Next Article in Journal
The Role of Selected Dietary Factors in the Development and Course of Endometriosis
Previous Article in Journal
Lifestyles, Food Consumption Frequencies, and Eating Behaviors among Three Main Disciplines of Undergraduate Students during the Early COVID-19 Outbreak in Thailand
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Comparative Effects of Different Nutritional Supplements on Inflammation, Nutritional Status, and Clinical Outcomes in Colorectal Cancer Patients: A Systematic Review and Network Meta-Analysis

West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(12), 2772; https://doi.org/10.3390/nu15122772
Submission received: 9 May 2023 / Revised: 3 June 2023 / Accepted: 12 June 2023 / Published: 16 June 2023
(This article belongs to the Section Clinical Nutrition)

Abstract

:
Nutritional supplements have been widely used in colorectal cancer (CRC) patients. The aim of this network meta-analysis (NMA) was to compare the effects of different nutritional supplements on inflammation, nutritional status, and clinical outcomes in CRC patients. Four electronic databases were searched until December 2022. Randomized controlled trials (RCTs) comparing nutritional supplements of omega-3 fatty acids (omega-3), arginine, vitamin D, glutamine, probiotics, or their combinations with placebo or standard treatment were selected. The outcomes were inflammatory indicators, nutritional indicators, and clinical outcomes. A random-effects Bayesian NMA was performed to rank the effect of each supplement. In total, 34 studies involving 2841 participants were included. Glutamine was superior in decreasing tumor necrosis factor-α (MD −25.2; 95% CrI [−32.62, −17.95]), whereas combined omega-3 and arginine supplementation was more effective in decreasing interleukin-6 (MD −61.41; 95% CrI [−97.85, −24.85]). No nutritional supplements significantly maintained nutritional indicators in CRC patients. Regarding clinical outcomes, glutamine ranked highest in reducing the length of hospital stay (MD −3.71; 95% CrI [−5.89, −1.72]) and the incidence of wound infections (RR 0.12; 95% CrI [0, 0.85]), and probiotics were rated as best in reducing the incidence of pneumonia (RR 0.38; 95% CrI [0.15, 0.81]). Future well-designed RCTs are needed to further confirm these findings.

1. Introduction

Globally, colorectal cancer (CRC) is the third most frequently diagnosed cancer and the second leading cause of cancer-related deaths. In 2020, approximately 1.9 million new cases of CRC emerged and 935,000 deaths occurred, accounting for approximately one-tenth of cancer cases and deaths [1]. Standard conventional treatments for CRC include surgery, chemotherapy, and radiation therapy. Depending on the localization and progression of the tumor, these treatments can be used in combination [2,3,4,5]. Due to long-term tumor consumption, inadequate nutritional intake, as well as stress responses and metabolic disorders caused by surgical trauma, chemotherapy, or radiotherapy, patients are prone to chronic inflammation, malnutrition, and complications [6,7,8,9]. Inflammation is a driving factor contributing to cancer progression, invasion, metastasis, and adverse clinical characteristics, such as malnutrition, weight loss, and fever, which can severely affect patients’ prognosis [10]. The prevalence of malnutrition in CRC patients ranges from 20 to 37% [11], which can cause treatment outcomes to decrease and negatively impact patient prognosis and long-term quality of life [6,12,13,14,15]. Furthermore, inflammation and malnutrition may be the causal factors for poor clinical outcomes such as wound infections (WI), anastomotic leaks (AL), and pneumonia in CRC patients [10,16]. The incidence of adverse clinical outcomes raises patients’ medical expenses and impedes their recovery process [17,18]. Therefore, it is critical to find effective interventions to reduce inflammation and improve the nutritional status and clinical outcomes of CRC patients.
Several clinical studies have demonstrated the potency of nutritional supplements, such as omega-3 fatty acids (omega-3), arginine, vitamin D, glutamine, and probiotics, as an effective intervention in decreasing inflammation and enhancing the nutritional status, as well as the overall clinical outcomes of CRC patients [8,19,20,21,22,23,24,25,26,27,28]. For instance, glutamine supplementation was discovered to modulate the immunometabolic response of CRC patients and lower their susceptibility to infections [22,23]. CRC patients consuming vitamin D and omega-3 also showed low inflammation and better nutritional status [19]. Furthermore, it has been observed that probiotic supplementation modulated gut microbiota in CRC patients, reduced bacterial translocation and inflammatory cytokine release, and improved clinical outcomes in patients [24].
To date, multiple systematic reviews and meta-analyses have reported similar results in which nutritional supplements have been shown to improve inflammation, nutritional status, and clinical outcomes in CRC patients [8,25,26]. However, the comparative effects of different nutritional supplements in CRC patients have not been estimated. Network meta-analysis (NMA) allows for the simultaneous comparison of the effect of multiple interventions, as direct comparisons between these nutritional supplements are lacking [29]. Moreover, NMA can also rank interventions based on various outcomes, aiding health professionals and clinicians in evidence-based decision-making. Much prior research suggests that omega-3, arginine, vitamin D, glutamine, and probiotics are the most effective and widely studied nutritional supplements for reducing inflammation and improving nutritional status and clinical outcomes in CRC patients [8,19,20,21,22,23,24,25,26,27,28]. Therefore, in this study, we selected these nutritional supplements, aiming to compare and rank the effects of these nutritional supplements on inflammation, nutritional status, and clinical outcomes in CRC patients, as well as provide evidence-based data on nutritional supplementation for CRC patients.

2. Method

2.1. Design and Registration

We conducted the NMA following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement [30], and this study has been registered in the International Prospective Register of Systematic Reviews (PROSPERO; identifier CRD42023392142).

2.2. Inclusion and Exclusion Criteria

Inclusion criteria were as follows: (1) Patients diagnosed with CRC, colon cancer, or rectal cancer of any age. (2) Patients in the intervention group received at least one of the following nutritional supplements (in any form, dose, or duration): glutamine, arginine, omega-3, probiotics, vitamin D, or a combination of these supplements. These nutritional supplements are the most well-studied in CRC patients. (3) Patients in the control group received either placebo or standard treatment. If patients in both intervention groups and control groups received general adjuvant therapy at the same time, the adjuvant therapy should be the same. (4) Studies reported at least one of the following outcomes: inflammatory indicators such as C-reactive protein (CRP), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6); nutritional indicators such as albumin (Alb), weight, and body mass index (BMI); clinical outcomes such as length of hospital stay (LOH), the incidence of urinary tract infections (UTI), the incidence of WI, the incidence of AL, and the incidence of pneumonia. (5) The study design was a randomized controlled trial (RCT). (6) The language of the studies was limited to English.
Exclusion criteria were as follows: (1) Patients with cancer types other than CRC. (2) Studies with unclear diagnostic and efficacy criteria. (3) Studies that were cohort studies, review articles, case reports, descriptive studies, opinion articles, or abstracts. (4) Studies with incomplete or erroneous data that could not be merged.

2.3. Search Methods

Two researchers (Y.J.Y. and H.Y.J.) independently performed an extensive search on PubMed, Embase, the Cochrane Central Register of Controlled Trials, and Web of Science up to 25 December 2022, without restrictions in terms of the document type, date/time, and publication status. Medical Subject Headings (MeSH) and the free words of the keywords, including all known spellings of “colorectal cancer”, “glutamine”, “arginine”, “omega-3 fatty acids”, “probiotics”, and “vitamin D” were applied for document retrieval (for searching strategies, see Supplementary Table S1).

2.4. Study Selection

According to our predefined inclusion and exclusion criteria mentioned above, two researchers (Y.J.Y. and H.Y.J.) independently conducted the study selection. First, all potentially relevant studies were imported into EndNote 20 to remove repetitive studies. Then, we screened titles and abstracts to exclude ineligible studies. Finally, the full texts were further screened. Disagreements were resolved through discussion or negotiation with a third researcher (C.X.R.).

2.5. Data Extraction and Quality Assessment

The Cochrane data extraction form was used to extract the following data and information: (1) basic information such as title, name of the first author, and year of publication; (2) basic characteristics of the research subjects, including age, gender, the numbers of cases in each group, and diagnosis criteria; (3) details about the intervention, including specific measures and timing; (4) outcome indicators such as the levels of CRP, TNF-α, IL-6, Alb, weight, BMI, and LOH, the incidence of UTI, WI, AL, and pneumonia were extracted for the baseline and post-treatment timepoints; (5) key elements of risk assessment for bias. One researcher (Y.J.Y.) extracted data, which was then confirmed for accuracy by another researcher (H.Y.J.).
The methodological quality of each included study was assessed using the modified Jadad scale [31]. Two researchers (Y.J.Y. and H.Y.J.) independently scored the following items: generation of random sequences, allocation concealment, intervention blinding of participants and investigators, and incomplete outcome data. The modified Jadad scale scores range from 0 to 7. Studies with scores >3 were considered high quality, while studies with scores ≤3 were classified as low quality. The above scores were cross-checked and solutions were discussed with the third researcher (C.X.R.) if there were any discrepancies.

2.6. Data Synthesis and Statistical Analysis

Statistical models based on the Bayesian framework were constructed using the JAGS software (gemtc 0.8–2 and rjags 4–10 package) in R (version 4.1.2) (Rstudio, Boston, MA, USA). The mean difference (MD) with a 95% credible interval (CrI) was calculated for continuous data to determine the effect size. A pooled risk ratio (RR) with a 95% CrI was calculated for categorical data. Random-effect models for all NMA were employed because the trials included were clinically heterogeneous (different countries, doses of nutritional supplements, routes of supplementation, duration of supplementation, and anti-cancer treatment). Four Markov chains were set for each outcome, and each chain produced 50,000 iterations, with 20,000 iterations discarded as a burn-in period. The convergence of iterations was assessed with plots and the Gelman–Rubin–Brooks statistic [32]. We used the surface under the cumulative rank curve (SUCRA) to estimate the relative rank of different nutritional supplements for each outcome of interest [33]. The higher the SUCRA value, the higher the intervention in the rank [33]. In addition, the consistency model and inconsistency model were compared using the deviation information criterion (DIC). If the difference in DIC was less than 5 points, consistency was considered good and consistency modeling was used [34]. Heterogeneity was assessed using the I2 statistic. I2 values below 25% were considered low heterogeneity, those of 25 to 75% were considered moderate heterogeneity, and those higher than 75% were considered high heterogeneity [35]. Comparison-adjusted funnel plots were used to test for publication bias. Network plots and comparison-adjusted funnel plots of NMA were drawn by Stata (version 17.0) (StataCorp, College Station, Texas, USA).

