You are on page 1of 11

Nutrition 102 (2022) 111725

Contents lists available at ScienceDirect

Nutrition
journal homepage: www.nutritionjrnl.com

Review

Improving nutrition in cystic fibrosis: A systematic literature review


Monika Mielus a,b,*, Dorota Sands a,b, Marek Woynarowski b,c
a
Cystic Fibrosis Department, Institute of Mother and Child, Warsaw, Poland
b w Lesny, Poland
Cystic Fibrosis Centre, Pediatric Hospital, Dziekano
c
Collegium Medicum of Jan Kochanowski University, Kielce, Poland

A R T I C L E I N F O A B S T R A C T

Article History: With increasing life expectancy of patients with cystic fibrosis (CF), gastrointestinal manifestations of the dis-
Received 4 November 2021 ease have been increasingly brought into focus. This was a systematic review of the PubMed database and
Accepted 25 April 2022 ongoing phase III clinical trials that aimed to summarize recent (published after June 1 2016) studies report-
ing the effects of nutritional interventions on anthropometric measures (weight, height, and body mass
Keywords: index) in patients with CF. Two ongoing trials and 40 published studies (18 interventional and 22 observa-
Systematic literature review
tional) were identified. Key results supported the benefits of comprehensive, individualized nutritional plans,
Nutrition
high-fat, high-calorie diet including high-quality carbohydrates, and enteric tube feeding (albeit the latter
Diet
Supplementation
was derived from observational studies only). In contrast, the supplementation of probiotics, lipids, docosa-
Weight hexaenoic, glutathione, or antioxidant-enriched multivitamin appeared to have little effect on anthropomet-
Hight, Body mass index ric measures.
© 2022 The Author(s). Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/)

Introduction men and 22 kg/m2 for women, whereas 42% of children are not
meeting the BMI goal of 50 percentile [4].
Pulmonary disease is the main cause of mortality and morbidity There is a clear link between improved nutritional status and
in patients with cystic fibrosis (CF) [1]; yet, with increasing life better lung function and longer survival in patients with CF [5].
expectancy of these patients, gastrointestinal (GI) manifestations Cystic fibrosis transmembrane regulator (CFTR) modulation ther-
of the disease have been increasingly brought into focus. Approxi- apy revolutionized the treatment of CF, but a recent comprehen-
mately 85% of patients with CF present with pancreatic insuffi- sive systematic review reported mixed effects of CFTR modulators
ciency, usually starting shortly after birth [2]. Insufficient on anthropometric measures [6]. In addition to treatment targeting
pancreatic enzyme production results in malabsorption of the disease mechanism of CF, a range of nutritional interventions,
nutrients and fat-soluble vitamins, which may be exacerbated by from dietary advice, through supplements, to enteric tube feeding
concomitant liver disease [3]. On the other hand, pulmonary infec- may be employed to improve nutrition in patients with CF. Despite
tion can result in larger caloric need and reduced appetite [3]. Con- the wide array of interventions available, an analysis of gaps in the
sequently, poor nutrition in CF is common. Data from the Cystic evidence for treatment decisions in CF identified as many as 20 evi-
Fibrosis Foundation registry has shown that 47% of adults with CF dence gaps related to GI care of patients with CF [7], highlighting
are not meeting the body mass index (BMI) goals of 23 kg/m2 for the need for high-quality research in the area. Against this back-
drop, we conducted a systematic review of published literature
and ongoing clinical trials that aimed to summarize recent
research into nutrition and GI care in people with CF.
The conduct and publication preparation of this review were supported by Mylan
Healthcare Sp.zo.o. The authors had full independence in the conduct of the study Review methodology
and the drafting of the manuscript. M.M. reported personal fees from Nestle, Nutri-
cia, and Vertex Pharmaceuticals and serves an advisory board for Mylan outside the The protocol of the review (Supplementary Material) was developed in accor-
submitted work. D.S. reports personal fees from Chiesi, Nutricia, Pfeizer, Roche, and dance with Preferred Reporting Items for Systematic review and Meta-Analysis
Vertex Pharmaceuticals and serves on advisory boards for Vertex and Mylan outside Protocols (PRISMA-P) [8] criteria and the results of the review are reported herein
the submitted work. MW reports personal fees from Nutricia and Vertex in accordance with the PRISMA 2009 Checklist [9].
Pharmaceuticals. The present review focused solely on nutritional interventions and gene ther-
*Corresponding author. apy; CFTR modulators were not captured as these had been recently described in a
E-mail address: monika.mielus@imid.med.pl (M. Mielus). comprehensive systematic review by Bailey et al. [6].

https://doi.org/10.1016/j.nut.2022.111725
0899-9007/© 2022 The Author(s). Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/)
2 M. Mielus et al. / Nutrition 102 (2022) 111725

Table 1
Eligibility criteria for published studies describing nutritional interventions in cystic fibrosis.

Inclusion criteria Exclusion criteria

Population Individuals with CF Individuals without CF; Animal studies


Intervention and comparator Diet; nutrients; appetite stimulants; PN; ETF; PERT; supple- Intervention of interest not reported
ments; gene therapy
Outcomes Dosing of the intervention; efficacy of the intervention, Outcomes of interest not reported
including change in weight/weight z-score/weight percentile
(for pediatric patients) from baseline; change in BMI/ zBMI/
BMI percentile (for pediatric patients) from baseline; change
in height/height z-score/height percentile (for pediatric
patients) from baseline; change in the proportion of patients
who are underweight, from baseline
Study design Original research articles describing: clinical trials: RCTs; non- Case reports or case series; Reviews, editorials, letters, com-
randomized controlled trials; single-arm studies; observa- ments, notes; animal or in vitro studies; pharmacokinetic
tional studies: cohort studies; case control studies; cross- studies; cost-effectiveness studies
sectional studies; registry/database studies
Language restrictions Studies published in English or Polish Studies published in languages other than English or Polish
Publication date June 1, 2016 March 14, 2021 Studies published before June 1, 2016; Studies published after
April 22, 2020
BMI, body mass index; ETF, enteral tube feeding; PERT, pancreatic enzyme replacement therapy; PN, parenteral nutrition; RCT, randomized controlled trial; zBMI, BMI z-score

