

x x

4.03

6)

x

x x

x

Gra

y* (

1989

) (1

4)

Hea

lthy

adu

lts

(19-

74 y

ears

) w

ith

BM

I; 1

9.6-

53.3

kg

/m2 (

n=87

)

Who

le-b

ody

SF

-B

IA (

RJL

-mod

el

101,

RJL

Sys

tem

s,

Det

roit

. MI,

US

A)

x D

ensi

tom

etry

F

FM

for

men

=

(0.0

0139

x H

t2 ) –

(0.0

801

x R

50)

+ (

0.18

7 x

wei

ght)

+

39.8

30

FF

M f

or

wom

an =

(0

.001

51 x

Ht2 )

– (0

.034

4 x

R50

) +

0.1

40 x

w

eigh

t – (

0.15

8 x

age)

+ 2

0.38

7

(x

) x

x x

Page 156: Malnutrition and dietary interventions in colorectal

6 The

equ

atio

n te

sted

(“T

he L

ohm

an e

quat

ion”

) is

take

n fr

om a

n ar

ticl

e by

Gra

ves

et a

l (19

89)

(11)

ref

erri

ng to

unp

ubli

shed

res

ults

fro

m th

e 19

86 V

alha

lla

inte

r-la

bora

tory

inve

stig

atio

n of

BIA

(1

0).

7 The

equ

atio

n te

sted

(“T

he S

chol

s* e

quat

ion”

) is

take

n fr

om a

n ar

ticl

e by

Ste

iner

et a

l (20

02)

(12)

ref

erri

ng to

sex

-spe

cifi

c re

gres

sion

equ

atio

ns d

eriv

ed f

rom

Sch

ols

et a

l (U

nive

rsit

y of

M

aast

rich

t, M

aast

rich

t, th

e N

ethe

rlan

ds)

(per

sona

l com

mun

icat

ion)

.

Abb

revi

atio

ns: B

IA; B

ioel

ectr

ical

impe

danc

e an

alys

is, D

XA

; Dua

l ene

rgy

x-ra

y ab

sorp

tiom

etry

, AD

P; A

ir d

ispl

acem

ent p

leth

ysm

ogra

phy,

D2O

; Deu

teri

um o

xide

, SF

; Sin

gle-

freq

uenc

y, M

F;

Mul

ti-f

requ

ency

, BM

I; B

ody

mas

s in

dex,

FF

M; F

at f

ree

mas

s, F

M; F

at m

ass,

CO

PD

; Chr

onic

obs

truc

tive

pul

mon

ary

dise

ase,

Ht;

Hei

ght,

R50

; Res

ista

nce

at 5

0 kH

z, X

c; r

eact

ance

at 5

0 kH

z.

Page 157: Malnutrition and dietary interventions in colorectal

Ref

eren

ces

1.

Bos

y-W

estp

hal A

, Sch

autz

B, L

ater

W, K

ehay

ias

JJ, G

alla

gher

D, M

ulle

r M

J. W

hat m

akes

a B

IA e

quat

ion

uniq

ue?

Val

idit

y of

eig

ht-

elec

trod

e m

ulti

freq

uenc

y B

IA to

est

imat

e bo

dy c

ompo

siti

on in

a h

ealt

hy a

dult

pop

ulat

ion.

Eur

opea

n jo

urna

l of

clin

ical

nut

riti

on. 2

013;

67 S

uppl

1:

S14

-21.

2.

K

yle

UG

, Gen

ton

L, K

arse

gard

L, S

losm

an D

O, P

icha

rd C

. Sin

gle

pred

icti

on e

quat

ion

for

bioe

lect

rica

l im

peda

nce

anal

ysis

in a

dult

s ag

ed

20--

94 y

ears

. Nut

riti

on (

Bur

bank

, Los

Ang

eles

Cou

nty,

Cal

if).

200

1;17

(3):

248-

53.

3.

Luk

aski

HC

, Bol

onch

uk W

W, H

all C

B, S

ider

s W

A. V

alid

atio

n of

tetr

apol

ar b

ioel

ectr

ical

impe

danc

e m

etho

d to

ass

ess

hum

an b

ody

com

posi

tion

. Jou

rnal

of

appl

ied

phys

iolo

gy (

Bet

hesd

a, M

d : 1

985)

. 198

6;60

(4):

1327

-32.

4.

H

aver

kort

EB

, Rei

jven

PL

, Bin

neka

de J

M, d

e va

n de

r S

chue

ren

MA

, Ear

thm

an C

P, G

oum

a D

J, e

t al.

Bio

elec

tric

al im

peda

nce

anal

ysis

to

esti

mat

e bo

dy c

ompo

siti

on in

sur

gica

l and

onc

olog

ical

pat

ient

s: a

sys

tem

atic

rev

iew

. Eur

opea

n jo

urna

l of

clin

ical

nut

riti

on. 2

015;

69(1

):3-

13.