3. Results

3.1. Search Outcomes

The detailed procedure of study selection is shown as a PRISMA flow diagram in Figure 1. Initially, a total of 16,414 potentially relevant studies were identified from the four electronic databases mentioned above. After removing 5563 repetitive studies, the title and/or abstract of each study were screened based on the inclusion and exclusion criteria. Subsequently, 10,757 studies were excluded. The remaining 94 studies were further assessed for eligibility by examining their full texts, out of which 60 studies were excluded as they did not meet the criteria of RCTs, did not target the desired patients, and lacked data (details see Figure 1). Ultimately, 34 studies were deemed suitable and included in the NMA.

3.2. Characteristics of Included Studies

The characteristics and details of each included study in our NMA are presented in Table 1. Of the 34 eligible studies published from 1998 to 2022, 20 were conducted in Asia [19,20,22,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52], 10 in Europe [27,53,54,55,56,57,58,59,60,61], and 4 in South America [24,62,63,64]. Overall, 2841 CRC patients participated in the 34 RCTs, with study sample sizes ranging from 11 to 362 and mean ages ranging from 50 to 72 years old. Among them, 25 studies recruited patients with CRC surgery [20,22,24,27,36,37,40,41,42,43,44,45,47,48,49,50,51,52,53,54,55,56,57,61,63], 6 studies recruited patients with CRC chemotherapy [19,38,39,60,62,64], and 3 studies recruited patients with CRC chemoradiotherapy [46,58,59]. During the time of nutritional interventions, 7 studies intervened at the preoperative time point [24,27,40,41,43,47,57], 12 studies intervened during the perioperative period [22,42,44,45,48,49,50,51,53,54,56,63], 6 studies intervened postoperatively [20,36,37,52,55,61], 6 studies intervened during chemotherapy [19,38,39,60,62,64], and 3 studies intervened during chemoradiotherapy [46,58,59]. Depending on the means of nutritional intervention, 8 studies were administrated through parenteral nutrition [22,36,37,51,52,53,60,61], 24 studies were administrated through oral nutrition [19,20,24,38,39,40,41,42,43,44,45,46,47,48,49,50,54,55,57,58,59,62,63,64], and 2 studies were administrated through oral and enteral nutrition [27,56]. These studies evaluated a total of 9 nutritional supplements: glutamine (n = 7) [22,37,51,58,59,60,61], arginine (n = 1) [57], omega-3 (n = 9) [19,36,37,38,52,53,54,62,64], probiotics (n = 16) [20,24,40,41,42,43,44,45,46,47,48,49,50,55,56,63], vitamin D (n = 1) [19], combined with omega-3 and arginine (omega-3 + arginine) (n = 1) [27], combined with omega-3 and vitamin D (omega-3 + vitamin D) (n = 1) [19], combined with omega-3 and probiotics (omega-3 + probiotics) (n = 1) [39], and combined with omega-3 and glutamine (omega-3 + glutamine) (n = 1) [37]. As for the study design, all included studies were RCTs, among which 32 studies were two-arm studies [20,22,24,27,36,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64] and 2 were four-arm studies [19,37].

3.3. Quality Assessment

The Jadad quality assessment scale showed that all 34 studies were of high quality (Jadad score > 3) (Table 1).

3.4. Network Meta-Analysis

3.4.1. Inflammatory Indicators

Tumor Necrosis Factor-α

Overall, eight RCTs assessed the effects of six nutritional supplements on TNF-α (Figure 2A). The main findings of the NMA are shown in Figure 2B. Compared with placebo, glutamine (MD −25.2; 95% CrI [−32.62, −17.95]) and probiotics (MD −12.55; 95% CrI [−15.19, −9.92]) significantly reduced TNF-α levels in CRC patients (Figure 2B). Based on SUCRA, glutamine was the best nutritional supplement to reduce TNF-α (SUCRA = 99.9%) (Supplementary Table S5).

Interleukin-6

In total, fourteen RCTs and seven nutritional supplements were analyzed for their effects on IL-6 (Figure 3A). Compared with the placebo, omega-3 + arginine (MD −61.41; 95% CrI [−97.85, −24.85]) and probiotics (MD −21.12; 95% CrI [−40.38, −2.89])] significantly reduced IL-6 levels in CRC patients (Figure 3B). The remaining nutritional supplements also reduced IL-6 levels in CRC patients, but the difference did not reach significance (Figure 3B). The ranking based on SUCRA showed that omega-3 + arginine was the best choice to reduce IL-6 levels (SUCRA = 98.6%) (Supplementary Table S5).

C-reactive Protein

Seven nutritional supplements (arginine, probiotics, omega-3 + probiotics, omega-3 + vitamin D, omega-3, vitamin D, and glutamine) from nine RCTs were included in the analysis of CRP (Supplementary Figure S1). It was found that none of the nutritional supplements significantly reduced CRP levels compared with the placebo (Supplementary Table S2). There were also no significant differences in the comparative effects among these nutritional supplements (for details please see Supplementary Table S2). According to the SUCRA ranking, vitamin D may be the best nutritional supplement to reduce CRP (SUCRA = 87.2%) (Supplementary Table S5).

3.4.2. Nutritional Indicators

Nutritional indicators were evaluated, including Alb, weight, and BMI levels. The network plot was shown in Supplementary Figure S2. In total, the relative effects of four nutritional supplements (omega-3, omega-3 + vitamin D, vitamin D, and probiotics) on Alb levels were envaulted from six RCTs, the relative effects of six nutritional supplements (omega-3, vitamin D, omega-3 + vitamin D, omega-3 + probiotics, probiotics, and glutamine) on the levels of weight were evaluated from six RCTs, and the relative effects of five nutritional supplements (probiotics, omega-3 + probiotics, vitamin D, omega-3, and omega-3 + vitamin D) from five RCTs on BMI levels were analyzed (Supplementary Figure S2). These nutritional supplementation interventions were administered prior to, or in conjunction with, treatment (details see Table 1). The NMA showed that most nutritional supplements reduced the decline in Alb, weight, and BMI levels during patient treatment relative to placebo, but the differences were not statistically significant (Supplementary Table S3). The SUCRA ranking showed that omega-3 + vitamin D may be the best choice to maintain the Alb (SUCRA = 76.9%), weight (SUCRA = 66.6%), and BMI of patients during treatment (SUCRA = 63.6%) (Supplementary Table S6).

3.4.3. Clinical Outcomes

Length of Hospital Stay

Five nutritional supplements across thirteen RCTs were analyzed using NMA to evaluate the effects of nutritional supplements on the LOH in CRC patients (Figure 4A). The NMA showed that glutamine (MD −3.71; 95% CrI [−5.89, −1.72]) and omega-3 (MD −3.41; 95% CrI [−6.03, −0.81]) significantly reduced the LOH in CRC patients compared with placebo (Figure 4B). In terms of SUCRA ranking, glutamine was the best nutritional supplement to reduce the LOH (SUCRA = 78.7%) (Supplementary Table S7).

Urinary Tract Infections

The network plot of UTIs is shown in Supplementary Figure S3A, which includes a total of four nutritional supplements (probiotics, glutamine, omega-3, and omega-3 + arginine) from nine RCTs. According to the results of the NMA, none of the supplements significantly reduced the incidence of UTI, and there were no significant differences in the relative effects among the various supplements (Supplementary Table S4A). Based on SUCRA, probiotics may be the best nutritional supplement to reduce the incidence of UTI (SUCRA = 83.5%) (Supplementary Table S7).

Wound Infections

The four nutritional supplements from the seventeen RCTs provided data to compare their relative effects on the incidence of WIs (Figure 5A). Compared with placebo, glutamine and probiotics reduced the risk of WIs by 88% (RR 0.12; 95% CrI [0, 0.85]) and 39% (RR 0.61; 95% CrI [0.41, 0.86]), respectively (Figure 5B). In addition, a similar advantage exists for glutamine compared to omega-3 (RR 0.11; 95% CrI [0, 0.94]) in reducing the risk of WIs (Figure 5B). According to SUCRA, glutamine was the best nutritional supplement to reduce the incidence of WIs (SUCRA = 94.5%) (Supplementary Table S7).

Anastomotic Leaks

There were four nutritional supplements (omega-3 + arginine, omega-3, glutamine, and probiotics) from twelve RCTs included in the analysis of Als (Supplementary Figure S3B). The NMA showed that no nutritional supplements significantly reduced the incidence of ALs (Supplementary Table S4B). Moreover, the relative effects between different nutritional supplements were also not statistically significant (Supplementary Table S4B). Based on SUCRA, glutamine may be the best choice to reduce the incidence of ALs (SUCRA = 84.1%) (Supplementary Table S7).

Pneumonia

The incidence of pneumonia was reported in ten RCTs and involved three different nutritional supplements (Figure 6A). The analysis revealed that probiotics reduced the risk of pneumonia by 62% (RR 0.38; 95% CrI [0.15, 0.81]) compared to placebo (Figure 6B). The relative comparisons between different nutritional supplements were not statistically significant (Figure 6B). According to SUCRA, probiotics were the best nutritional supplement to reduce the incidence of pneumonia (SUCRA = 82.0%) (Supplementary Table S7).

3.5. Consistency and Publication Bias Assessment

The consistency model and inconsistency model were compared using DIC. Good consistency with DIC was indicated by changes less than five for all closed-loop models present. In terms of publication bias assessment, there was no evidence of publication bias in the comparison-adjusted funnel plots (Supplementary Figure S4).