The present review comprised two independent components: a review of the liprotamase compared with Pancreaze in patients with CF-related
published literature and a review of ongoing clinical trials. The PubMed database exocrine pancreatic insufficiency (EPI). EASY was an open-label,
was initially searched to identify relevant studies published between June 1, 2016
single-arm study to evaluate long-term safety of liprotamase in
and April 22, 2020. Ongoing clinical trials posted between those dates were
searched across the clinicaltrials.gov, EduraCT, and ISRCTN registers. These patients with CF-related EPI.
searches were subsequently updated using the same search terms as in the origi- The final trial, NCT04411901, was a single-arm trial that
nal review, except for the updated date restriction so that, overall, the date range assessed the effect of vitamin D3 on the severity of CF and non-CF
for the review was from June 1, 2016 to March 14, 2021.
pediatric bronchiectasis. The primary end point of this trial was
Publication date was restricted to identify studies published from June 2016,
to update on an earlier comprehensive review performed to inform the develop- reaching vitamin D levels of >30 ng/dL, while decreased number
ment of Australian and New Zealand guidelines on nutrition in CF [10]. of exacerbations and increased lung function were the secondary
Screening of titles and abstracts for published studies was performed in End- end points. It was not clear from the records available if the trial
Note X8 and EndNote20 to facilitate duplicate removal. Full texts of potentially eli- assessed anthropometric measures of interest. Although the trial
gible studies were subsequently reviewed to determine eligibility based on the
Population, Intervention, Comparator, Outcomes, and Study Design (PICOS) crite-
was recorded as completed, no results were available on Clinical-
ria provided in Table 1. trials.gov. A publication based on this trial was therefore identified
For ongoing trials, results from each registry search were exported into Micro- [13]. Although the improvement in vitamin D levels was signifi-
soft Excel and duplicates were removed manually to enable identification of cantly higher in patients with CF than in the non-CF bronchiectasis
unique studies. The review of ongoing clinical trials applied the eligibility criteria
outlined in Table 1 for population, intervention and comparator, and outcomes. In
group, changes in anthropometric measures with vitamin D sup-
terms of study design, however, only phase III trials were included. plementation were not reported in the publication [13].
Data extraction was performed by a single reviewer in a consistent manner
using two piloted forms in Microsoft Excel, one for published studies and one for
ongoing trials. Risk of bias in included studies was not assessed. Published reports
Outcomes of interest to the review focused solely on anthropometric meas-
ures of nutritional status (weight, height, and BMI), which are known to correlate The initial search strategy defined in the review protocol ren-
with lung function [11,12] and are easy to evaluate even in less developed health
systems, potentially expanding the geographical scope of the review.
dered 506 hits. An additional 150 hits were identified at the
update, resulting in 656 hits. We excluded 578 abstracts, and 77
full-text articles were screened for eligibility. Of these, 36 were
Search results
excluded; the vast majority (n = 33) because they did not report
outcomes of interest. Overall, 40 studies corresponding to 41
Ongoing phase III trials
articles were included (one identified article [14] was a correction
to an included study [15]). Outcomes of the selection process are
Clinicaltrials.gov (37 hits initial search, 7 hits update), EduraCT
documented in Figure 1.
(36 hits initial search, 15 hits update), and ISRCTN (0 hits at both
initial search and update) registers were searched, rendering 61
unique trials across the three registries. Of these trials, 58 were Overview of published studies
excluded due to population (n. = 6) or intervention (n = 52) not fit-
ting the review’s PICOS criteria. Three trials were included. Table 2 summarizes the characteristics of included studies.
The intervention assessed in two of these trials (EduraCT num-
bers 2017-000571-85 [Reliable, Emergent Solution Using Liprota- Interventional studies
mase Treatment (RESULT)] and 2016-002851-92 [Extended Access
to Sollpura over Years (EASY)]) was liprotamase, a pancreatic Of the 40 included studies, 18 were interventional. Of these, 11
enzyme replacement therapy (PERT) from non-animal sources, the were clinical trials, of which 7 were placebo-controlled [15 21],
development of which has been discontinued due to a failure to and the remaining were active controlled (n = 3) [22 24] or sin-
meet the primary end point in the phase III RESULT trial. Conse- gle-arm (n = 1) [25]. Six studies were prospective, interventional
quently, both EASY and RESULT were listed as prematurely ended. cohort studies [26 31], including a patient access scheme [30].
RESULT was a phase III, randomized, open-label, assessor-blind, The final publication reported on a small, prospective, cross-over
non-inferiority trial evaluating the efficacy and safety of study [32].
M. Mielus et al. / Nutrition 102 (2022) 111725 3

Fig. 1. Preferred Reporting Items for Systematic review and Meta-Analysis flow diagram of the PubMed review. *Forty studies corresponding to 41 articles as 1 identified arti-
cle was a correction to another included study. OrR, original review; U, update.

A wide range of pharmacologic, dietary, and other interventions Australasian Clinical Practice Guidelines for Nutrition in Cystic
was tested, including probiotic/symbiotic supplementation (n = 3) Fibrosis [51].
[17 19], dietary modification (n = 3) [22,28,31], patient education/
dietary management frameworks (n = 2) [25,29], vitamin supple-
Populations included
mentation (n = 2) [23,26], PERT (n = 1) [24], gene therapy (n = 1)
[16], choline (n = 1) [27], glutathione (Glu) [20], docosahexaenoic
Included studies varied substantially in cohort size (range:
acid (DHA) [21], a readily absorbable structured lipid (Encala) [15],
10 2304) and apparent methodological quality, although the latter
and supplemental foods (n = 1) [32]. Additionally, one study
was not formally assessed. Four studies included adults only
assessed a device, an immobilized lipase cartridge for extracorpo-
[27,35,38,39], 21 included solely pediatric patients
real digestion of enteral feedings (RELiZORB) [30].
[15,17 20,22,25,28,29,32,33,36,37,40,41,43,47,48,51,52,54], 8 did
not report the proportion of pediatric patients
Observational studies
[21,24,42,44 46,49,53], and the remaining 7 studies included a
mixed population of children and adults [16,23,26,30,31,34,50].
Twenty-two observational studies were included. Among these
22, four were prospective cohort studies [33 36] and 5 were retro-
spective studies [37 41]. Other study designs included cross-sec- Key results from identified published studies
tional studies (n = 8) [42 49], registry-based studies (n = 3)
[50 52], and case control studies (n = 2) [53,54]. Two Canadian Dietary management plans and frameworks
publications reported on the same cohort of 200 patients treated
at the Centre Hospitalier de l’Universite de Montreal [35,38]. No Four interventional studies examined holistic approaches to
overlap was detected between any of the remaining studies. nutritional care in the form of dietary plans, frameworks, or man-
Included studies assessed the relationships between nutrition- agement tools.
related outcomes and PERT (n = 3) [33,37,52] or pancreatic suffi- El-Koofy et al. enrolled 50 pediatric patients with CF in a pro-
ciency status (n = 1) [36], enteral feeding (n = 5) [34,39,50,53,54], spective, interventional cohort study of a comprehensive nutri-
vitamin intake (n = 8) [35,38,40 42,44 47], and calorie and/or tional plan with vitamin and mineral supplementation. The study
macronutrient intake (n = 3) [41,43,48]. One cross-sectional study was conducted in a single center in Egypt. Implementation of the
assessed the effects of oral nutritional supplements [49]. The final nutritional plan was associated with a significant improvement in
study reported on the effects of implementing the 2006 several nutritional indices, including weight z-score, after a period
4
Table 2
Summary of the design of included studies.

Study name Country Study design Cohort size Intervention Comparator Treatment duration

Dietary management plans and frameworks


Boon 2020 [25] International (Portugal, Spain, 6-mo, open label, prospective, 171 Mobile MyCyFAPP app None 6 mo
Italy, Belgium, Netherlands) multicenter interventional clini-
cal trial
El-Koofy 2020 [28] Egypt Prospective, interventional 50 Nutritional plan plus vitamin None 3 mo
cohort study and mineral supplementation
Revert 2018 [29] France Prospective, interventional Open cohort: 34 patients at Individually adapted therapeutic None 3y
cohort study study start and 44 at study end patient education
Van Biervliet 2021 Belgium Prospective pre post-interven- 34 Residential rehabilitation pro- None 3 wk
tion study gram combining nutritional
optimization, therapy support,
and physical activity
General calorie and macronutrient intake and diet
Gorji 2020 [22] Iran Randomized, double-blinded, 44 (22 per arm) Low glycemic index/HFCD HFCD 3 mo
parallel-group, clinical trial
Neri 2019 [43] Brazil Cross-sectional study 101, including 9 infants (<2 y of None (reported nutritional out- None N/A
age), 23 preschoolers (2 5 y), 18 comes by macronutrient intake
students (5 10 y), and 51 ado- and age group)