5.

Seg

al K

R, V

an L

oan

M, F

itzg

eral

d PI

, Hod

gdon

JA

, Van

Ita

llie

TB

. Lea

n bo

dy m

ass

esti

mat

ion

by b

ioel

ectr

ical

impe

danc

e an

alys

is: a

fo

ur-s

ite

cros

s-va

lida

tion

stu

dy. A

m J

Cli

n N

utr.

198

8;47

(1):

7-14

. 6.

H

eitm

ann

BL

. Pre

dict

ion

of b

ody

wat

er a

nd f

at in

adu

lt D

anes

fro

m m

easu

rem

ent o

f el

ectr

ical

impe

danc

e. A

val

idat

ion

stud

y.

Inte

rnat

iona

l jou

rnal

of

obes

ity.

199

0;14

(9):

789-

802.

7.

D

eure

nber

g P

, van

der

Koo

ij K

, Eve

rs P

, Hul

shof

T. A

sses

smen

t of

body

com

posi

tion

by

bioe

lect

rica

l im

peda

nce

in a

pop

ulat

ion

aged

gr

eate

r th

an 6

0 y.

Am

J C

lin

Nut

r. 1

990;

51(1

):3-

6.

8.

Deu

renb

erg

P, v

an d

er K

ooy

K, L

eene

n R

, Wes

tstr

ate

JA, S

eide

ll J

C. S

ex a

nd a

ge s

peci

fic

pred

icti

on f

orm

ulas

for

est

imat

ing

body

co

mpo

siti

on f

rom

bio

elec

tric

al im

peda

nce:

a c

ross

-val

idat

ion

stud

y. I

nter

nati

onal

jour

nal o

f ob

esit

y. 1

991;

15(1

):17

-25.

9.

K

yle

UG

, Bos

aeus

I, D

e L

oren

zo A

D, D

eure

nber

g P

, Eli

a M

, Gom

ez J

M, e

t al.

Bio

elec

tric

al im

peda

nce

anal

ysis

--pa

rt I

: rev

iew

of

prin

cipl

es a

nd m

etho

ds. C

lini

cal n

utri

tion

(E

dinb

urgh

, Sco

tlan

d). 2

004;

23(5

):12

26-4

3.

10.

Loh

man

TG

. Pre

lim

inar

y re

sult

s fr

om th

e 19

86 V

alha

lla

inte

rlab

orat

ory

inve

stig

atio

n on

bio

elec

tric

al im

peda

nce.

Unp

ubli

shed

dat

a. 1

988.

11

. G

rave

s JE

P, M

.L. C

olvi

n, A

.B. V

an L

oan,

M. a

nd L

ohm

an, T

.G. .

Com

pari

son

of D

iffe

rent

Bio

elec

tric

al I

mpe

danc

e A

naly

zers

in th

e P

redi

ctio

n of

Bod

y C

ompo

siti

on. A

m J

Hum

Bio

l. 19

89.

12.

Ste

iner

MC

, Bar

ton

RL

, Sin

gh S

J, M

orga

n M

D. B

edsi

de m

etho

ds v

ersu

s du

al e

nerg

y X

-ray

abs

orpt

iom

etry

for

bod

y co

mpo

siti

on

mea

sure

men

t in

CO

PD

. The

Eur

opea

n re

spir

ator

y jo

urna

l. 20

02;1

9(4)

:626

-31.

13

. K

yle

UG

, Pic

hard

C, R

ocha

t T, S

losm

an D

O, F

itti

ng J

W, T

hieb

aud

D. N

ew b

ioel

ectr

ical

impe

danc

e fo

rmul

a fo

r pa

tien

ts w

ith

resp

irat

ory

insu

ffic

ienc

y: c

ompa

riso

n to

dua

l-en

ergy

X-r

ay a

bsor

ptio

met

ry. T

he E

urop

ean

resp

irat

ory

jour

nal.

1998

;12(

4):9

60-6

. 14

. G

ray

DS

, Bra

y G

A, G

emay

el N

, Kap

lan

K. E

ffec

t of

obes

ity

on b

ioel

ectr

ical

impe

danc

e. A

m J

Cli

n N

utr.

198

9;50

(2):

255-

60.

15.

Luk

aski

HC

, Joh

nson

PE

, Bol

onch

uk W

W, L

ykke

n G

I. A

sses

smen

t of

fat-

free

mas

s us

ing

bioe

lect

rica

l im

peda

nce

mea

sure

men

ts o

f th

e hu

man

bod

y. A

m J

Cli

n N

utr.

198

5;41

(4):

810-

7.