4. Discussion

We conducted a comprehensive search of relevant publications and analyzed all available evidence from 34 RCTs with NMA to compare the effects of various nutritional supplements on inflammation, nutritional status, and clinical outcomes in CRC patients. Regarding inflammatory indicators, glutamine was rated as the most effective in reducing TNF-α levels, while omega-3 + arginine had the best potency to reduce IL-6 levels. However, none of the nutritional supplements significantly maintained the nutritional indicators of CRC patients. Regarding clinical outcomes, glutamine was the most effective nutritional supplement to reduce the LOH and the incidence of WI, whereas probiotics were rated as best in lowering the incidence of pneumonia in CRC patients.
Based on NMA, it was found that glutamine was significantly effective in reducing TNF-α levels in CRC patients, whereas omega-3 + arginine significantly inhibited IL-6 expression in CRC patients. Inflammation is a crucial factor in the proliferation and invasion of CRC, and severe inflammation can lead to cancer progression and worsen the prognosis of patients [10]. Previous systematic reviews and meta-analyses have shown that supplements such as glutamine, probiotics, omega-3, and arginine can attenuate inflammatory responses [65,66,67], coinciding with our results. The present study further confirms that optimal nutritional supplement intake may be used to suppress the expression of different inflammatory cytokines in CRC patients. As an immune nutrient, glutamine exhibits important regulatory effects on the proliferation and function of lymphocytes, macrophages, and neutrophils [68]. Furthermore, it can also reduce excessive inflammatory responses [68]. Taniguchi et al. showed that glutamine supplementation in colitis-associated CRC mice significantly reduced TNF-α levels [69]. Additionally, omega-3 can also decrease the expression of nuclear factor- κB (NF-κB) and peroxisome proliferator-activated receptors (PPARs), thereby suppressing the production of inflammatory cytokines [70]. Omega-3 can also be converted into anti-inflammatory lipid mediators that bind to specific receptors on immune cells to inhibit the production of proinflammatory cytokines [71]. Arginine can also suppress excessive inflammatory responses through the production of nitric oxide and immunomodulators such as resolvin and protectin [72,73]. Consistent with previous studies, we found that nutritional supplements did not substantially reduce CRP levels in CRC patients [15,73]. However, these findings should be interpreted cautiously due to the limited number of available trials. Of note, our study provides a rationale for the use of nutritional supplementation regimens with glutamine and omega-3 + arginine in patients with CRC. These supplements may improve the prognosis of CRC patients by reducing their excessive inflammatory response, which provides new insight for the future adjunctive treatment of CRC patients.
Based on our study, none of the nutritional supplements had a significant effect on maintaining the Alb, weight, and BMI levels of CRC patients during treatment. A meta-analysis study showed that perioperative omega-3 supplementation had no significant effect on maintaining weight, BMI, and Alb levels in patients with gastrointestinal cancer [74]. Furthermore, Brown et al. [75] reported that vitamin D supplementation during chemotherapy did not significantly improve weight and BMI in CRC patients. The results of the above study were consistent with our analysis. However, there were diverse findings, such as those by Yue et al. [8]. The results of a meta-analysis by Yue et al. showed that perioperative omega-3 supplementation significantly increased Alb levels in CRC patients [8]. This discrepancy may be attributed to heterogeneity in the study populations, including the baseline weight loss, types of anticancer treatment, and tolerance to the anticancer treatment. Furthermore, Alb, although used as a surrogate for nutritional status, can be influenced by other factors such as inflammation, renal function, and hydration status [76]. Although the results of our study did not reach significance, it is noteworthy that nutritional supplementation interventions before or during treatment were shown to be associated with maintaining the nutritional status of CRC patients during treatment. Importantly, maintaining the nutritional status of CRC patients during treatment can help to reduce the toxicity of the treatment and decrease the incidence of complications, thereby improving the prognosis and quality of life of patients [6,12]. Moreover, our NMA showed that omega-3 + vitamin D ranked highest in maintaining Alb, weight, and BMI levels in CRC patients during treatment. Malnutrition in CRC patients is typically associated with cancer-induced systemic inflammation, leading to anabolic resistance and muscle loss [6]. Omega-3 and vitamin D can stimulate immune cells in specific ways, such as by enhancing response function, maintaining a normal and moderate immune response, regulating cytokine production and release, and reducing harmful or excessive inflammatory responses, thereby maintaining the nutritional status of CRC patients [77,78,79,80]. The above evidence indicates that omega-3 + vitamin D has the most potential to be used to maintain the nutritional status of CRC patients during treatment.
Despite advancements in surgical procedures and perioperative care, CRC patients still experience adverse postoperative clinical outcomes after surgery. These poor outcomes not only increase medical expenses but also reduce the quality of life of patients [81,82]. Previous meta-analysis results showed that glutamine and omega-3 supplementation can significantly reduce LOH in CRC patients [28,83]. Our NMA confirmed these results and demonstrated that glutamine was the most effective nutritional supplement for reducing LOH in CRC patients. Glutamine is an immune nutrient that can improve the immune response of CRC patients, decrease the incidence of infectious complications, and subsequently reduce the LOH of CRC patients [68,84]. Similar to a previous study, we also found that glutamine was the most effective nutritional supplement to reduce the incidence of WI in CRC patients [28]. This may be related to the anti-inflammatory and healing-promoting effects of glutamine. Glutamine not only inhibits the excessive inflammatory response by regulating the body’s immune response but also promotes nitric oxide synthesis, which modulates cell proliferation, collagen formation, and wound contraction, thereby facilitating wound healing [68,85,86,87]. However, the extent of the effect of glutamine on the incidence of WI in CRC patients in the present study should be interpreted with caution, as only one trial was available. Moreover, our NMA showed that probiotics performed best in decreasing the incidence of pneumonia in CRC patients. This result aligns with a meta-analysis study by Araújo et al. [26]. The mechanism by which probiotics reduce the risk of postoperative pneumonia in CRC remains unclear. However, some proposed mechanisms include increasing the number of beneficial bacteria in the gut microbiota and maintaining the balance of the gut microbiota [88]. The balance of microbiota plays an essential role in the development and function of the immune system, both locally and systemically [89]. Secondly, probiotic supplementation significantly increases the levels of short-chain fatty acids, which promote anti-inflammatory mechanisms to maintain intestinal and systemic homeostasis [89]. Third, probiotic supplementation could maintain the intestinal epithelial barrier. Probiotics stimulate tight junction proteins in the intestinal epithelial cell, which promotes mucin secretion, increases the protective layer, prevents pathogenic bacteria from adhering to intestinal epithelial cells, and consequently reduces the translocation of pathogenic bacteria [90]. Finally, probiotics can regulate innate and adaptive immune responses and reduce systemic inflammatory responses [91]. These changes suggest that probiotic supplementation can have a beneficial effect on reducing the occurrence of pneumonia. Nonetheless, this study did not find that nutritional supplements significantly improved UTI incidence and AL incidence in CRC patients, which may be due to differences in the study population, study design, and protocol (the variability in the nutritional supplement routes, doses, start time, and duration). Based on our research findings, glutamine and probiotics can be used in patients undergoing CRC surgery to improve their clinical outcomes.

5. Strengths and Limitations

To our knowledge, this is the first NMA to compare the effects of different nutritional supplements on inflammation, nutritional status, and clinical outcomes in CRC patients, and to rank all supplements based on these factors. This study provides valuable information when deciding on the best nutritional intervention for CRC patients. Nevertheless, this NMA has several limitations that should be acknowledged. Firstly, the small sample sizes and the limited number of studies may affect the accuracy and applicability of our results. Secondly, the varying treatment of patients, the formula, dose, route, and time of administration of nutrition supplements among the studies included may result in heterogeneity. Unfortunately, subgroup analyses could not be performed due to the limited number of studies. Lastly, our study was limited to research published in English, introducing selectivity bias. Hence, more high-quality RCTs are required to confirm our findings.

6. Conclusions

There is no single nutritional supplement that works optimally for all indicators in CRC patients. Glutamine was found to be the most effective in lowering TNF-α levels, shortening LOH, and reducing the incidence of WI in CRC patients. Omega-3 + arginine performed optimally in reducing IL-6 levels in CRC patients. Probiotics performed best in reducing the incidence of pneumonia in CRC patients. Due to some limitations of existing clinical studies and evidence, future studies should focus on larger sample sizes, longer follow-ups, and more rigorous study designs to confirm these findings.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/nu15122772/s1. Supplementary Table S1: Search strategy to find potentially relevant articles for inclusion in the network meta-analysis. Supplementary Table S2 League Table: Nutritional supplements network meta-analysis results for C-reactive protein (CRP, mg/L). Supplementary Table S3 League Table: Nutritional supplements network meta-analysis results for nutritional indicators. (A) Albumin (Alb, g/L); (B) weight (kg); (C) body mass index (BMI, kg/m2). Supplementary Table S4 League Table: Nutritional supplements network meta-analysis results for clinical outcomes. (A) The incidence of urinary tract infections (UTI); (B) the incidence of anastomotic leaks (AL). Supplementary Table S5: SUCRA values of nutritional supplements according to inflammatory indicators. Supplementary Table S6: SUCRA values of nutritional supplements according to nutritional indicators. Supplementary Table S7: SUCRA values of nutritional supplements according to clinical outcomes. Supplementary Figure S1: Network plots for C-reactive protein (CRP, mg/L). Supplementary Figure S2: Network plots for nutritional indicators. (A) Albumin (Alb, g/L); (B) weight (kg); (C) body mass index (BMI, kg/m2). Supplementary Figure S3: Network plots for clinical outcomes. (A) The incidence of urinary tract infections; (B) the incidence of anastomotic leaks. Supplementary Figure S4 Funnel Plot: Funnel plot for the network of (A) tumor necrosis factor- α (TNF-α); (B) interleukin-6 (IL-6); (C) C-reactive protein (CRP); (D) the length of hospital stay (LOH); (E) the incidence of urinary tract infections (UTI); (F) the incidence of wound infections (WI); (G) the incidence of anastomotic leaks (AL); (H) the incidence of pneumonia.