M. Mielus et al. / Nutrition 102 (2022) 111725


lescents (10+ y)
Poulimeneas 2020 [48] Greece Cross-sectional study 76 None (non-interventional study None N/A
that assessed nutritional intake
in general)
Ruseckaite 2018 [51] Australia Australian Cystic Fibrosis Data 2304 Implementation of the 2006 None N/A
Registry-based study Australasian Clinical Practice
Guidelines for Nutrition in Cystic
Fibrosis
Woestenenk 2019 [41] Netherlands Retrospective cohort study 191 None (reported on the relation- None N/A (mean § SD follow-up
ship between calorie intake and period was 4.7 § 2.3 y)
nutritional outcomes)
Supplemental foods
Pitman 2019 [32] U.S. Crossover study 10 2 flavors of peanut-based ready- None 2 wk
to-use supplemental food
Victoria 2021 [49] Spain Cross-sectional observational 59, including 13 patients (22%) ONS None N/A
study receiving ONS
Probiotics
Bruzzese 2018 [17] Italy Prospective, multicenter, dou- 95, including 41 in the probiotic Lactobacillus GG probiotics Placebo 1y
ble-blind, randomized, placebo- group and 40 in the placebo
controlled, interventional trial group
De Freitas 2018 [18] Brazil Randomized, placebo-controlled, 58, including 17 non-CF controls, Symbiotic supplementation Placebo 90 d
double-blind trial 19 CF patients in the placebo
group, and 22 patients in the
symbiotic group
Van Biervliet 2018 [19] Belgium Randomized, double blind pla- 31 Probiotic Placebo 4 mo, separated by a wash-out of
cebo-controlled crossover-study 1 mo
Vitamin supplementation general
Sagel 2018 [23] US Randomised, multicenter, dou- 73 in the mITT population, Antioxidant-enriched Standard multivitamin 16 wk
ble-blind, controlled trial including 36 randomized to anti- multivitamin
oxidant multivitamin and 37 to
control multivitamin

(continued on next page)


Table 2 (Continued)
Study name Country Study design Cohort size Intervention Comparator Treatment duration

Tham 2020 [47] Australia Cross-sectional study 164 including 82 children with None (reported the intake of None N/A
CF and 82 age- and sex-matched several vitamins and micronu-
controls trients, and nutritional out-
comes, for CF patients vs
controls)
Vitamin D supplementation
Abu-Fraiha 2019 [26] Israel Prospective, interventional 90 Vitamin D supplementation None 12+ mo (median 16.28 mo)
cohort study
Coriati 2017 [38] Canada Retrospective study 200 Vitamin D3 supplementation NR mean § SD: 5 § 0.9 y
(reports on the same cohort as
Lehoux Dubois 2019)
Lehoux Dubois 2019 [35] Canada Prospective observational cohort 200 Vitamin D3 supplementation NR mean § SD: 5 § 0.9 y
study (reports on the same cohort as
Coriati 2017)
Ongaratto 2018 [44] Brazil Retrospective cross-sectional 37 None (reported nutritional out- None N/A
study comes by vitamin D deficiency
status)
Vitamin A supplementation
Sapiejka 2017 [45] Poland Cross-sectional study 196 None (reported nutritional out- None N/A
comes by vitamin A deficiency

M. Mielus et al. / Nutrition 102 (2022) 111725


status)
Woestenenk 2016 [40] Netherlands Retrospective, observational 221 None (reported nutritional out- None N/A
study comes by serum vitamin A
levels)
Supplementation of other vitamins
Krzyzanowska 2018 [42] Ukraine Cross-sectional study 79 None (reported nutritional out- None N/A
comes by vitamin K deficiency
status)
Sapiejka 2018 [46] Poland Cross-sectional study 211 None (reported nutritional out- None N/A
comes by vitamin E deficiency
status)
Pancreatic sufficiency status and PERT
Calvo-Lerma 2017 [37] Spain Retrospective observational 16 PERT None 1y
study
Gelfond 2018 [33] US Prospective, observational 231 including 205 with pancre- PERT None Up to 1 y
cohort study atic insufficiency who initiated
PERT, 6 with pancreatic insuffi-
ciency but no PERT use, and 20
with pancreatic sufficiency
Munck 2018 [36] France Prospective, longitudinal, obser- 105, including 86 with pancre- None (reported nutritional out- None N/A
vational, multicenter study atic insufficiency and 19 with comes by pancreatic sufficiency
pancreatic sufficiency at baseline status)
Schechter 2018 [52] US Retrospective cohort analysis 502 PERT None 2y
based on the Cystic Fibrosis
Foundation Registry, including a
nested case control component

(continued on next page)

3
of

5
6
Table 2 (Continued)
Study name Country Study design Cohort size Intervention Comparator Treatment duration

Taylor 2016 [24] International: 34 sites in 7 Euro- Randomized, double-blind, 96 randomized (48 to Zenpep Kreon 28 d on each PERT
pean countries including Bel- active-controlled, crossover, Zenpep ! Kreon and 48 to
gium, Bulgaria, Germany, multinational, non-inferiority Kreon ! Zenpep)
Hungary, Italy, Poland, and the study 83 completers (41
UK Zenpep ! Kreon and 42
Kreon ! Zenpep)
Enteral tube feeding
Declercq 2020 [54] Belgium Retrospective case control 24 ETF None Median 4.7 y (IQR 2.8 5.7 y)
study
Hollander 2017 [39] Netherlands Retrospective observational 26 eTF, comprising at baseline: None 6 mo
study 57.7% nasogastric tube, 30.8%
PEG, 7.7% PRG, 3.8% nasoduode-
nal tube
Kedzior 2019 [34] Poland Prospective, observational 53 Enteral nutrition (PEG) None Range: 6 mo to 7 y
cohort study <1 y in 21%
1 2 y in 56%
2+ y in 23%
Khalaf 2019 [53] US Retrospective case control 60, including 20 patients who Enteral nutrition (PEG) No PEG NR
study received PEG and 40 controls

M. Mielus et al. / Nutrition 102 (2022) 111725


who did not
Libeert 2018 [50] Belgium Belgian Cystic Fibrosis Registry- 339, including 113 cases receiv- ETF No ETF Median (IQR): 2 (1 5) y
based, retrospective, longitudi- ing ETF and 226 controls not
nal study receiving ETF
Other interventions
Alton 2016 [16] UK Randomized, double-blind, pla- 136 (ITT) 60 in placebo arm and gGene therapy (CFTR gen- Placebo 1y
cebo-controlled phase IIb trial 76 in gene therapy arm e liposome complex pGM169/
116 (PP) 54 in placebo arm and GL67A)
62 in gene therapy arm
Bernhard 2019 [27] Germany Prospective, interventional 10 Choline None Median (IQR): 84 (84 91) d
cohort study
Bozic 2020 [20] United States Randomised, Double-blind, mul- 60 Reduced L-glutathione (GSH) Placebo 24 wk
ticenter, placebo-controlled
phase II trial
 pez-Neyra 2020 [21]
Lo Spain Randomized, double-blind, par- 87 Docosahexaenoic acid Placebo 48 wk
allel-group, placebo-controlled
trial
Sathe 2021 [30] Not reported explicitly, most Patient access program 100 RELiZORB: immobilized lipase None 12 mo
likely United States cartridge for extracorporeal
digestion of enteral feedings
Stallings 2020 [15] United States Double-blind placebo-controlled 66 Readily absorbable structured Placebo 3 mo presented in the analysis
study lipid (Encala) (total study duration was 12 mo)
CF, cystic fibrosis; ETF, enteral feeding tube; GSH, glutathione; HFCD, high-fat, high-calorie diet; IQR, interquartile range; ITT, intention-to-treat; mITT, modified intention-to-treat; N/A, not applicable; NR, not reported; ONS, oral
nutritional supplement; PEG, percutaneous endoscopic gastrostomy; PERT, pancreatic enzyme replacement therapy; PP, per-protocol; PRG, percutaneous radiologic gastrostomy;
M. Mielus et al. / Nutrition 102 (2022) 111725 7

mo; however, it should be noted that the study population was girls [41]. A longitudinal analysis over a mean follow-up of 4.7 y
substantially malnourished at baseline [28]. revealed that increased calorie intake was associated with signifi-
Revert et al. reported on a quality-of-care improvement pro- cant increases in WAZ in both sexes and a significant ZBMI increase
gram at a specialist CF center in France, aiming to improve the in boys only [41]. There was no association between calorie intake
nutritional status of children with CF. Initial cohort size was 34 and height-for-age adjusted-for-target-height z-score [41]. The die-
children, increasing to 44 by the end of the study. The program tary contribution of protein, fat, and carbohydrates was not associ-
was multistep, and involved, among other steps taken, effective ated with any of the three nutritional and growth parameters [41].
follow-up of each patient’s nutritional status, individualized The Australian Cystic Fibrosis Data Registry (ACFDR)-based
weight gain goals, and intensified follow-up for more difficult study by Ruseckaite et al. reported on the effects of implementing
cases. Implementation of the program over a 3-y period led to a the 2006 Australasian Clinical Practice Guidelines for Nutrition in
numerical improvement in mean BMI z-score (zBMI; P-value not Cystic Fibrosis [51]. Nutritional data from two independent pediat-
reported) in addition to achieving a high level of patient and parent ric-age cohorts (2 5 y and 6 11 y) were collected longitudinally
satisfaction [29]. between 1998 and 2014 (8 y before and after the publication of the
The study by Boon et al. [25], was a single-arm, open-label, pro- guidelines) [51]. After adjusting for cofounders, there was a signifi-
spective, multicenter, international, interventional clinical trial cant increase in weight and height z-scores and zBMI in both age
investigating the effect on MyCyFAPP mobile app on GI-related cohorts following the implementation of the nutritional guidelines
quality of life (QoL) in 171 pediatric patients with CF. The app in 2006, suggesting that implementing these guidelines in clinical
allowed recording of meals to be eaten (returning recommended practice had a positive effect on nutritional status of patients with
PERT dose in real-time) and GI symptoms, contained educational CF [51].
materials, and enabled contact with a health care professional. Glycemic index of consumed carbohydrates also appears to affect
Although usage of the tool over 6 mo improved patient QoL, there nutritional outcomes. In a randomized controlled, double-blind, par-
were contradictory effects on anthropometric parameters (signifi- allel-group, single-center trial conducted in Iran, Gorji et al. com-
cant increases in height z-score, but statistically significant pared low-glycemic index, high-fat, high-calorie diet (HFCD;
decreases in weight z-score and zBMI), making it difficult to draw macronutrient percentage as per the HFCD group, with carbohy-
any conclusions. drate source restricted to glycemic index <50) with HFCD diet (40%
Finally, an interventional pre post study conducted in Belgium fat, 20% protein, and 40% carbohydrate of any source regardless of
by Van Biervliet et al. described a 3-wk residential rehabilitation glycemic index). Forty-four pediatric patients (22 in each arm) were
program combining nutritional optimization, therapy support, and included in the analysis. Both diets resulted in significant increases
physical activity. In a cohort of 34 patients (mixed pediatric and in body weight over a 3-mo period; however, this was significantly
adult), the program resulted in significant increases in weight and higher in the low-glycemic index, HFCD group [22].
zBMI, as well as a reduction in the proportion of patients who
could be considered malnourished [31]. Supplemental foods

Calorie and macronutrient intake and diet In a crossover, U.S.-based, single-center study by Pitman et al.,
two types of ready-to-use, high-energy supplemental foods
Four observational studies examined calorie and macronutrient resulted in small but statistically significant increases in zBMI and
intake, strongly suggesting that nutritional intake [41] and growth weight [32]. However, this study was both small (N = 10) and
parameters [43,48] remain suboptimal in some children with CF; short-term (2 wk), so that no conclusions on the effects of supple-
however, implementation of nutritional guidelines may improve mental foods could be drawn based on it.
these parameters [51]. A cross-sectional study in Spain identified oral nutritional sup-
Neri et al. performed a cross-sectional study of 101 children and plement (ONS) use in 13 (22%) of 59 included patients [49].
adolescents with CF attending a single center in Brazil and reported Patients using ONS had significantly lower BMI, although the pro-
adequate calorie and macronutrient consumption and adequate portion of malnourished participants (fat-free mass index <17 kg/
nutritional status in most patients [43]. However, lower zBMIs m2 in males and <15 kg/m2 in females) was not significantly differ-
were observed in children 5 to <10 y of age and adolescents 10 y ent between users and non-users of ONS [49].
of age, with 35.3% and 33.3%, respectively, classed as underweight
based on a zBMI below 1 [43]. Similarly, in a cross-sectional study Probiotics
enrolling 76 children with CF in Greece, 9% of boys and 5% of girls
were underweight (BMI-for-age z-score [BAZ] < 2) and 19% of Probiotic/symbiotic supplementation was assessed in three pla-
boys and 18% of girls experienced nutritional failure (BAZ < 1.04), cebo-controlled randomized controlled trials (RCTs) enrolling 31
despite all patients meeting or exceeding the recommended total to 95 pediatric patients with CF [17 19]. Two of the RCTs followed
energy intake [48]. Carbohydrate and fiber intake was generally a parallel-group design, one for 3 mo [18] and the other for 1 y
inadequate, suggesting there remains room for improvement in [17]. The final RCT was a crossover study, with probiotic and pla-
terms of diet quality [48]. cebo administered for 4 mo each, separated by a washout period of
In contrast to the results from Brazil [43] and Greece [48], a ret- 1 mo [19]. None of the three probiotic supplements improved
rospective, single-center cohort study of 191 Dutch children with CF growth parameters. In the multicenter Italian study by Bruzzese
(2 10 y of age) reported that dietary intake exceeded 110% of the et al., BMI did not significantly differ between the study groups
estimated average requirement in only 47% of dietary measure- after 12 mo of supplementation with Lactobacillus rhamnosus GG
ments, and therefore did not fully achieve the recommended high or placebo [17]. Similarly, the single-center Brazilian study by de
energy levels [41]. Energy intake in relation to estimated require- Freitas et al. reported that 90 d of supplementations with Lactofos
ments decreased with increasing age [41]. In a cross-sectional analy- (containing L. paracasei, L. rhamnosus, L. acidophilus, and Bifidobac-
sis, higher calorie intake was associated with significantly higher terium lactis) did not result in improvements in WAZ, BAZ, or
weight-for-age z-score (WAZ), height-for-age-adjusted-for-target- height-for-age z-score (HAZ) relative to presupplementation base-
height, and zBMI in boys, but only with significantly higher WAZ in line, and the effect of probiotic supplementation on these
8 M. Mielus et al. / Nutrition 102 (2022) 111725