Author Contributions

Conceptualization: J.Y. and Y.H.; methodology: J.Y., Y.H. and X.C.; formal analysis and investigation: J.Y.; writing—original draft preparation: J.Y.; writing—review and editing: J.Y., C.C. and K.L.; supervision: X.C. and K.L.; all authors have commented on previous versions of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by National Natural Science Foundation of China Regional Joint Project (No. U22A20334), Chengdu Science and Technology Bureau (No. 2022-YF05-01440-SN), and Chengdu Municipal Health Commission (No. 2022040).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Schmoll, H.J.; Van Cutsem, E.; Stein, A.; Valentini, V.; Glimelius, B.; Haustermans, K.; Nordlinger, B.; van de Velde, C.J.; Balmana, J.; Regula, J.; et al. ESMO Consensus Guidelines for management of patients with colon and rectal cancer. a personalized approach to clinical decision making. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2012, 23, 2479–2516. [Google Scholar] [CrossRef]
  3. Van Cutsem, E.; Cervantes, A.; Nordlinger, B.; Arnold, D. Metastatic colorectal cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2014, 25 (Suppl. S3), iii1–iii9. [Google Scholar] [CrossRef] [PubMed]
  4. Yoshino, T.; Arnold, D.; Taniguchi, H.; Pentheroudakis, G.; Yamazaki, K.; Xu, R.H.; Kim, T.W.; Ismail, F.; Tan, I.B.; Yeh, K.H.; et al. Pan-Asian adapted ESMO consensus guidelines for the management of patients with metastatic colorectal cancer: A JSMO-ESMO initiative endorsed by CSCO, KACO, MOS, SSO and TOS. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2018, 29, 44–70. [Google Scholar] [CrossRef] [PubMed]
  5. Van Cutsem, E.; Cervantes, A.; Adam, R.; Sobrero, A.; Van Krieken, J.H.; Aderka, D.; Aranda Aguilar, E.; Bardelli, A.; Benson, A.; Bodoky, G.; et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2016, 27, 1386–1422. [Google Scholar] [CrossRef]
  6. Arends, J.; Baracos, V.; Bertz, H.; Bozzetti, F.; Calder, P.C.; Deutz, N.E.P.; Erickson, N.; Laviano, A.; Lisanti, M.P.; Lobo, D.N.; et al. ESPEN expert group recommendations for action against cancer-related malnutrition. Clin. Nutr. 2017, 36, 1187–1196. [Google Scholar] [CrossRef] [Green Version]
  7. Mendes, M.C.; Pimentel, G.D.; Costa, F.O.; Carvalheira, J.B. Molecular and neuroendocrine mechanisms of cancer cachexia. J. Endocrinol. 2015, 226, R29–R43. [Google Scholar] [CrossRef] [Green Version]
  8. Yue, T.; Xiong, K.; Deng, J.; Hu, W.; Tan, T.; Li, S.; Yang, T.; Xiao, T. Meta-analysis of omega-3 polyunsaturated fatty acids on immune functions and nutritional status of patients with colorectal cancer. Front. Nutr. 2022, 9, 945590. [Google Scholar] [CrossRef] [PubMed]
  9. Lam, C.N.; Watt, A.E.; Isenring, E.A.; de van der Schueren, M.A.E.; van der Meij, B.S. The effect of oral omega-3 polyunsaturated fatty acid supplementation on muscle maintenance and quality of life in patients with cancer: A systematic review and meta-analysis. Clin. Nutr. 2021, 40, 3815–3826. [Google Scholar] [CrossRef]
  10. Moore, M.M.; Chua, W.; Charles, K.A.; Clarke, S.J. Inflammation and cancer: Causes and consequences. Clin. Pharmacol. Ther. 2010, 87, 504–508. [Google Scholar] [CrossRef]
  11. Burden, S.T.; Hill, J.; Shaffer, J.L.; Todd, C. Nutritional status of preoperative colorectal cancer patients. J. Hum. Nutr. Diet. Off. J. Br. Diet. Assoc. 2010, 23, 402–407. [Google Scholar] [CrossRef] [PubMed]
  12. Pressoir, M.; Desné, S.; Berchery, D.; Rossignol, G.; Poiree, B.; Meslier, M.; Traversier, S.; Vittot, M.; Simon, M.; Gekiere, J.P.; et al. Prevalence, risk factors and clinical implications of malnutrition in French Comprehensive Cancer Centres. Br. J. Cancer 2010, 102, 966–971. [Google Scholar] [CrossRef] [Green Version]
  13. Maasberg, S.; Knappe-Drzikova, B.; Vonderbeck, D.; Jann, H.; Weylandt, K.H.; Grieser, C.; Pascher, A.; Schefold, J.C.; Pavel, M.; Wiedenmann, B.; et al. Malnutrition Predicts Clinical Outcome in Patients with Neuroendocrine Neoplasia. Neuroendocrinology 2017, 104, 11–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Santarpia, L.; Contaldo, F.; Pasanisi, F. Nutritional screening and early treatment of malnutrition in cancer patients. J. Cachexia Sarcopenia Muscle 2011, 2, 27–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Mocellin, M.C.; Camargo, C.Q.; Nunes, E.A.; Fiates, G.M.R.; Trindade, E. A systematic review and meta-analysis of the n-3 polyunsaturated fatty acids effects on inflammatory markers in colorectal cancer. Clin. Nutr. 2016, 35, 359–369. [Google Scholar] [CrossRef]
  16. Song, H.N.; Wang, W.B.; Luo, X.; Huang, D.D.; Ruan, X.J.; Xing, C.G.; Chen, W.Z.; Dong, Q.T.; Chen, X.L. Effect of GLIM-defined malnutrition on postoperative clinical outcomes in patients with colorectal cancer. Jpn. J. Clin. Oncol. 2022, 52, 466–474. [Google Scholar] [CrossRef]
  17. Tahiri, M.; Sikder, T.; Maimon, G.; Teasdale, D.; Hamadani, F.; Sourial, N.; Feldman, L.S.; Guralnick, J.; Fraser, S.A.; Demyttenaere, S.; et al. The impact of postoperative complications on the recovery of elderly surgical patients. Surg. Endosc. 2016, 30, 1762–1770. [Google Scholar] [CrossRef] [PubMed]
  18. Pradarelli, J.C.; Healy, M.A.; Osborne, N.H.; Ghaferi, A.A.; Dimick, J.B.; Nathan, H. Variation in Medicare Expenditures for Treating Perioperative Complications: The Cost of Rescue. JAMA Surg. 2016, 151, e163340. [Google Scholar] [CrossRef]
  19. Haidari, F.; Abiri, B.; Iravani, M.; Ahmadi-Angali, K.; Vafa, M. Randomized Study of the Effect of Vitamin D and Omega-3 Fatty Acids Cosupplementation as Adjuvant Chemotherapy on Inflammation and Nutritional Status in Colorectal Cancer Patients. J. Diet. Suppl. 2020, 17, 384–400. [Google Scholar] [CrossRef]
  20. Zaharuddin, L.; Mokhtar, N.M.; Muhammad Nawawi, K.N.; Raja Ali, R.A. A randomized double-blind placebo-controlled trial of probiotics in post-surgical colorectal cancer. BMC Gastroenterol. 2019, 19, 131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Hossain, T.; Phillips, B.E.; Doleman, B.; Lund, J.N.; Williams, J.P. A double-blind randomized controlled trial of the effects of eicosapentaenoic acid supplementation on muscle inflammation and physical function in patients undergoing colorectal cancer resection. Clin. Nutr. 2020, 39, 2055–2061. [Google Scholar] [CrossRef] [PubMed]
  22. Cui, Y.; Hu, L.; Liu, Y.J.; Wu, Y.M.; Jing, L. Intravenous alanyl-L-glutamine balances glucose-insulin homeostasis and facilitates recovery in patients undergoing colonic resection: A randomised controlled trial. Eur. J. Anaesthesiol. 2014, 31, 212–218. [Google Scholar] [CrossRef]
  23. Jing-Xiang, S.; Xiao-Huang, T.; Lie, W.; Chen-Jing, L. Glutamine dipeptide-supplemented parenteral nutrition in patients with colorectal cancer. Clin. Nutr. Suppl. 2004, 1, 49–53. [Google Scholar] [CrossRef]
  24. Polakowski, C.B.; Kato, M.; Preti, V.B.; Schieferdecker, M.E.M.; Ligocki Campos, A.C. Impact of the preoperative use of synbiotics in colorectal cancer patients: A prospective, randomized, double-blind, placebo-controlled study. Nutrition 2019, 58, 40–46. [Google Scholar] [CrossRef] [PubMed]
  25. Buijs, L.F.; Dunlop, M.G.; Vaughan-Shaw, P.G. Vitamin D supplementation and colorectal cancer survival: Systematic review and meta-analysis. Color. Dis. 2020, 22, 47. [Google Scholar] [CrossRef]
  26. Araújo, M.M.; Montalvão-Sousa, T.M.; Teixeira, P.D.C.; Figueiredo, A.; Botelho, P.B. The effect of probiotics on postsurgical complications in patients with colorectal cancer: A systematic review and meta-analysis. Nutr. Rev. 2023, 81, 493–510. [Google Scholar] [CrossRef]
  27. Braga, M.; Gianotti, L.; Vignali, A.; Carlo, V.D. Preoperative oral arginine and n-3 fatty acid supplementation improves the immunometabolic host response and outcome after colorectal resection for cancer. Surgery 2002, 132, 805–814. [Google Scholar] [CrossRef] [PubMed]
  28. Yang, T.; Yan, X.; Cao, Y.; Bao, T.; Li, G.; Gu, S.; Xiong, K.; Xiao, T. Meta-analysis of Glutamine on Immune Function and Post-Operative Complications of Patients With Colorectal Cancer. Front. Nutr. 2021, 8, 765809. [Google Scholar] [CrossRef]
  29. Hoaglin, D.C.; Hawkins, N.; Jansen, J.P.; Scott, D.A.; Itzler, R.; Cappelleri, J.C.; Boersma, C.; Thompson, D.; Larholt, K.M.; Diaz, M.; et al. Conducting indirect-treatment-comparison and network-meta-analysis studies: Report of the ISPOR Task Force on Indirect Treatment Comparisons Good Research Practices: Part 2. Value Health J. Int. Soc. Pharm. Outcomes Res. 2011, 14, 429–437. [Google Scholar] [CrossRef] [Green Version]