parameters was not statistically different from placebo [18]. supplementation with antioxidant-enriched or standard multivita-
Finally, the Belgian-based multicenter study by van Biervliet et al. min and weight z-score changed little over the course of the study
reported no significant differences in zBMI, weight or height z- in both groups [23].
scores after 4 mo of supplementation with probiotics (containing L.
rhamnosus and B. animalis) or placebo [19].
PERT
Vitamin supplementation
A prospective, longitudinal, observational, multicenter study
The intake of various vitamins was assessed in two interven- reported on the effects of pancreatic (in)sufficiency on nutritional
tional studies and eight observational studies. status and found longitudinal patterns of weight and length
Dietary intake of micronutrients, including vitamins, in patients z-scores in infants with pancreatic insufficiency to be significantly
with CF was assessed in a cross-sectional study by Tham et al., who lower compared with infants with pancreatic sufficiency [36].
reported on micronutrient intake in a pediatric CF population PERT itself was assessed in three observational studies
treated at a single center in Australia [47]. Absolute intakes of all [33,37,52] and one RCT [24].
assessed micronutrients apart from vitamin C and folate were sig- Of the three observational studies, two were U.S.-based, includ-
nificantly higher in patients with CF than in non-CF controls [47]. ing a large prospective, observational cohort study by Gelfond
Furthermore, for most micronutrients, the proportion of children et al. using the BONUS (The Baby Observational and Nutrition
not meeting adequate intake was lower in patients with CF than in Study) cohort [33] and a retrospective cohort study based on the
controls [47]. Cystic Fibrosis Registry [52]. The registry-based study by Schechter
Vitamin D intake was assessed by four studies, including three et al. reported greater improvements in WAZ in infants receiving
observational studies [35,38,44] and an interventional study [26]. higher PERT doses [52]. Conversely, Gelfond et al. reported that
Two of the observational studies (one retrospective and one pro- 12-mo weight z-scores were numerically lower in infants receiving
spective) [35,38] reported on the same cohort of 200 adults with higher PERT doses, although this relationship did not reach statisti-
CF from a single center in Canada. The remaining observational cal significance [33]. The authors concluded these results likely
study, a retrospective cross-sectional study based in a single center represented indication bias, due to clinicians increasing PERT dos-
in Brazil, was small (N = 37) [44]. Vitamin D levels did not appear ing in patients with failing growth and not increasing it in those
to significantly associate with differences in BMI in any of the three growing well [33]. The final study on PERT was a small, retrospec-
studies [35,38,44]. Similarly, in a prospective, interventional cohort tive, observational study, conducted at a single center in Spain that
study conducted by Abu-Fraiha et al. in a single center in Israel, reported stable BMI in a cohort of 16 children receiving individual-
vitamin D supplementation according to the CF Foundation guide- ized PERT doses over a 12-mo period [37].
lines resulted in only a very modest numerical increase in BMI The relevant RCT was a double-blind, active-controlled, cross-
(from mean [SD] 19.40 [3.87] to mean [SD] 19.74 [3.83]) over a over, multinational, non-inferiority trial comparing two PERT prep-
median supplementation period of 16.28 mo; no P-values for the arations, Zenpep and Kreon. Although non-inferiority and
change in BMI were reported [26]. equivalence of Zenpep to Kreon were demonstrated in this trial,
Two studies assessed the effect of vitamin A intake on nutri- both PERT preparations resulted in a similar, modest increase in
tional status, identifying no clear associations [40,45]. A retrospec- body weight (least squares mean of 0.5 kg) over a 28-d period that
tive, single-center, observational study of 221 pediatric Dutch did not differ significantly between trial arms. P-values for change
patients with CF reported that vitamin A deficiency was rare and from baseline were not reported [24].
median serum retinol values were within the reference age across
all age groups [40]. This study also reported WAZ, HAZ, and
weight-for-height z-score (WHZ) for the different pediatric age Enteral tube feeding
groups, with the two former measures consistently <0 [40]. In a
Polish cross-sectional study of 196 patients with CF, those with Enteral tube feeding (ETF) was investigated in four observa-
vitamin A deficiency and normal vitamin A status did not differ sig- tional studies, the largest of which was a Belgian Cystic Fibrosis
nificantly in terms of weight or height z-scores [45]. Registry-based, retrospective, longitudinal study enrolling 339
The role of intake of vitamins K and E was assessed in one study patients with CF (113 cases receiving ETF for a median of 2 y and
each. A cross-sectional study of 79 Ukrainian patients with CF 226 controls not receiving ETF; »70% of patients were pediatric)
employed two different measures of assessing vitamin K defi- [50]. Patients who progressed to receiving ETF had significantly
ciency: prothrombin concentration induced by vitamin K absence worse nutritional status than controls; this difference was evident
(PIVKA-II) and undercarboxylated osteocalcin percentage (u-OC) both at ETF initiation and earlier at first entry in the registry [50]. It
[42]. Weight z-score, but not height z-score, was significantly was also maintained at 3 y from ETF initiation [50]. Although zBMI
lower in patients with abnormal PIVKA-II level [42]. However, improved significantly after 3 y, there was no effect on height in
when u-OC was used to determine vitamin K status, patients with children [50].
normal, insufficient, and deficient u-OC levels did not differ signifi- The remaining four studies were smaller and included a pro-
cantly in terms of weight or height z-scores [42]. Therefore, the spective, observational cohort study of 53 patients in Poland
relationship between vitamin K status and anthropometric param- receiving ETF [34], a retrospective observational study of 26
eters depended on definition of vitamin K status used. In terms of patients receiving ETF in a single center in the Netherlands [39],
vitamin E status, a cross-sectional study of 211 Polish patients with and two retrospective case control studies: a U.S.-based, single-
CF found that those with and without vitamin E deficiency did not center study including 20 patients who received ETF and 40 con-
differ significantly in terms of weight and height z-scores [46]. trols who did not [53] and a Belgian study of 24 patients receiving
Finally, Sagel et al. reported on a comparison of two multivita- ETF and 18 age, sex, and pancreatic status-matched controls not
min preparations, based on a double-blind RCT conducted across receiving ETF [54]. All four studies reported numerical increases in
multiple centers in the United States [23]. There were no differen- body weight and/or BMI with ETF, although statistical significance
ces between the two groups in weight or BMI after 16 wk of of these findings varied between the studies [34,39,53,54].
M. Mielus et al. / Nutrition 102 (2022) 111725 9

Other supplements A recent systematic review was not able to identify and associa-
tion between macronutrient distribution and BMI in adults [57]
Four interventional studies assessed various types of supple- and a similar lack of association between anthropometric parame-
ments: choline [27], Glu [20], a readily absorbable structured lipid ters and dietary contribution of protein, fat, and carbohydrates
[15], and DHA [21]. The study by Bernhard et al. was a small, inter- was observed in a pediatric study [41] identified in this review.
ventional cohort study conducted in a single center in Germany However, evidence from a small RCT conducted by Gorji et al. sug-
that investigated the effects of choline supplementation in adult gests that deriving the carbohydrate contribution from low glyce-
men with CF for a median of 84 d [27]. Weight and BMI were mic index sources enhances weight gain [22]. It would be of
within the normal range at the beginning of the study and did not interest to attempt to replicate these results in a larger, interna-
change during its course; however, statistical significance of these tional study enrolling a more diverse population to ascertain the
findings was not reported [27]. Glu [20] and DHA [21] displayed no relative roles of glycemic index and carbohydrate quality in dietary
difference versus placebo in terms of BMI or weight in double- management.
blind RCTs enrolling 60 and 87 patients, respectively. Similarly, the Many studies identified in the review investigated probiotics
change in weight, height, and BMI z-scores over 3 mo with the [17 19], vitamin supplementation [23,26,35,38,40,42,44 47], or
readily absorbable lipid (Encala) was no different from the results other supplements, including choline [27], Glu [20], a readily absorb-
of the placebo group [15]. However, the identified study only able structured lipid [15], and DHA [21]. In general, none of these sup-
reported 3-mo data despite the total study duration being specified plements were associated with improvements in anthropometric
as 12 mo [15]. Inspection of the original report from this trial [55] measures. This result is not surprising given that the primary aim of
also did not permit assessment of Encala effects on anthropometric probiotic supplementation is to restore normal intestinal microbiota
parameters over the full 12-mo study duration. and thus potentially reduce inflammation within the gut, while vita-
min supplementation aims primarily to prevent and correct deficien-
Other interventions cies. With regard to lipid and fatty acid supplements, CF is associated
with a dysregulation of fatty acid metabolism [58], so interventions
Two additional studies assessing unique types of interventions aiming to correct this may not necessarily readily translate into
were also identified. The first was a patient access scheme for an improvements in anthropometric measures. However, outcomes per-
immobilized lipase cartridge for extracorporeal digestion of enteral taining to vitamin deficiency status, gut function and lipid metabo-
feedings (RELiZORB), which aims to improve digestibility of enteral lism were not assessed in this review.
feeds in patients with pancreatic insufficiency [30]. Significant A single study assessed the effects of gene therapy [16] and,
increases in weight, height,and weight z-score were reported although the trial was not specifically focused on anthropometric
among 100 (mostly pediatric) included patients over 1 y of using measures, no treatment-related differences in weight or BMI were
the device [30]. observed [16]. This is in stark contrast with CFTR modulators, for
The final study was a double-blind, placebo-controlled, multi- which some positive effects on anthropometric measures were
center, UK-based phase IIb RCT assessing nebulized gene therapy noted in a recent systematic review, although these varied depend-
over a period of 1 y [16]. Efficacy assessments focused primarily on ing on the CFTR mutation present and the type of modulation ther-
respiratory function, whereas weight and BMI were only included apy used [6].
as safety assessments [16]. No treatment-related differences in Identified evidence suggests that pancreatic insufficiency in
weight or BMI were observed [16]. infants with CF is associated with hampered growth [36]. Replen-
ishment of pancreatic enzymes through PERT is the mainstay of
Discussion nutritional management in patients with CF and pancreatic insuffi-
ciency [5,10] and plays a key role in maintaining an adequate
This systematic review of available literature collects and sum- nutritional status through facilitating the digestion and absorption
marized recent evidence on nutrition in CF. The 40 identified stud- of nutrients. Best practices pertaining to PERT use have been
ies provided evidence on specific nutritional interventions, described in recent clinical guidelines on nutrition in CF [5;10].
nutritional status, and holistic approaches to nutritional manage- The non-inferiority RTC by Taylor et al. identified in this review
ment in CF. compared two porcine PERT preparations formulated as enteric-
European Society for Clinical Nutrition and Metabolism coated microspheres (Kreon and Zenpep) and determined their
(ESPEN); European Society for Paediatric Gastroenterology, Hepa- equivalence [24]. The benefits of enteric-coated microsphere PERT
tology, and Nutrition (ESPGHAN); and European Cystic Fibrosis compared with enteric-coated tablets are well known [7]. A recent
Society (ECFS) guidelines [5] and Australian and New Zealand Cochrane review reported beneficial bowel-related outcomes with
guidelines [10] on nutrition in CF both recommend regular assess- enteric coated microspheres when compared with enteric-coated
ment of anthropometric parameters and nutritional markers, and tablets (and non enteric coated pancreatic enzyme prepara-
highlight the importance of patient or parent education about tions); however, limitations to the evidence included small
nutrition. Similarly, the 2020 guidelines from the Academy of study size and risk of bias [59]. Another Cochrane review is
Nutrition and Dietetics recommend that nutritional assessment planned to assess the effects of different PERT regimens
findings are translated into individualized patient education and (including dose and administration timing) on clinical out-
counseling, to obtain a personalized nutrition care plan [56]. comes in patients with CF [60] and its publication may help to
Implementation of comprehensive individual nutritional plans further optimize PERT use. Another avenue of research on PERT
resulted in improvement of anthropometric parameters in three is the identification of non-porcine enzyme sources, which may
studies [28,29,31] identified in our review. Technology such as be preferred in patients with allergies or those abstaining from
mobile apps could be employed to facilitate implementation of the pork-derived products for religious reasons. Both ongoing trials
nutritional plan by the patient. Although the evidence identified in identified in this review assessed liprotamase, a non-porcine
this review did not conclusively support a positive effect linked to biotechnology-derived PERT. The development of this product
using an app for CF nutritional management on anthropometric was, however, discontinued as its phase III trial failed to meet
parameters, there was an improvement in QoL [25]. its primary end point.
10 M. Mielus et al. / Nutrition 102 (2022) 111725