  30. Hutton, B.; Salanti, G.; Caldwell, D.M.; Chaimani, A.; Schmid, C.H.; Cameron, C.; Ioannidis, J.P.; Straus, S.; Thorlund, K.; Jansen, J.P.; et al. The PRISMA extension statement for reporting of systematic reviews incorporating network meta-analyses of health care interventions: Checklist and explanations. Ann. Intern. Med. 2015, 162, 777–784. [Google Scholar] [CrossRef] [Green Version]
  31. Oremus, M.; Wolfson, C.; Perrault, A.; Demers, L.; Momoli, F.; Moride, Y. Interrater reliability of the modified Jadad quality scale for systematic reviews of Alzheimer’s disease drug trials. Dement. Geriatr. Cogn. Disord. 2001, 12, 232–236. [Google Scholar] [CrossRef] [PubMed]
  32. Brooks, S.P.; Gelman, A. General methods for monitoring convergence of iterative simulations. J. Comput. Graph. Stat. 1998, 7, 434–455. [Google Scholar]
  33. Veroniki, A.A.; Straus, S.E.; Fyraridis, A.; Tricco, A.C. The rank-heat plot is a novel way to present the results from a network meta-analysis including multiple outcomes. J. Clin. Epidemiol. 2016, 76, 193–199. [Google Scholar] [CrossRef]
  34. Dempster, A.P. The direct use of likelihood for significance testing. Stat. Comput. 1997, 7, 247–252. [Google Scholar] [CrossRef]
  35. Cumpston, M.; Li, T.; Page, M.J.; Chandler, J.; Welch, V.A.; Higgins, J.P.; Thomas, J. Updated guidance for trusted systematic reviews: A new edition of the Cochrane Handbook for Systematic Reviews of Interventions. Cochrane Database Syst. Rev. 2019, 10, Ed000142. [Google Scholar] [CrossRef] [Green Version]
  36. Liang, B.; Wang, S.; Ye, Y.J.; Yang, X.D.; Wang, Y.L.; Qu, J.; Xie, Q.W.; Yin, M.J. Impact of postoperative omega-3 fatty acid-supplemented parenteral nutrition on clinical outcomes and immunomodulations in colorectal cancer patients. World J. Gastroenterol. 2008, 14, 2434–2439. [Google Scholar] [CrossRef] [PubMed]
  37. Aliyazicioglu, T.; Cantürk, N.; Şimşek, T.; Kolayli, F.; Çekmen, M. Effects of Standard and/or Glutamine Dipeptide and/or Omega-3 Fatty Ascid-Supplemented Parenteral Nutrition on Neutrophil Functions, Interleukin-8 Level and Length of Stay-A Double Blind, Controlled, Randomised Study. East Afr. Med. J. 2013, 90, 59–66. [Google Scholar]
  38. Esfahani, A.; Somi, M.H.; Ayromlou, H.; Nikanfar, A.; Jafarabadi, M.A.; Sadat, B.E.; Ghoreishi, Z. The effect of n-3 polyunsaturated fatty acids on incidence and severity of oxaliplatin induced peripheral neuropathy: A randomized controlled trial. Biomark. Res. 2016, 4, 13. [Google Scholar] [CrossRef] [Green Version]
  39. Golkhalkhali, B.; Rajandram, R.; Paliany, A.S.; Ho, G.F.; Wan Ishak, W.Z.; Johari, C.S.; Chin, K.F. Strain-specific probiotic (microbial cell preparation) and omega-3 fatty acid in modulating quality of life and inflammatory markers in colorectal cancer patients: A randomized controlled trial. Asia-Pac. J. Clin. Oncol. 2018, 14, 179–191. [Google Scholar] [CrossRef]
  40. Tan, C.K.; Said, S.; Rajandram, R.; Wang, Z.; Roslani, A.C.; Chin, K.F. Pre-surgical Administration of Microbial Cell Preparation in Colorectal Cancer Patients: A Randomized Controlled Trial. World J. Surg. 2016, 40, 1985–1992. [Google Scholar] [CrossRef]
  41. Zhang, J.W.; Du, P.; Gao, J.; Yang, B.R.; Fang, W.J.; Ying, C.M. Preoperative probiotics decrease postoperative infectious complications of colorectal cancer. Am. J. Med. Sci. 2012, 343, 199–205. [Google Scholar] [CrossRef] [PubMed]
  42. Sadahiro, S.; Suzuki, T.; Tanaka, A.; Okada, K.; Kamata, H.; Ozaki, T.; Koga, Y. Comparison between oral antibiotics and probiotics as bowel preparation for elective colon cancer surgery to prevent infection: Prospective randomized trial. Surgery 2014, 155, 493–503. [Google Scholar] [CrossRef]
  43. Xu, Q.; Xu, P.; Cen, Y.; Li, W. Effects of preoperative oral administration of glucose solution combined with postoperative probiotics on inflammation and intestinal barrier function in patients after colorectal cancer surgery. Oncol. Lett. 2019, 18, 694–698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Mizuta, M.; Endo, I.; Yamamoto, S.; Inokawa, H.; Kubo, M.; Udaka, T.; Sogabe, O.; Maeda, H.; Shirakawa, K.; Okazaki, E.; et al. Perioperative supplementation with bifidobacteria improves postoperative nutritional recovery, inflammatory response, and fecal microbiota in patients undergoing colorectal surgery: A prospective, randomized clinical trial. Biosci. Microbiota Food Health 2016, 35, 77–87. [Google Scholar] [CrossRef] [Green Version]
  45. Komatsu, S.; Sakamoto, E.; Norimizu, S.; Shingu, Y.; Asahara, T.; Nomoto, K.; Nagino, M. Efficacy of perioperative synbiotics treatment for the prevention of surgical site infection after laparoscopic colorectal surgery: A randomized controlled trial. Surg. Today 2016, 46, 479–490. [Google Scholar] [CrossRef]
  46. Farshi Radvar, F.; Mohammad-Zadeh, M.; Mahdavi, R.; Andersen, V.; Nasirimotlagh, B.; Faramarzi, E.; Lotfi Yagin, N. Effect of synbiotic supplementation on matrix metalloproteinase enzymes, quality of life and dietary intake and weight changes in rectal cancer patients undergoing neoadjuvant chemoradiotherapy. Mediterr. J. Nutr. Metab. 2020, 13, 225–235. [Google Scholar] [CrossRef]
  47. Xie, X.; He, Y.; Li, H.; Yu, D.; Na, L.; Sun, T.; Zhang, D.; Shi, X.; Xia, Y.; Jiang, T.; et al. Effects of prebiotics on immunologic indicators and intestinal microbiota structure in perioperative colorectal cancer patients. Nutrition 2019, 61, 132–142. [Google Scholar] [CrossRef]
  48. Liu, Z.; Li, C.; Huang, M.; Tong, C.; Zhang, X.; Wang, L.; Peng, H.; Lan, P.; Zhang, P.; Huang, N.; et al. Positive regulatory effects of perioperative probiotic treatment on postoperative liver complications after colorectal liver metastases surgery: A double-center and double-blind randomized clinical trial. BMC Gastroenterol. 2015, 15, 34. [Google Scholar] [CrossRef] [Green Version]
  49. Liu, Z.; Qin, H.; Yang, Z.; Xia, Y.; Liu, W.; Yang, J.; Jiang, Y.; Zhang, H.; Yang, Z.; Wang, Y.; et al. Randomised clinical trial: The effects of perioperative probiotic treatment on barrier function and post-operative infectious complications in colorectal cancer surgery—A double-blind study. Aliment. Pharmacol. Ther. 2011, 33, 50–63. [Google Scholar] [CrossRef]
  50. Yang, Y.; Xia, Y.; Chen, H.; Hong, L.; Feng, J.; Yang, J.; Yang, Z.; Shi, C.; Wu, W.; Gao, R.; et al. The effect of perioperative probiotics treatment for colorectal cancer: Short-term outcomes of a randomized controlled trial. Oncotarget 2016, 7, 8432–8440. [Google Scholar] [CrossRef] [Green Version]
  51. Oguz, M.; Kerem, M.; Bedirli, A.; Mentes, B.B.; Sakrak, O.; Salman, B.; Bostanci, H. L-alanin-L-glutamine supplementation improves the outcome after colorectal surgery for cancer. Color. Dis. Off. J. Assoc. Coloproctology Great Br. Irel. 2007, 9, 515–520. [Google Scholar] [CrossRef]
  52. Zhu, M.W.; Tang, D.N.; Hou, J.; Wei, J.M.; Hua, B.; Sun, J.H.; Cui, H.Y. Impact of fish oil enriched total parenteral nutrition on elderly patients after colorectal cancer surgery. Chin. Med. J. 2012, 125, 178–181. [Google Scholar] [PubMed]
  53. Bakker, N.; van den Helder, R.S.; Stoutjesdijk, E.; van Pelt, J.; Houdijk, A.P.J. Effects of perioperative intravenous ω-3 fatty acids in colon cancer patients: A randomized, double-blind, placebo-controlled clinical trial. Am. J. Clin. Nutr. 2020, 111, 385–395. [Google Scholar] [CrossRef] [PubMed]
  54. Sorensen, L.S.; Thorlacius-Ussing, O.; Schmidt, E.B.; Rasmussen, H.H.; Lundbye-Christensen, S.; Calder, P.C.; Lindorff-Larsen, K. Randomized clinical trial of perioperative omega-3 fatty acid supplements in elective colorectal cancer surgery. Br. J. Surg. 2014, 101, 33–42. [Google Scholar] [CrossRef]
  55. Bajramagic, S.; Hodzic, E.; Mulabdic, A.; Holjan, S.; Smajlovic, S.V.; Rovcanin, A. Usage of Probiotics and its Clinical Significance at Surgically Treated Patients Sufferig from Colorectal Carcinoma. Med. Arch. 2019, 73, 316–320. [Google Scholar] [CrossRef]
  56. Kotzampassi, K.; Stavrou, G.; Damoraki, G.; Georgitsi, M.; Basdanis, G.; Tsaousi, G.; Giamarellos-Bourboulis, E.J. A Four-Probiotics Regimen Reduces Postoperative Complications After Colorectal Surgery: A Randomized, Double-Blind, Placebo-Controlled Study. World J. Surg. 2015, 39, 2776–2783. [Google Scholar] [CrossRef]