Another notable intervention examined in the identified Acknowledgments


studies was ETF. ETF is recommended in patients who remain
persistently undernourished despite less invasive nutritional The authors acknowledge Karolina Badora of Proper Medical
interventions [5]. All studies examining ETF that were identi- Writing sp. z o. o. for editorial assistance in the preparation of this
fied in this review were observational; the largest reported a manuscript.
sustained increase in BMI in Belgian Cystic Fibrosis Registry
patients receiving ETF [50]. Indeed, a recent Cochrane review
identified no RCTs of ETF, likely due to the ethical challenges of References
withholding this intervention in patients who need it [61].
[1] Sathe MN, Freeman AJ. Gastrointestinal, pancreatic, and hepatobiliary mani-
Despite this limitation of available evidence, comprehensive festations of cystic fibrosis. Pediatr Clin North Am 2016;63:679–98.
guidelines on ETF are available from the Cystic Fibrosis Founda- [2] Singh VK, Schwarzenberg SJ. Pancreatic insufficiency in cystic fibrosis. J Cyst
tion [62]. Fibros 2017;16:S70–8.
[3] Sabharwal S. Gastrointestinal manifestations of cystic fibrosis. Gastroenterol
Limitations of the current review include a lack of formal Hepatol (N Y) 2016;12:43–7.
assessment risk of bias in included studies, involvement of a [4] 2019 Cystic Fibrosis Foundation Patient Registry Highlights. Bethesda, MD:
single reviewer, and limiting the searches to the PubMed data- Cystic Fibrosis Foundation; 2020.
[5] Turck D, Braegger CP, Colombo C, Declercq D, Morton A, Pancheva R, et al.
base. The outcomes of interest were restricted to include only ESPEN-ESPGHAN-ECFS guidelines on nutrition care for infants, children, and
anthropometric measures, which are readily measurable and adults with cystic fibrosis. Clin Nutr 2016;35:557–77.
routinely used across different health systems. Although body [6] Bailey J, Rozga M, McDonald CM, Bowser EK, Farnham K, Mangus M, et al.
Effect of CFTR modulators on anthropometric parameters in individuals with
composition (fat versus muscle mass) would also be of interest
cystic fibrosis: an evidence analysis center systematic review. J Acad Nutr Diet
to the review, as fat-free mass has been reported to correlate 2021;121:1364–78. e2.
with respiratory function in patients with CF [63], there is no [7] Rowbotham NJ, Smith S, Prayle AP, Robinson KA, Smyth AR. Gaps in the evi-
standardized way to measure body composition, limiting com- dence for treatment decisions in cystic fibrosis: a systematic review. Thorax
2019;74:229–36.
parisons across studies and making the data potentially difficult [8] Moher D, Shamseer L, Clarke M, Ghersi D, Liberati A, Petticrew M, et al. Pre-
to interpret. Therefore, we refrained from collecting data on ferred reporting items for systematic review and meta-analysis protocols
body composition. The evidence collected in this systematic (PRISMA-P) 2015 statement. Syst Rev 2015;4:1.
[9] Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for sys-
review varied substantially in terms of study design and appar- tematic reviews and meta-analyses: the PRISMA statement. PLoS Med 2009;6:
ent quality as well as geographic location, resulting in different e1000097.
populations and health systems being captured, which should [10] Saxby N, Painter C, Kench A, King S, Crowder T, van der Haak N, et al. Nutrition
guidelines for cystic fibrosis in Australia and New Zealand. In: Bell SC, ed. Syd-
be considered when making any comparisons between the dif- ney, Australia: Thoracic Society of Australia and New Zealand; 2017.
ferent studies identified. In general, few RCTs were identified, [11] Pedreira CC, Robert RG, Dalton V, Oliver MR, Carlin JB, Robinson P, et al. Associ-
and those captured were often small. Although ethical consid- ation of body composition and lung function in children with cystic fibrosis.
Pediatr Pulmonol 2005;39:276–80.
erations may prohibit the conduct of RCTs assessing ETF, there [12] Sheikh S, Zemel BS, Stallings VA, Rubenstein RC, Kelly A. Body composition and
is a need for comprehensive, high-quality evidence on a num- pulmonary function in cystic fibrosis. Front Pediatr 2014;2:33.
ber of other common nutritional interventions in CF, such as [13] Ali H, Deraz T, Mohamed D, Mahmoud M. The role of vitamin D3 Therapy in
pediatric bronchiectasis severity (CF versus non-CF patients). Open J Pediatr
optimizing PERT and the role of supplemental foods in ensuring
2020;10:521–34.
adequate nutrition. Another important consideration, particu- [14] Stallings VA, Tindall AM, Mascarenhas MR, Maqbool A, Schall JI. Correction:
larly when interpreting the real-world, observational studies improved residual fat malabsorption and growth in children with cystic fibro-
identified, is the potential for confounding. Nutrition is a com- sis treated with a novel oral structured lipid supplement: a randomized con-
trolled trial. PloS One 2020;15:e0239642.
plex area and nutritional status may be affected by several fac- [15] Stallings VA, Tindall AM, Mascarenhas MR, Maqbool A, Schall JI. Improved
tors that are difficult to adjust for. Examples include poor residual fat malabsorption and growth in children with cystic fibrosis treated
adherence to therapeutic recommendations where there is high with a novel oral structured lipid supplement: a randomized controlled trial.
PloS One 2020;15:e0232685.
treatment burden and low socioeconomic status, which are [16] Alton E, Armstrong DK, Ashby D, Bayfield KJ, Bilton D, Bloomfield EV, et al. A
known risk factors for poor clinical outcomes in CF [64]. Fur- randomised, double-blind, placebo-controlled trial of repeated nebulisation of
thermore, CF affects multiple organ systems, and its manifesta- non-viral cystic fibrosis transmembrane conductance regulator (CFTR) gene
therapy in patients with cystic fibrosis. Southampton (UK): NIHR Journals
tions may vary between individuals, making it difficult to Library; 2016.
firmly establish relationships between specific nutritional inter- [17] Bruzzese E, Raia V, Ruberto E, Scotto R, Giannattasio A, Bruzzese D, et al. Lack of
ventions and nutritional status. Currently available guidelines efficacy of Lactobacillus GG in reducing pulmonary exacerbations and hospital
admissions in children with cystic fibrosis: a randomised placebo controlled
offer direction in establishing appropriate nutritional care for trial. J Cyst Fibros 2018;17:375–82.
patients with CF, and implementation of guidelines is associ- [18] de Freitas MB, Moreira EAM, Oliveira DL, Tomio C, da Rosa JS, Moreno YMF,
ated with long-term improvements in nutritional status of et al. Effect of synbiotic supplementation in children and adolescents with cys-
tic fibrosis: a randomized controlled clinical trial. Eur J Clin Nutr 2018;72:
these patients [51]. However, nutritional care and its goals
736–43.
should be individualized and regularly reviewed, to ensure [19] Van Biervliet S, Hauser B, Verhulst S, Stepman H, Delanghe J, Warzee JP, et al.
they continue to optimally fit the patient’s needs. Probiotics in cystic fibrosis patients: a double blind crossover placebo con-
trolled study: Pilot study from the ESPGHAN Working Group on Pancreas/CF.
Clin Nutr ESPEN 2018;27:59–65.
[20] Bozic M, Goss CH, Tirouvanziam RM, Baines A, Kloster M, Antoine L, et al.
Conclusion Oral glutathione and growth in cystic fibrosis: a multicenter, randomized,
placebo-controlled, double-blind trial. J Pediatr Gastroenterol Nutr
This systematic review of recent studies assessing nutritional 2020;71:771–7.
[21] Lopez-Neyra A, Suarez L, Mun ~ oz M, de Blas A, Ruiz de Valbuena M, Garriga M,
interventions in patients with CF confirmed the benefits of com-
et al. Long-term docosahexaenoic acid (DHA) supplementation in cystic fibro-
prehensive, individualized nutritional plans, HFCD diet including sis patients: a randomized, multi-center, double-blind, placebo-controlled
high-quality carbohydrates, and ETF. No evidence has been identi- trial. Prostaglandins Leukot Essent Fatty Acids 2020;162:102186.
fied to support a significant positive effect on anthropometric [22] Gorji Z, Modaresi M, Yekanni-Nejad S, Mahmoudi M. Effects of low glycemic
index/high-fat, high-calorie diet on glycemic control and lipid profiles of chil-
measures of the supplementation of probiotics, lipids, DHA, Glu, or dren and adolescence with cystic fibrosis: a randomized double-blind con-
antioxidant-enriched multivitamin. trolled clinical trial. Diabetes Metab Syndr 2020;14:87–92.
M. Mielus et al. / Nutrition 102 (2022) 111725 11