  57. Szefel, J.; Ślebioda, T.; Walczak, J.; Kruszewski, W.J.; Szajewski, M.; Ciesielski, M.; Stanisławowski, M.; Buczek, T.; Małgorzewicz, S.; Owczarzak, A.; et al. The effect of l-arginine supplementation and surgical trauma on the frequency of myeloid-derived suppressor cells and T lymphocytes in tumour and blood of colorectal cancer patients. Adv. Med. Sci. 2022, 67, 66–78. [Google Scholar] [CrossRef]
  58. Rotovnik Kozjek, N.; Kompan, L.; Soeters, P.; Oblak, I.; Mlakar Mastnak, D.; Možina, B.; Zadnik, V.; Anderluh, F.; Velenik, V. Oral glutamine supplementation during preoperative radiochemotherapy in patients with rectal cancer: A randomised double blinded, placebo controlled pilot study. Clin. Nutr. 2011, 30, 567–570. [Google Scholar] [CrossRef]
  59. Rotovnik Kozjek, N.; Kompan, L.; Žagar, T.; Mrevlje, Ž. Influence of enteral glutamine on inflammatory and hormonal response in patients with rectal cancer during preoperative radiochemotherapy. Eur. J. Clin. Nutr. 2017, 71, 671–673. [Google Scholar] [CrossRef] [PubMed]
  60. Decker-Baumann, C.; Buhl, K.; Frohmüller, S.; von Herbay, A.; Dueck, M.; Schlag, P.M. Reduction of chemotherapy-induced side-effects by parenteral glutamine supplementation in patients with metastatic colorectal cancer. Eur. J. Cancer 1999, 35, 202–207. [Google Scholar] [CrossRef] [PubMed]
  61. Morlion, B.J.; Stehle, P.; Wachtler, P.; Siedhoff, H.P.; Köller, M.; König, W.; Fürst, P.; Puchstein, C. Total parenteral nutrition with glutamine dipeptide after major abdominal surgery: A randomized, double-blind, controlled study. Ann. Surg. 1998, 227, 302–308. [Google Scholar] [CrossRef] [PubMed]
  62. Mocellin, M.C.; Pastore e Silva Jde, A.; Camargo Cde, Q.; Fabre, M.E.; Gevaerd, S.; Naliwaiko, K.; Moreno, Y.M.; Nunes, E.A.; Trindade, E.B. Fish oil decreases C-reactive protein/albumin ratio improving nutritional prognosis and plasma fatty acid profile in colorectal cancer patients. Lipids 2013, 48, 879–888. [Google Scholar] [CrossRef]
  63. Flesch, A.T.; Tonial, S.T.; Contu, P.C.; Damin, D.C. Perioperative synbiotics administration decreases postoperative infections in patients with colorectal cancer: A randomized, double-blind clinical trial. Rev. Do Col. Bras. De Cir. 2017, 44, 567–573. [Google Scholar] [CrossRef] [Green Version]
  64. Silva Jde, A.; Trindade, E.B.; Fabre, M.E.; Menegotto, V.M.; Gevaerd, S.; Buss Zda, S.; Frode, T.S. Fish oil supplement alters markers of inflammatory and nutritional status in colorectal cancer patients. Nutr. Cancer 2012, 64, 267–273. [Google Scholar] [CrossRef] [PubMed]
  65. Dikeocha, I.J.; Al-Kabsi, A.M.; Eid, E.E.M.; Hussin, S.; Alshawsh, M.A. Probiotics supplementation in patients with colorectal cancer: A systematic review of randomized controlled trials. Nutr. Rev. 2021, 80, 22–49. [Google Scholar] [CrossRef] [PubMed]
  66. Guo, Y.; Ma, B.; Li, X.; Hui, H.; Zhou, Y.; Li, N.; Xie, X. Omega-3 Polyunsaturated Fatty Acids can Reduce IL-6 and TNF Levels in Patients with Cancer. Br. J. Nutr. 2022, 7, 1–34. [Google Scholar] [CrossRef]
  67. Miller, L.J.; Douglas, C.; McCullough, F.S.; Stanworth, S.J.; Calder, P.C. Impact of enteral immunonutrition on infectious complications and immune and inflammatory markers in cancer patients undergoing chemotherapy: A systematic review of randomised controlled trials. Clin. Nutr. 2022, 41, 2135–2146. [Google Scholar] [CrossRef] [PubMed]
  68. Newsholme, P. Why is L-glutamine metabolism important to cells of the immune system in health, postinjury, surgery or infection? J. Nutr. 2001, 131, 2515S–2522S, discussion 2523S–2524S. [Google Scholar] [CrossRef] [Green Version]
  69. Taniguchi, K.; Karin, M. IL-6 and related cytokines as the critical lynchpins between inflammation and cancer. Semin. Immunol. 2014, 26, 54–74. [Google Scholar] [CrossRef]
  70. Tapia, G.; Valenzuela, R.; Espinosa, A.; Romanque, P.; Dossi, C.; Gonzalez-Mañán, D.; Videla, L.A.; D’Espessailles, A. N-3 long-chain PUFA supplementation prevents high fat diet induced mouse liver steatosis and inflammation in relation to PPAR-α upregulation and NF-κB DNA binding abrogation. Mol. Nutr. Food Res. 2014, 58, 1333–1341. [Google Scholar] [CrossRef]
  71. Weylandt, K.H.; Chiu, C.Y.; Gomolka, B.; Waechter, S.F.; Wiedenmann, B. Omega-3 fatty acids and their lipid mediators: Towards an understanding of resolvin and protectin formation. Prostaglandins Other Lipid Mediat. 2012, 97, 73–82. [Google Scholar] [CrossRef]
  72. Liang, M.; Wang, Z.; Li, H.; Liu, B.; Yang, L. l-Arginine prevents 4-hydroxy-2-nonenal accumulation and depresses inflammation via inhibiting NF-κB activation. J. Biochem. Mol. Toxicol. 2022, 36, e23087. [Google Scholar] [CrossRef] [PubMed]
  73. Nazarian, B.; Fazeli Moghadam, E.; Asbaghi, O.; Zeinali Khosroshahi, M.; Choghakhori, R.; Abbasnezhad, A. Effect of l-arginine supplementation on C-reactive protein and other inflammatory biomarkers: A systematic review and meta-analysis of randomized controlled trials. Complement. Ther. Med. 2019, 47, 102226. [Google Scholar] [CrossRef]
  74. Wan, G.Y.; Zheng, L.Y.; Li, H.Q.; Yuan, H.; Xue, H.; Zhang, X.Y. Effects of enteral nutritional rich in n-3 polyunsaturated fatty acids on the nutritional status of gastrointestinal cancer patients: A systematic review and meta-analysis. Eur. J. Clin. Nutr. 2020, 74, 220–230. [Google Scholar] [CrossRef] [PubMed]
  75. Brown, J.C.; Rosenthal, M.H.; Ma, C.; Zhang, S.; Nimeiri, H.S.; McCleary, N.J.; Abrams, T.A.; Yurgelun, M.B.; Cleary, J.M.; Rubinson, D.A.; et al. Effect of High-Dose vs Standard-Dose Vitamin D(3) Supplementation on Body Composition among Patients with Advanced or Metastatic Colorectal Cancer: A Randomized Trial. Cancers 2020, 12, 3451. [Google Scholar] [CrossRef]
  76. Cabrerizo, S.; Cuadras, D.; Gomez-Busto, F.; Artaza-Artabe, I.; Marín-Ciancas, F.; Malafarina, V. Serum albumin and health in older people: Review and meta analysis. Maturitas 2015, 81, 17–27. [Google Scholar] [CrossRef] [PubMed]
  77. Liu, W.; Zhang, L.; Xu, H.J.; Li, Y.; Hu, C.M.; Yang, J.Y.; Sun, M.Y. The Anti-Inflammatory Effects of Vitamin D in Tumorigenesis. Int. J. Mol. Sci. 2018, 19, 2736. [Google Scholar] [CrossRef] [Green Version]
  78. Li, K.; Huang, T.; Zheng, J.; Wu, K.; Li, D. Effect of marine-derived n-3 polyunsaturated fatty acids on C-reactive protein, interleukin 6 and tumor necrosis factor α: A meta-analysis. PLoS ONE 2014, 9, e88103. [Google Scholar] [CrossRef] [Green Version]
  79. Martens, P.J.; Gysemans, C.; Verstuyf, A.; Mathieu, A.C. Vitamin D’s Effect on Immune Function. Nutrients 2020, 12, 1248. [Google Scholar] [CrossRef]
  80. Gutiérrez, S.; Svahn, S.L.; Johansson, M.E. Effects of Omega-3 Fatty Acids on Immune Cells. Int. J. Mol. Sci. 2019, 20, 5028. [Google Scholar] [CrossRef] [Green Version]
  81. Soreide, K.; Berg, M.; Skudal, B.S.; Nedreboe, B.S. Advances in the understanding and treatment of colorectal cancer. Discov. Med. 2011, 12, 393–404. [Google Scholar]
  82. Vonlanthen, R.; Slankamenac, K.; Breitenstein, S.; Puhan, M.A.; Muller, M.K.; Hahnloser, D.; Hauri, D.; Graf, R.; Clavien, P.A. The impact of complications on costs of major surgical procedures: A cost analysis of 1200 patients. Ann. Surg. 2011, 254, 907–913. [Google Scholar] [CrossRef]
  83. Bai, H.; Li, Z.; Meng, Y.; Yu, Y.; Zhang, H.; Shen, D.; Chen, L. Effects of parenteral ω-3 fatty acid supplementation in postoperative gastrointestinal cancer on immune function and length of hospital stay: A systematic review and meta-analysis. Asia Pac. J. Clin. Nutr. 2018, 27, 121–128. [Google Scholar] [CrossRef]
  84. Cruzat, V.; Macedo Rogero, M.; Noel Keane, K.; Curi, R.; Newsholme, P. Glutamine: Metabolism and Immune Function, Supplementation and Clinical Translation. Nutrients 2018, 10, 1564. [Google Scholar] [CrossRef] [Green Version]
  85. Witte, M.B.; Barbul, A. Arginine physiology and its implication for wound healing. Wound Repair Regen. Off. Publ. Wound Heal. Soc. Eur. Tissue Repair Soc. 2003, 11, 419–423. [Google Scholar] [CrossRef]
  86. Malone-Povolny, M.J.; Maloney, S.E.; Schoenfisch, M.H. Nitric Oxide Therapy for Diabetic Wound Healing. Adv. Healthc. Mater. 2019, 8, e1801210. [Google Scholar] [CrossRef] [PubMed]
  87. Arribas-López, E.; Zand, N.; Ojo, O.; Snowden, M.J.; Kochhar, T. The Effect of Amino Acids on Wound Healing: A Systematic Review and Meta-Analysis on Arginine and Glutamine. Nutrients 2021, 13, 2498. [Google Scholar] [CrossRef]