[23] Sagel SD, Khan U, Jain R, Graff G, Daines CL, Dunitz JM, et al. Effects of an anti- [44] Ongaratto R, Rosa KMD, Eloi JC, Epifanio M, Marostica P, Pinto LA. Association
oxidant-enriched multivitamin in cystic fibrosis. A randomized, controlled, between hypovitaminosis D and frequency of pulmonary exacerbations in
multicenter clinical trial. Am J Respir Crit Care Med 2018;198:639–47. children and adolescents with cystic fibrosis. Einstein (Sao Paulo) 2018;16:
[24] Taylor CJ, Thieroff-Ekerdt R, Shiff S, Magnus L, Fleming R, Gommoll C. Compari- eAO4143.
son of two pancreatic enzyme products for exocrine insufficiency in patients _
[45] Sapiejka E, Krzyzanowska P, Walkowiak D, Wenska-Chyzy _ E, Szczepanik M,
with cystic fibrosis. J Cyst Fibros 2016;15:675–80. Cofta S, et al. Vitamin A status and its determinants in patients with cystic
[25] Boon M, Calvo-Lerma J, Claes I, Havermans T, Asseiceira I, Bulfamante A, et al. fibrosis. Acta Sci Pol Technol Aliment 2017;16:345–54.
Use of a mobile application for self-management of pancreatic enzyme [46] Sapiejka E, Krzyzanowska-Jankowska P, Wenska-Chyzy E, Szczepanik M, Wal-
replacement therapy is associated with improved gastro-intestinal related kowiak D, Cofta S, et al. Vitamin E status and its determinants in patients with
quality of life in children with Cystic Fibrosis. J Cyst Fibros 2020;19:562–8. cystic fibrosis. Adv Med Sci 2018;63:341–6.
[26] Abu-Fraiha Y, Elyashar-Earon H, Shoseyov D, Cohen-Cymberknoh M, Armoni S, [47] Tham A, Katz TE, Sutherland RE, Garg M, Liu V, Tong CW, et al. Micronutrient
Kerem E, et al. Increasing vitamin D serum levels is associated with reduced intake in children with cystic fibrosis in Sydney, Australia. J Cyst Fibros
pulmonary exacerbations in patients with cystic fibrosis. J Pediatr Gastroen- 2020;19:146–52.
terol Nutr 2019;68:110–5. [48] Poulimeneas D, Grammatikopoulou MG, Devetzi P, Petrocheilou A, Kaditis AG,
[27] Bernhard W, Lange R, Graepler-Mainka U, Engel C, Machann J, Hund V, et al. Papamitsou T, et al. Adherence to dietary recommendations, nutrient intake
Choline supplementation in cystic fibrosis—the metabolic and clinical impact. adequacy and diet quality among pediatric cystic fibrosis patients: results
Nutrients 2019:11. from the GreeCF Study. Nutrients 2020;12:3126.
[28] El-Koofy N, El-Mahdy M, Fathy M, El Falaki M, El Dine Hamed DH. Nutritional  AF, María GO, Jose
[49] Victoria CB, Casilda O, Nuria P, Jose  SF, et al. Oral nutritional
rehabilitation for children with cystic fibrosis: single center study. Clin Nutr supplements in adults with cystic fibrosis: effects on intake, levels of fat-solu-
ESPEN 2020;35:201–6. ble vitamins, and bone remodeling biomarkers. Nutrients 2021;13:669.
[29] Revert K, Audran L, Pengam J, Lesne P, Pougheon Bertrand D. A quality [50] Libeert D, Declercq D, Wanyama S, Thomas M, Van Daele S, De Baets F, et al.
improvement program to improve nutritional status of children with cystic The effect of enteral tube feeding in cystic fibrosis: a registry based study. J
fibrosis aged 2-12 years old over a 3 year period at CF center Roscoff, Brittany. Cyst Fibros 2018;17:264–70.
Orphanet J Rare Dis 2018;13:8. [51] Ruseckaite R, Pekin N, King S, Carr E, Ahern S, Oldroyd J, et al. Evaluating the
[30] Sathe MN, Patel D, Stone A, First E. Evaluation of the effectiveness of in-line impact of 2006 Australasian Clinical Practice Guidelines for nutrition in chil-
immobilized lipase cartridge in enterally fed patients with cystic fibrosis. J dren with cystic fibrosis in Australia. Respir Med 2018;142:7–14.
Pediatr Gastroenterol Nutr 2021;72:18–23. [52] Schechter MS, Michel S, Liu S, Seo BW, Kapoor M, Khurmi R, et al. Relationship
[31] Van Biervliet S, Declercq D, Dereeper S, Vermeulen D, Wu € rth B, De Guschte- of initial pancreatic enzyme replacement therapy dose with weight gain in
naere A. The effect of an intensive residential rehabilitation program on body infants with cystic fibrosis. J Pediatr Gastroenterol Nutr 2018;67:520–6.
composition in patients with cystic fibrosis. Eur J Pediatr 2021;180:1981–5. [53] Khalaf RT, Green D, Amankwah EK, Peck J, Carr V, Goldenberg NA, et al.
[32] Pitman RT, Mui M, Michelson PH, Manary MJ. Ready-to-use supplemental food Percutaneous endoscopic gastrostomy tubes may be associated with pres-
for nutritional supplementation in cystic fibrosis. Curr Dev Nutr 2019;3. ervation of lung function in patients with cystic fibrosis. Nutr Clin Pract
nzz016. 2019;34:290–6.
[33] Gelfond D, Heltshe SL, Skalland M, Heubi JE, Kloster M, Leung DH, et al. Pancre- [54] Declercq D, Huysentruyt K, Hauser B, De Wachter E, Van Daele S, De Baets F,
atic Enzyme replacement therapy use in infants with cystic fibrosis diagnosed et al. Long-term use of tube feeding in children with cystic fibrosis: results
by newborn screening. J Pediatr Gastroenterol Nutr 2018;66:657–63. from two Belgian CF centers. Eur J Clin Nutr 2021;75:620–7.
˛
[34] Kedzior A, ºabuz-Roszak B, Jezierska S. Enteral nutrition in cystic fibrosis. [55] Stallings VA, Schall JI, Maqbool A, Mascarenhas MR, Alshaikh BN, Dougherty
Wiad Lek 2019;72:1890–3. KA, et al. Effect of oral lipid matrix supplement on fat absorption in cystic
[35] Lehoux Dubois C, Labreche E, Boudreau V, Colomba J, Mailhot M, Lavoie A, et al. fibrosis: a randomized placebo-controlled trial. J Pediatr Gastroenterol Nutr
Extra-skeletal impact of vitamin D supplementation protocol in an adult popu- 2016;63:676–80.
lation with cystic fibrosis. Clin Nutr 2019;38:1666–71. [56] McDonald CM, Alvarez JA, Bailey J, Bowser EK, Farnham K, Mangus M, et al.
[36] Munck A, Boulkedid R, Weiss L, Foucaud P, Wizla-Derambure N, Reix P, et al. Academy of Nutrition and Dietetics: 2020 Cystic Fibrosis Evidence Analysis
Nutritional status in the first 2 years of life in cystic fibrosis diagnosed by new- Center Evidence-Based Nutrition Practice Guideline. J Acad Nutr Diet
born screening. J Pediatr Gastroenterol Nutr 2018;67:123–30. 2020;121:1591–636. e3.
[37] Calvo-Lerma J, Martinez-Barona S, Masip E, Fornes V, Ribes-Koninckx C. Pan- [57] McDonald CM, Bowser EK, Farnham K, Alvarez JA, Padula L, Rozga M. Dietary
creatic enzyme replacement therapy in cystic fibrosis: dose, variability and macronutrient distribution and nutrition outcomes in persons with cystic
coefficient of fat absorption. Rev Esp Enferm Dig 2017;109:684–9. fibrosis: an evidence analysis center systematic review. J Acad Nutr Diet
[38] Coriati A, Labreche E, Mailhot M, Mircescu H, Berthiaume Y, Lavoie A, et al. 2020;121:1574–90. e3.
Vitamin D3 supplementation among adult patients with cystic fibrosis. Clin [58] Vandebrouck C, Ferreira T. Glued in lipids: lipointoxication in cystic fibrosis.
Nutr 2017;36:1580–5. EBioMedicine 2020;61:103038.
[39] Hollander FM, de Roos NM, Belle van Meerkerk G, Teding van Berkhout F, Hei- [59] Somaraju URR, Solis-Moya A. Pancreatic enzyme replacement therapy for peo-
jerman HGM, van de Graaf EA. Body weight and body mass index in patients ple with cystic fibrosis. Cochrane Database Syst Rev 2020;8:CD008227.
with end-stage cystic fibrosis stabilize after the start of enteral tube feeding. J [60] Ng C, Major G, Smyth AR. Timing of pancreatic enzyme replacement therapy
Acad Nutr Diet 2017;117:1808–15. (PERT) in cystic fibrosis. Cochrane Database Syst Rev 2021;8:CD013488.
[40] Woestenenk JW, Broos N, Stellato RK, Arets HG, van der Ent CK, Houwen RH. [61] Shimmin D, Lowdon J, Remmington T. Enteral tube feeding for cystic fibrosis.
Vitamin A intake and serum retinol levels in children and adolescents with Cochrane Database Syst Rev 2019;7:CD001198.
cystic fibrosis. Clin Nutr 2016;35:654–9. [62] Schwarzenberg SJ, Hempstead SE, McDonald CM, Powers SW, Wooldridge
[41] Woestenenk JW, Dalmeijer GW, van der Ent CK, Houwen RH. The relationship J, Blair S, et al. Enteral tube feeding for individuals with cystic fibrosis:
between energy intake and body-growth in children with cystic fibrosis. Clin Cystic Fibrosis Foundation evidence-informed guidelines. J Cyst Fibros
Nutr 2019;38:920–5. 2016;15:724–35.
[42] Krzyzanowska P, Drzymala-Czyz S, Rohovyk N, Bober L, Moczkco J, Rachel M, [63] Calella P, Valerio G, Brodlie M, Donini LM, Siervo M. Cystic fibrosis, body com-
et al. Prevalence of vitamin K deficiency and associated factors in non-supple- position, and health outcomes: a systematic review. Nutrition
mented cystic fibrosis patients. Arch Argent Pediatr 2018;116:e19–25. 2018;55 56:131–9.
[43] Neri LCL, Bergamaschi DP, Silva Filho L. Evaluation of nutritional status in [64] Bell SC, Mall MA, Gutierrez H, Macek M, Madge S, Davies JC, et al. The
patients with cystic fibrosis according to age group. Rev Paul Pediatr future of cystic fibrosis care: a global perspective. Lancet Respir Med
2019;37:58–64. 2020;8:65–124.

You might also like