  88. Li, H.Y.; Zhou, D.D.; Gan, R.Y.; Huang, S.Y.; Zhao, C.N.; Shang, A.; Xu, X.Y.; Li, H.B. Effects and Mechanisms of Probiotics, Prebiotics, Synbiotics, and Postbiotics on Metabolic Diseases Targeting Gut Microbiota: A Narrative Review. Nutrients 2021, 13, 3211. [Google Scholar] [CrossRef]
  89. Dang, A.T.; Marsland, B.J. Microbes, metabolites, and the gut-lung axis. Mucosal Immunol. 2019, 12, 843–850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Markowiak, P.; Śliżewska, K. Effects of Probiotics, Prebiotics, and Synbiotics on Human Health. Nutrients 2017, 9, 1021. [Google Scholar] [CrossRef] [PubMed]
  91. Zhang, C.X.; Wang, H.Y.; Chen, T.X. Interactions between Intestinal Microflora/Probiotics and the Immune System. BioMed Res. Int. 2019, 2019, 6764919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. PRISMA flow diagram for search and selection of eligible studies included in the network meta-analysis.
Figure 1. PRISMA flow diagram for search and selection of eligible studies included in the network meta-analysis.
Nutrients 15 02772 g001
Figure 2. Network plot and results of network meta-analysis. (A) Network plot for tumor necrosis factor-α (TNF-α, pg/mL). (B) Relative effects of different nutritional supplements on TNF-α. Notes: estimates are presented as mean difference (MD) and 95% credible intervals (CrI; in parentheses). Comparisons between treatments should be read from left to right. The estimate of supplementation effectiveness is located at the intersection of the column-defining supplementation and the row-defining supplementation. Significant results are presented in bold. Combined with omega-3 fatty acids and probiotics: omega3 + probiotics; combined with omega-3 fatty acids and vitamin D: omega3 + vitamind; omega-3 fatty acids: omega3; vitamin D: vitamind.
Figure 2. Network plot and results of network meta-analysis. (A) Network plot for tumor necrosis factor-α (TNF-α, pg/mL). (B) Relative effects of different nutritional supplements on TNF-α. Notes: estimates are presented as mean difference (MD) and 95% credible intervals (CrI; in parentheses). Comparisons between treatments should be read from left to right. The estimate of supplementation effectiveness is located at the intersection of the column-defining supplementation and the row-defining supplementation. Significant results are presented in bold. Combined with omega-3 fatty acids and probiotics: omega3 + probiotics; combined with omega-3 fatty acids and vitamin D: omega3 + vitamind; omega-3 fatty acids: omega3; vitamin D: vitamind.
Nutrients 15 02772 g002
Figure 3. Network plot and results of network meta-analysis. (A) Network plot for interleukin-6 (IL-6, pg/mL). (B) Relative effects of different nutritional supplements on IL-6. Notes: estimates are presented as mean difference (MD) and 95% credible intervals (CrI; in parentheses). Comparisons between treatments should be read from left to right. The estimate of supplementation effectiveness is located at the intersection of the column-defining supplementation and the row-defining supplementation. Significant results are presented in bold. Combined with omega-3 fatty acids and arginine: omega3 + arginine; combined with omega-3 fatty acids and probiotics: omega3 + probiotics; combined with omega-3 fatty acids and vitamin D: omega-3 + vitamind; omega-3 fatty acids: omega3; vitamin D: vitamind.
Figure 3. Network plot and results of network meta-analysis. (A) Network plot for interleukin-6 (IL-6, pg/mL). (B) Relative effects of different nutritional supplements on IL-6. Notes: estimates are presented as mean difference (MD) and 95% credible intervals (CrI; in parentheses). Comparisons between treatments should be read from left to right. The estimate of supplementation effectiveness is located at the intersection of the column-defining supplementation and the row-defining supplementation. Significant results are presented in bold. Combined with omega-3 fatty acids and arginine: omega3 + arginine; combined with omega-3 fatty acids and probiotics: omega3 + probiotics; combined with omega-3 fatty acids and vitamin D: omega-3 + vitamind; omega-3 fatty acids: omega3; vitamin D: vitamind.
Nutrients 15 02772 g003
Figure 4. Network plot and results of network meta-analysis. (A) Network plot for the length of hospital stay (LOH). (B) Relative effects of different nutritional supplements on the LOH. Notes: estimates are presented as mean difference (MD) and 95% credible intervals (CrI; in parentheses). Comparisons between treatments should be read from left to right. The estimate of supplementation effectiveness is located at the intersection of the column-defining supplementation and the row-defining supplementation. Significant results are presented in bold. Combined with omega-3 fatty acids and arginine: omega3 + arginine; combined with omega-3 fatty acids and glutamine: omega3 + glutamine; omega-3 fatty acids: omega3.
Figure 4. Network plot and results of network meta-analysis. (A) Network plot for the length of hospital stay (LOH). (B) Relative effects of different nutritional supplements on the LOH. Notes: estimates are presented as mean difference (MD) and 95% credible intervals (CrI; in parentheses). Comparisons between treatments should be read from left to right. The estimate of supplementation effectiveness is located at the intersection of the column-defining supplementation and the row-defining supplementation. Significant results are presented in bold. Combined with omega-3 fatty acids and arginine: omega3 + arginine; combined with omega-3 fatty acids and glutamine: omega3 + glutamine; omega-3 fatty acids: omega3.
Nutrients 15 02772 g004
Figure 5. Network plot and results of network meta-analysis. (A) Network plot for the incidence of wound infections (WI). (B) Relative effects of different nutritional supplements on the incidence of WI. Notes: estimates are presented as risk ratio (RR) and 95% credible intervals (CrI; in parentheses). Comparisons between treatments should be read from left to right. The estimate of supplementation effectiveness is located at the intersection of the column-defining supplementation and the row-defining supplementation. Significant results are presented in bold. Combined with omega-3 fatty acids and arginine: omega3 + arginine; omega-3 fatty acids: omega3.
Figure 5. Network plot and results of network meta-analysis. (A) Network plot for the incidence of wound infections (WI). (B) Relative effects of different nutritional supplements on the incidence of WI. Notes: estimates are presented as risk ratio (RR) and 95% credible intervals (CrI; in parentheses). Comparisons between treatments should be read from left to right. The estimate of supplementation effectiveness is located at the intersection of the column-defining supplementation and the row-defining supplementation. Significant results are presented in bold. Combined with omega-3 fatty acids and arginine: omega3 + arginine; omega-3 fatty acids: omega3.
Nutrients 15 02772 g005
Figure 6. Network plot and results of network meta-analysis. (A) Network plot for the incidence of pneumonia. (B) Relative effects of different nutritional supplements on the incidence of pneumonia. Notes: estimates are presented as risk ratio (RR) and 95% credible intervals (CrI; in parentheses). Comparisons between treatments should be read from left to right. The estimate of supplementation effectiveness is located at the intersection of the column-defining supplementation and the row-defining supplementation. Significant results are presented in bold. Omega-3 fatty acids: omega3.
Figure 6. Network plot and results of network meta-analysis. (A) Network plot for the incidence of pneumonia. (B) Relative effects of different nutritional supplements on the incidence of pneumonia. Notes: estimates are presented as risk ratio (RR) and 95% credible intervals (CrI; in parentheses). Comparisons between treatments should be read from left to right. The estimate of supplementation effectiveness is located at the intersection of the column-defining supplementation and the row-defining supplementation. Significant results are presented in bold. Omega-3 fatty acids: omega3.
Nutrients 15 02772 g006
Table 1. Main characteristics of the studies included in the network meta-analysis.
Table 1. Main characteristics of the studies included in the network meta-analysis.
Author (Year)NationalityParticipant TypesSample Size (n)Age (Years)InterventionRouteOutcomesJadad Score
TreatmentControlTreatmentControl
Aliyazicioglu et al. (2013) [37]TurkeyCRC surgery patients8/8/101058.0 ± 15.6/
63.9 ± 17.2/
59.9 ± 15.1
56.3 ± 14.3glutamine/omega3 fatty acid/
glutamine + omega 3 fatty acid/glutamine:0.3–0.4 g/kg/d, omega 3 fatty acid: 0.1–0.2 g/kg/d, 2 postoperative days to 7 postoperative days
PNg4
Bakker et al. (2020) [53]NetherlandsCRC surgery patients182368.2475 ± 8.84968.6415 ± 8.6919omega 3 fatty acid (fish oil)/0.2 g/kg/d, 1 preoperative day and 1 postoperative dayPNa, c, h, i, j, k6
Sorensen et al. (2014) [54]DenmarkCRC surgery patient747469 ± 1171 ± 10omega 3 fatty acid (EPA:2.0 g/d and DHA:1.0 g/d)/400 mL/d, 7 preoperative days and 7 postoperative daysOralh, i, j, k7
Liang et al. (2008) [36]ChinaCRC surgery patients202155.80 ± 10.1059.19 ± 10.61omega 3 fatty acid/0.2 g/kg/d, 7 postoperative daysPNb, c, g7
Esfahani et al. (2016) [38]IranCRC chemotherapy
patients
363554.14 ± 10.5353.40 ± 15.70omega 3 fatty acid (54% DHA, 10% EPA)/1920 mg/d,
1 month
Oralb, c7
Mocellin et al. (2013) [62]BrazilCRC chemotherapy
patients
6555.2 ± 7.753.6 ± 12.9omega 3 fatty acid (90 mg EPA and 60 mg DHA)/2 g/d, 9 weeksOrala, b, d, e, f4
Haidari et al. (2020) [19]IranCRC chemotherapy
patients
20/21/202056.75 ± 10.60/
56.90 ± 12.45/
57.15 ± 10.17
59.90 ± 8.75omega 3 fatty acid (54 mg EPA, 250 mg DHA, 26 mg other omega-3 fatty acids)/
Vitamin D/
omega 3 fatty acid (54 mg EPA, 250 mg DHA, 26 mg other omega-3 fatty acids) + vitamin D/vitamin D: 50,000 IU soft gel/w, omega3 fatty acid: 660 mg/d, 8 weeks
Orala, b, c, d, e, f4
Golkhalkhali et al. (2017) [39]MalaysiaCRC chemotherapy
patients
7070≤56:20
57–66: 22
≥67:23
≤56:28
57–66:19
≥67:19
omega 3 fatty acid (700 mg EPA) + probiotics (L. acidophilus, Lactobacillus lactis, Bififidobacterium
bififidum, Bififidobacterium longum, Bififidobacterium infantis)/omega 3 fatty acid: 2 g/d + probiotics:2 sachets/d, 8 weeks
Orala, b, c, e, f7
Bajramagic et al. (2019) [55]SarajevoCRC surgery patients3939NRNRprobiotics (Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus plantarum, Lactobacillus rhamnosus, Bifidobacterium lactis, Bifidobacterium bifidum, Bifidobacterium breve, Streptococcusthermophilus)/4 capsules/d for 30 days, 1 capsule/d for 2 weeks in each month to 1 yearOrali, j4
Tan et al. (2016) [40]MalaysiaCRC surgery patients202064.3 ± 14.568.4 ± 11.9probiotics (Lactobacillus acidophilus, Lactobacillus lactis, Bififidobacterium bififidum, Bififidobacterium longum, and Bififidobacterium infantis)/2 sachets/d, 7 preoperative daysOralg, i, j, k6
Zhang et al. (2012) [41]ChinaCRC surgery patients303067.14 ± 10.2962.09 ± 8.82probiotics (B longum, L acidophilus, and Enterococcus faecalis)/0.63 g/d, 5 preoperative days to 3 preoperative daysOralc, i, j, k5
Flesch et al. (2017) [63]BrazilCRC surgery patients494264.561.1probiotics (Lactobacillus acidophilus, Lactobacillus rhamnosus, Lactobacillus paracasei, and Bifidobacterium lactis)/12 g/d, 5 preoperative days and 14 postoperative daysOrali, k4
Kotzampassi et al. (2015) [56]GreeceCRC surgery patients848065.9 ±11.566.4 ± 11.9probiotics (Lactobacillus acidophilus LA-5, Lactobacillus plantarum, Bififidobacterium lactis BB-12, and Saccharomyces boulardii)/2 capsules/d, on the day of operation and 14 postoperative daysOral/
EN
h, i, j, k4
Sadahiro et al. (2014) [42]JapanCRC surgery patients1009567 ± 966 ± 12probiotics (Bifidobacteria)/9 tablets/d, 7 preoperative days and 5 postoperative days to 15 postoperative daysOrali, j4
Xu et al. (2019) [43]ChinaCRC surgery patients303061.03 ± 15.2862.35 ± 13.71probiotics (bifidus-triple viable preparation)/NA,7 preoperative daysOrala, i4
Polakowski et al. (2019) [24]BrazilCRC surgery patients363760.9 ± 6.758.9 ± 6.3probiotics (fructooligosaccharide, Lactobacillus acidophilus NCFM, L. rhamnosus HN001, L. casei LPC-37, and Bififidobacterium lactis HN019)/2 times/d, 7 preoperative daysOrala, c, g7
Mizuta et al.(2016) [44]JapanCRC surgery patients312968.9 ± 10.471.2 ± 9.5probiotics (B. longum)/2 g/d, 7–14 preoperative days and 14 postoperative daysOralc, d, g, i, j7
Komatsu et al. (2016) [45]JapanCRC surgery patients16819466.7 ± 11.667.7 ± 10.7probiotics (Lactobacillus casei, galactooligosaccharides, and Bifidobacterium breve)/NA, 7–11 preoperative days and 2–7 postoperative daysOrali, j5
Radvar et al. (2020) [46]IranCRC chemoradiotherapy patients232357.58 ± 12.7862.89 ± 13.93probiotics (Lactobacillus casei PXN 37, Lactobacillus rhamnosus PXN 54, Streptococcus thermophilus PXN 66, Bififidobacteriumbreve PXN 25, Lactobacillus acidophilus
PXN 35, Bififidobacteriumlongum PXN 30, Lactobacillus bulgaricus PXN 39, Fructooligosaccharide, magnesium stearate, and hydroxypropyl methyl- cellulose)/2 times/d, 6 weeks
Orale, f7
Xie et al. (2019) [47]ChinaCRC surgery patients666962.62 ± 9.62760.29 ± 9.54probiotics (fructooligosaccharide, xylooligosaccharide, polydextrose, and resistant dextrin)/30 g/d, 7 preoperative daysOrald6
Liu et al. (2015) [48]ChinaCRC surgery patients666865.62 ± 18.1860.16 ± 16.20probiotics (Lactobacillus plantarum, Lactobacillus acidophilus-11, and Bifido-bacterium longum-88)/2 g/d,
6 preoperative days and 10 postoperative days
Oralg, h, i, k7
Liu et al. (2011) [49]ChinaCRC surgery patient505065.3 ± 11.065.7 ± 9.9probiotics (Lactobacillus plantarum, Lactobacillus acidophilus-11, and Bifido-bacterium longum-88)/2 g/d,
6 preoperative days and 10 postoperative days
Oralg, h, i, k7
Yang et al. (2016) [50]ChinaCRC surgery patients303063.90 ± 12.2562.17 ± 11.06probiotics (Bifidobacterium longum, Lactobacillus
acidophilus, and Enterococcus faecalis)/6 g/d, 5 preoperative days and 7 postoperative days
Orald, g, h, i, j, k6
Szefel et al. (2022) [57]PolandCRC surgery patients283768.9 ± 10.968.7 ± 9.3arginine/20 capsules/d, 9 preoperative daysOrala7
Braga et al. (2002) [27]ItalyCRC surgery patients505063.0 ± 8.162.2 ± 10.4arginine (12.5 g/L) + omega 3 fatty acid (3.3 g/L)/1 L/d, 5 preoperative daysOral/
EN
c, g, h, i, j5
Rotovnik et al. (2011) [58]SloveniaCRC chemoradiotherapy patients141960.5 ± 14.263.6 ± 10.12glutamine/30 g/d, 5 weeksOrala, c7
Oguz et al. (2007) [51]TurkeyCRC surgery patients575252 ± 1257 ± 17glutamine/1 g/kg/day, 5 preoperative days and 5 postoperative daysPNg, h, i, j,4
Rotovnik et al. (2017) [59]SloveniaCRC chemoradiotherapy patients334060.8 ±11.961.4 ± 9.9glutamine/30 g/d, 5 weeksOralc6
Cui et al. (2014) [22]ChinaCRC surgery patients20
2055 ± 10.856 ± 10.7glutamine/0.5 g/kg, 1 preoperative day and the day of surgeryPNb, g6
Decker et al. (1999) [60]GermanCRC chemotherapy
patients
121256.1 ± 9.658.4 ± 7.2glutamine/14 ± 22 g/d, 18 daysPNe4
Zaharuddin et al. (2019) [20]MalaysiaCRC surgery patients2725NRNRprobiotics (Lactobacillus and Bifidobacteria)/2 times/d, 6 postoperative monthsOralb, c5
Morlion et al. (1998) [61]GermanCRC surgery patients151367.1 ± 10.768.2.1 ± 12.5glutamine/0.3 g/kg/d/5 postoperative daysPNg6
Silva et al. (2012) [64]BrazilCRC chemotherapy
patients
111250.1 ± 8.254.3 ± 9.3omega 3 fatty acid/0.6 g/d/9 weeksOrala, c, d, e, f4
Zhu et al. (2012) [52]ChinaCRC surgery patients292869.8 ± 10.570.8 ± 6.4omega 3 fatty acid/0.2 g/kg/d/7 postoperative daysPNb, c, g, h, i7
Note: NR, not report; PL, placebo; PN, parenteral nutrition; EN, enteral nutrition; CRC, Colorectal cancer. a, C-reactive protein; b, tumor necrosis factor- α; c, interleukin-6; d, albumin; e, weight; f, body mass index; g, the length of hospital stays; h, urinary infections; i, wound infections; j, anastomotic leaks; k, pneumonia.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ye, J.; Hu, Y.; Chen, X.; Chang, C.; Li, K. Comparative Effects of Different Nutritional Supplements on Inflammation, Nutritional Status, and Clinical Outcomes in Colorectal Cancer Patients: A Systematic Review and Network Meta-Analysis. Nutrients 2023, 15, 2772. https://doi.org/10.3390/nu15122772

AMA Style

Ye J, Hu Y, Chen X, Chang C, Li K. Comparative Effects of Different Nutritional Supplements on Inflammation, Nutritional Status, and Clinical Outcomes in Colorectal Cancer Patients: A Systematic Review and Network Meta-Analysis. Nutrients. 2023; 15(12):2772. https://doi.org/10.3390/nu15122772

Chicago/Turabian Style

Ye, Jiayi, Yanjie Hu, Xinrong Chen, Chengting Chang, and Ka Li. 2023. "Comparative Effects of Different Nutritional Supplements on Inflammation, Nutritional Status, and Clinical Outcomes in Colorectal Cancer Patients: A Systematic Review and Network Meta-Analysis" Nutrients 15, no. 12: 2772. https://doi.org/10.3390/nu15122772

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop