Next Article in Journal
Intergenerational Hyperglycemia Impairs Mitochondrial Function and Follicular Development and Causes Oxidative Stress in Rat Ovaries Independent of the Consumption of a High-Fat Diet
Previous Article in Journal
Adherence to Data-Driven Dietary Patterns and Lung Cancer Risk: A Systematic Review and Dose–Response Meta-Analysis
Previous Article in Special Issue
Antioxidant and Antiproliferative Activities of Several Garlic Forms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Synergistic Strategies for Gastrointestinal Cancer Care: Unveiling the Benefits of Immunonutrition and Microbiota Modulation

1
Department of Experimental and Clinical Medicine, University of Florence, 50134 Firenze, Italy
2
Department of Nutrition and Dietetics, Gazi University, 06560 Ankara, Turkey
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(20), 4408; https://doi.org/10.3390/nu15204408
Submission received: 20 September 2023 / Revised: 12 October 2023 / Accepted: 16 October 2023 / Published: 17 October 2023

Abstract

:
Gastrointestinal (GI) cancers are a group of highly prevalent malignant tumors affecting the gastrointestinal tract. Globally, one in four cancer cases and one in three cancer deaths are estimated to be GI cancers. They can alter digestive and absorption functions, leading to severe malnutrition which may worsen the prognosis of the patients. Therefore, nutritional intervention and monitoring play a fundamental role in managing metabolic alterations and cancer symptoms, as well as minimizing side effects and increasing the effectiveness of chemotherapy. In this scenario, the use of immunonutrients that are able to modulate the immune system and the modification/regulation of the gut microbiota composition have gained attention as a possible strategy to improve the conditions of these patients. The complex interaction between nutrients and microbiota might contribute to maintaining the homeostasis of each individual’s immune system; therefore, concurrent use of specific nutrients in combination with traditional cancer treatments may synergistically improve the overall care of GI cancer patients. This work aims to review and discuss the role of immunonutrition and microbiota modulation in improving nutritional status, postoperative recovery, and response to therapies in patients with GI cancer.

1. Introduction

Gastrointestinal (GI) cancers, a collective term encompassing a group of malignancies originating in various parts of the gastrointestinal tract, account for around 20% of newly diagnosed cancers and are responsible for over 25% of cancer-related deaths worldwide [1]. GI cancers differ in their potential to disrupt normal digestive functions and can have significant implications for a patient’s overall health and well-being, making early detection and appropriate treatment essential for optimal outcomes. The substantial global impact of GI cancers underscores the critical necessity to address the multifaceted challenges faced by these patients. Nutritional intervention and monitoring play pivotal roles in the comprehensive care of GI cancer patients. Managing the nutritional needs of these patients is essential not only to address the metabolic alterations associated with tumors but also to mitigate the adverse effects of cancer treatments. Adequate nutritional support can help optimize treatment outcomes, enhance the patient’s quality of life (QoL), and improve their overall prognosis. This review attempts to investigate the promising avenues of immunonutrition and microbiota modulation as innovative approaches to improve the nutritional status and overall well-being of GI cancer patients. By addressing these aspects, the goal is to offer insights into strategies that can complement traditional cancer treatments and enhance the holistic care of this cancer population.

2. Nutritional Status of GI Cancer Patients

In the realm of GI cancer care, understanding the nutritional status of patients is of paramount importance. Patients diagnosed with cancer frequently experience malnutrition as their tumors progress. The prevalence of malnutrition among cancer patients varies, ranging from 20% to 70%; in patients diagnosed with upper GI cancer, its prevalence can escalate to as high as 60.2% [2,3]. This malnutrition has been linked to increased postoperative complications and a decline in overall QoL. Malnutrition in cancer patients is a complex issue and can be classified into various categories, including anorexia, sarcopenia, and cachexia. These definitions are meant to help clinicians identify and treat metabolic and nutritional problems associated with cancer. Anorexia (loss of appetite) is quite common in cancer and, according to Abraham et al., 69% of patients newly diagnosed with gastric or gastroesophageal junction (GOJ) experience anorexia [4]. The primary cause of anorexia is often an increase in pro-inflammatory cytokines or an increase in lactate which can, in turn, modulate central nervous system neurotransmitter cascades [5,6]. Sarcopenia is characterized by diminished muscle strength, reduced muscle mass or quality, and decreased physical performance [7]. In GI cancer patients, among whom the prevalence of sarcopenia exceeds 40% [8], monitoring this condition is crucial as it significantly influences postoperative outcomes and is associated with a higher risk of complications [9,10]. Cancer cachexia, a syndrome characterized by loss of weight, muscle, and fat mass that cannot be reversed through conventional nutritional interventions, creates a proinflammatory environment leading to heightened energy expenditure [11]. The incidence of cancer cachexia in GI cancer patients varies widely, ranging from 15% in prostate cancer to as high as 90% in pancreatic cancer, with an overall occurrence between 40% and 80% [12,13]. GI cancer patients, in general, are at an elevated risk of experiencing cancer cachexia, often diagnosed too late for effective prevention or treatment of muscle and weight loss, resulting in heightened morbidity and mortality, reduced QoL, and suboptimal therapeutic outcomes [14].
Malnutrition in GI cancer patients has multiple underlying causes. The metabolic alterations induced by tumors and the adverse effects of cancer treatments can lead to a spectrum of nutritional challenges, including decreased appetite, difficulty swallowing, taste and smell changes, weight loss, fatigue, and a decline of QoL [2,15,16]. GI mucosa is sensitive to chemotherapy cytotoxicity: DNA damage, apoptosis, and inflammation of the healthy mucosa disrupt the GI architecture [17] by decreasing the mucosal area available for nutrient absorption and impairing the immune response [18]. This condition can be defined as gastrointestinal mucositis (GI-M) [19,20]. Consequently, patients who develop GI-M are at a significantly higher risk of secondary complications, especially nutritional deficiencies [21]. In addition, psychological distress and anxiety significantly influence dietary intake [22]. The connection between malnutrition and psychological distress has been evidenced in cancer patients. Those experiencing weight loss and other malnutrition-related symptoms often report heightened levels of psychological distress, manifested as increased fatigue, insomnia, heightened anxiety, and depression, ultimately exacerbating the progression of their illness [23]. When considered collectively, these factors can significantly impact the nutritional status of GI cancer patients and individuals who have undergone surgery as well as those currently undergoing radiotherapy and chemotherapy [24] (Figure 1).

3. Nutritional Strategies for GI Cancers Patients Care

Clinical management guidelines emphasize the importance of screening and assessing malnutrition in cancer patients. They support the provision of adequate and effective nutritional therapy, particularly for people who face problems with nutrition [2,25]. While the precise timing for initiating nutrition support is not yet fully defined, it is recommended to commence intervention before malnutrition becomes well established [2]. In cases where patients are severely malnourished and actively undergoing treatment, nutritional support should be implemented immediately to address their nutritional needs. GI cancer patients undergoing radiotherapy can benefit from early and intensive nutritional intervention, which will improve their nutritional status and QoL [26,27]. Nutritional therapy in GI patients should, at first, comprise an adequate calorie and protein intake, essential to preserve lean body mass, promote wound healing, and support the body’s immune function. High-calorie, high-protein foods should be included in the diet, along with small, frequent meals [28,29]. Secondly, it is important to manage digestive symptoms, since GI cancers and their post-surgical treatments can lead to nausea, vomiting, diarrhea, constipation, and loss of appetite [30].
The dose of nutritional therapy is based on the patient’s energy and nutrient requirements. The recommended energy intake for cancer patients is 25–30 kcal/kg/day. Protein intake should be above 1 g/kg/day and above 1.5 g/kg/day if possible [15,31].
Effectively managing symptoms is a crucial aspect of cancer care from the time of diagnosis to treatment. These factors not only impact the well-being of GI cancer patients but also have substantial implications for treatment outcomes.
Avoiding spicy or greasy foods, eating smaller portions, and consuming more frequent meals of easily digestible foods are dietary modifications that can help relieve these symptoms and improve nutrient absorption. Vomiting or diarrhea may also result in fluid loss; therefore, adequate hydration to support overall bodily functions should be implemented. Drinking water, clear broths, herbal teas, and consuming hydrating foods like fruits and vegetables can help meet hydration needs [32]. Certain micronutrients may be compromised in GI cancer patients due to reduced intake or absorption. Then, the healthcare team may recommend specific supplements to address deficiencies, such as vitamins (e.g., vitamin D, B vitamins) and minerals (e.g., iron, zinc), based on individual needs [33,34] (Figure 2). At last, in cases where oral food intake is insufficient or not possible, artificial nutritional may be required [35]. This includes enteral nutrition (EN) (delivery of nutrients through the GI tract, either orally or via a feeding tube), parenteral nutrition (PN) (delivery of nutrients directly into the bloodstream), nutrition counseling, and oral nutritional supplements (ONS). If the patient can eat but is malnourished or at risk of malnutrition, interventions to increase food intake or ONS are recommended. If the patient is undernourished, medical nutrition (EN or PN) is indicated [15]. After hospitalization, or when palliation is the main purpose of nutritional intervention, EN should be preferred, except when intestinal obstruction, ileus, severe shock, intestinal ischemia, high-flow fistula, or severe intestinal bleeding occurs [10]. Optimal preoperative nutritional support for at least 10 days has been reported to reduce the risk of postoperative surgical site infection (SSI) in patients with gastric cancer [36]. Clinicians should plan individualized nutritional interventions during screening, evaluation, and treatment processes and should not hesitate to prefer the EN and/or PN route when nutritional goals cannot be achieved with an oral diet alone [37] (Figure 2).
Preoperative nutrition and exercise intervention have been reported to provide perioperative functional improvement in esophagogastric cancer surgery patients [38]. According to the ESPEN guideline, routine postoperative nutritional support should be considered for surgical cancer patients (especially those undergoing upper GI cancer surgery) at moderate or severe nutritional risk [15]. Postponing a regular oral diet after major surgery is associated with an increased rate of infectious complications and a longer recovery [39].
Several studies have highlighted the critical role of adequate nutrition in mitigating postoperative complications, maintaining immune function, reducing treatment toxicity, enhancing overall survival rates, shortening length of stay (LOS) in hospital, and facilitating the timely administration of adjuvant oncologic therapy [40,41,42,43,44].
Following surgery, colorectal cancer (CRC) patients receiving oral nutritional supplements (ONS) demonstrated notable enhancements; however, no significant alterations were observed in metrics like body weight and body mass index (BMI) [45]. Nevertheless, a randomized trial showed that the use of ONS administration after GI cancer surgery may have positive outcomes on patients’ body weight and BMI [46]. Likewise, patients administered with ONS three months following GC surgery experienced significantly less reduction in body weight and exhibited a notably higher BMI compared with patients who received nutrition counseling alone [45]. A prospective randomized controlled study showed that patients experienced a decline in nutritional status after discharge, highlighting the critical role of postoperative nutritional supplementation in enhancing nutritional status, QoL, and reducing morbidity among surgical patients [47]. The European Society for Clinical Nutrition and Metabolism (ESPEN) practice guideline recommends an enhanced recovery after surgery (ERAS) program for all cancer patients undergoing surgery [15]. ERAS is an evidence-based, multicomponent perioperative protocol that aims at reducing stress and promoting a return to function [48]. Within the scope of this program, each patient should be assessed for malnutrition, and additional nutritional support should be applied if necessary. Nutritional components of ERAS are avoiding fasting, preoperative fluid, and carbohydrate overload, and recommencement of oral diet on the first postoperative day. The aim is to minimize the metabolic response to surgery [15].
Considering all these data, it becomes crucial that each step of the process for GI cancer surgery patients, starting from preoperative nutritional screening and extending to post-discharge nutritional support, is overseen by a healthcare professional. Adequate nutritional care should be meticulously planned for each patient (Figure 2).
However, long-term prospective studies of GI cancer patients’ preoperative and postoperative nutritional care in larger populations are necessary.

4. Immunonutrition and Gut Microbiota Modulation

Cancer and its treatment can weaken the immune system, making patients more susceptible to infections and impairing their ability to combat the disease. Moreover, GI surgical procedures trigger a natural inflammatory response in the body, which is intended to be protective and create an environment conducive to recovery. This response primarily aims to facilitate energy production, restore cardiovascular balance, promote tissue repair and wound healing, and ultimately ensure the state of well-being of the patient. However, there are instances when this inflammatory response becomes dysregulated, leading to the release of proinflammatory cytokines, endothelial dysfunction, glycocalyx damage, activation of neutrophils, and subsequent damage to tissues and multiple organ systems [49]. In such cases, patients undergoing GI surgery may experience postoperative complications such as anastomotic dehiscence and surgical site infections, or even face severe consequences due to an amplified and uncontrolled inflammatory reaction [50].
Immunonutrition in GI cancer care is designed for complementing traditional cancer treatments by supporting the immune system, boosting the host’s cancer-related immune response, reducing inflammation, promoting tissue repair, and improving overall nutritional status [51].
Interestingly, inflammation is closely linked to alterations in the gut microbiota (GM) and their metabolites, particularly short-chain fatty acids (SCFAs). As a counterbalance to the immune response, the human GM appears to play a significant role in the development of post-surgery complications [52]. Furthermore, the GM is integral in the absorption, storage, and utilization of energy derived from dietary intake [53]. It contributes to food regulation intake by influencing hormones related to metabolic function and brain regions associated with eating behavior [54]. In addition, it has beeen proven that the GM plays critical roles in protecting the integrity of the GI mucosa and the maintenance of its homeostasis [55]. GI cancers and also their treatments, such as chemotherapy, have been consistently demonstrated to induce changes in the GM composition and functions [56], contributing to GI-M severity and to malnutrition onset [57,58]. In this context, dietary interventions aimed at fostering a healthy GM before therapy and enhancing its resilience during and after therapy show potential as therapeutic strategies for GI-M and its related symptoms [59,60].
The maintenance of a functional gut barrier and of host homeostasis is granted by the action of beneficial bacteria like Bifidobacterium spp., Faecalibacterium prausnitzii, Lactobacillus spp., and the production of SCFAs, particularly butyrate. SCFAs stimulate the regeneration of epithelial cells, the production of mucus and antimicrobial peptides, and modulate T regulatory (Treg) cells [61,62]. Dendritic cells located in the lamina propria release transforming growth factor-β (TGF-β) in response to commensal antigens, activating Treg cells to secrete interleukin-10 (IL-10) and TGF-β, promoting a more immune-tolerant phenotype [63]. In addition, dietary and microbiota-derived ligands of the aryl hydrocarbon receptor (AhR) stimulate innate lymphoid cell 3 (ILC3) to produce IL-22, which plays a vital role in preserving intestinal barrier function [64]. Another hallmark of intestinal balance is a thicker mucus layer, acting as a barrier between luminal bacteria and epithelial cells.
When dysbiosis occurs, the lower abundance of beneficial bacteria and higher abundance of pathobionts (i.e., Clostridium difficile and Escherichia coli) promote production of inflammatory factors such as radical oxygen species (ROS), nitric oxide (NO), proinflammatory cytokines, and cyclo-oxygenase 2 (COX-2) [65,66]. The decrease in thickness of the mucus layer and cellular tight junction expression results in a compromised intestinal barrier function, allowing bacterial products, like lipopolysaccharides (LPSs), to leak from the intestinal lumen into the lamina propria. LPSs bind to toll-like receptors (TLRs), triggering macrophages to generate tumor necrosis factor-α (TNF-α). TNF-α promotes the proliferation of T helper cell type 1 (Th1) and the release of pro-inflammatory cytokines, including TNF-α and interferon-γ (IFN-γ), ultimately causing inflammation. This inflammatory process further undermines the integrity of the intestinal barrier. Additionally, the reduction in IL-10-producing Treg cells contributes to the inflammation within the intestine [67]. Finally, dysbiosis may increase colonic epithelial cells’ exposure to carcinogens [68]. Notably, the GM influences the host’s response to cancer therapy. Germ-free and antibiotic-treated mice showed reduced responses to immunotherapy and chemotherapy by CpG oligonucleotides, due to impaired function of myeloid-derived cells in the tumor microenvironment [69]. On the other hand, Barnesiella intestinihominis has been reported to have an adjuvant effect on cyclophosphamide (CTX)-induced tumor immunity by promoting infiltration of IFN-γ-producing γδT cells in cancer lesions [70].
Finally, studies report that the GM is involved in the management of cancer, as the composition of the GM can modulate the effect of anticancer drugs. Neoadjuvant chemoradiotherapy (nCRT) has become a standard treatment for locally advanced rectal cancer (LARC), with only 15–27% of patients achieving a pathological complete response and 20–40% achieving little to no response. Aiming to elucidate the mechanism underlying the response of LARC to nCRT, Teng et al. showed that GM-mediated nucleotide synthesis can modulate the response of LARC patients to nCRT. Multi-omics data integration showed that Bacteroides vulgatus-mediated nucleotide biosynthesis was associated with nCRT resistance in LARC patients, and non-responsive tumors were characterized by up-regulation of genes related to DNA repair and nucleoside transport [71].
Given these results, a more balanced microbiota obtained through microbiota manipulation may have positive contributions in preventing cancer formation and increasing response to medical treatment [72,73]. Therefore, managing the immune response during the post-surgical period, both in the short and long term, with a focus on the interaction between GM and inflammation (a bidirectional signaling axis regulating immune response, GI balance, and body weight through appetite control, energy storage, and expenditure), presents an advantageous strategy.

4.1. Immunonutrients Supplementation

Among nutritional interventions, immune-enhancing nutrient formulas, also called immunonutrients, can be supplemented via EN, PN, and ONS. The most common immunonutrients are arginine, glutamine (Gln), omega-3 (ω-3) fatty acids, nucleotides, or RNA, which have been seen to modulate inflammatory responses and increase protein synthesis following surgical procedures [74,75]. The advantages obtained from immunonutrient consumption encompass various mechanisms, among them GM modulation. Studies indicate that arginine treatment in mice results in a beneficial alteration of the Firmicutes-to-Bacteroidetes ratio to favor Bacteroidetes, along with decreased expression of nuclear factor-κB (NF-κB), mitogen-activated protein kinase (MAPK), and phosphatidyl inositol 3-kinase/protein kinase B (PI3K/Akt) signaling pathways [76]. Notably, Bacteroidetes promote intestinal innate and mediated immunity, including the secretion of immunoglobulin A (IgA) and various cytokines [76,77]. These findings are particularly significant in the context of diseases like Crohn’s disease (CD) and ulcerative colitis (UC), where Bacteroidetes’ presence is diminished, potentially contributing to the reduced anti-inflammatory effect observed during colitis [78,79]. Similarly, ω-3 fatty acids can modulate the abundance of gut microorganisms. Recent studies suggest that dietary supplementation with ω-3 polyunsaturated fatty acids increases the abundance of various health-promoting bacteria, including butyrate producers from genera such as Bifidobacterium, Roseburia, Lactobacillus, and the mucin specialist Akkermansia muciniphila [80,81,82]. Interestingly, in a cross-sectional study of breast cancer survivors, higher blood levels of docosahexaenoic acid (DHA) were positively associated with an increased abundance of Bifidobacterium in the GM, particularly in participants without a history of chemotherapy [83]. Nevertheless, the influence of immunonutrient use on microbiota is still inadequately investigated, highlighting the need for further research to comprehensively understand the impact of immunonutrition in prevent dysbiosis, especially in the context of GI cancer care. According to the ESPEN guidelines, oral or enteral immunonutrient administration is recommended for upper GI cancer surgery patients in preoperative and postoperative nutritional care [15]. However, more research is needed to assess the efficacy of immunonutrient-enriched formula supplementation compared with standard oral and enteral nutrition in the perioperative period [10]. In a recent meta-analysis, enteral immunonutrition was found to be both safe and effective in reducing overall complications, particularly infectious complications, and it also led to a shortened hospital stay. This positive outcome was observed in patients undergoing surgery for GI cancers, including GC, CRC, esophageal cancer, periampullary cancer, and pancreatic cancer [84].
In studies involving patients undergoing laparoscopic colorectal resection, preoperative and postoperative immunonutrient supplementation was associated with a lower incidence of SSI compared with those who received nutrition counseling alone [85]. Similarly, preoperative EN immunonutrition has been proven effective in preventing SSI in CRC patients without malnutrition [86].
However, when comparing standard ONS with ω-3-enriched ONS in CRC surgery patients, ω-3-enriched ONS did not significantly affect postoperative complications, LOS, postoperative blood loss, the need for intensive care, or hospital readmission [87].
On the other hand, Adiamah et al. reported a 48% risk reduction of postoperative infectious complications in patients receiving preoperative immunonutrition. This intervention also led to shortened LOS of 1.5 days, although it did not impact other complications or mortality [40].
Similarly, Probst et al. found that perioperative immunonutrition reduced infectious complications, general complications, and LOS, with no effect on mortality [88]. Furthermore, a pilot trial assessing perioperative nutritional supplementation in GI cancer patients demonstrated a feasible enrollment fraction of 49% and revealed a higher proportion of infectious complications in the control group, emphasizing the importance of infectious complications as a relevant outcome of interest in such studies [89].
Furthermore, a retrospective study by Franceschilli et al. suggested that the combination of preoperative immunonutrition within the context of the ERAS protocol for patients with normal nutritional status undergoing laparoscopic total gastrectomy (LTG) reduced postoperative complications [90].
In the case of malnourished patients, enteral immunonutrition affected postoperative complications and LOS. However, for patients without malnutrition, the content of nutritional support did not significantly impact complications or LOS [91].
Studies have shown that early postoperative enteral immunonutrition enriched with nutrients like arginine, ω-3 fatty acids, and RNA positively influenced surgical wound healing and immune function in patients undergoing gastrectomy for GC [92,93].
Additionally, immunonutrition has been reported as a safe and feasible nutritional therapy that positively modulates immune responses after esophagectomy [93]. However, in another randomized controlled trial, no significant immunomodulatory effect was observed when comparing immunonutrient-rich EN with standard EN [94].
Lastly, in GI cancer surgery patients, the switch from standard intravenous fluid to immune-enhancing EN reduced infectious complications by two-thirds, while non-infectious complications saw a 13.5% reduction. This result suggests that a nutritional intervention modulating the host immune response may positively influence the relationship between immune support and postoperative infections [95]. PN administration of immunonutrients gave similar results. In an interventional clinical study, GI cancer surgery patients were assigned to two different groups: one group received postoperative total parenteral nutrition (TPN), and the other group received TPN along with a daily supplementation of 0.4 g/kg of Gln. Following these interventions, the nutritional status improved in both groups; however, the group receiving supplementation exhibited significantly greater improvement and demonstrated better results in GI function assessment [96]. Lu et al. previously demonstrated that Gln-enriched TPN led to higher serum prealbumin levels, improved nitrogen balance, and lower levels of inflammatory markers such as IL-6 and C-reactive protein (CRP) compared with standard TPN in postoperative GI cancer patients [97]. These results suggest that Gln-enriched TPN may enhance both nutritional and inflammatory status and potentially reduce the risk of infectious complications in these patients. Regarding the roles of immunonutrition in modulating radio- and chemotherapy side effects, Gln may shorten the duration of chemotherapy-induced diarrhea but does not affect its severity [98]. A systematic review evaluating Gln intake among colon and CRC patients found that Gln may reduce some chemotherapy-induced complications, such as GI-M and diarrhea, and improve postoperative nitrogen balance, immunity, and wound healing, whereas Gln had no beneficial effects on the side effects of radio-chemotherapy [99]. On the other hand, long-chain ω-3 fatty acids and fish oil are recommended to improve body weight, food intake, and other components in patients undergoing chemotherapy and at risk of weight loss or malnutrition [15].
Anyway, the current evidence does not provide a clear role for immunonutrition in managing infectious episodes during chemotherapy in cancer patients [100]. Interestingly, ω-3 has shown potential to enhance the effectiveness of chemotherapy through its synergistic inhibition of cell growth [101]. Mechanistic insights into ω-3’s action were gained through in vitro studies conducted on CRC cell lines, revealing its antiproliferative effects [102,103], promotion of apoptosis [101,104], and improved chemotherapy efficacy [101,102].
The effect of eicosapentaenoic acid (EPA) supplementation in GI patients has been studied with varying outcomes. In a double-blind, placebo-controlled study involving advanced cancer patients with weight and appetite loss, daily administration of 1.8 g of EPA C20:5 ω-3 for two weeks did not result in significant improvements in appetite, fatigue, nausea, overall well-being, caloric intake, nutritional status, or functional abilities compared with the placebo group [105]. A clinical trial reported that dietary counseling by qualified dietitians and the use of EPA-ONS in advanced CRC patients receiving chemotherapy could help maintain weight and potentially enhance symptom control, nutritional status, and QoL [106]. A recent study assessed the impact of perioperative EPA supplementation in patients with localized gastric cancer, as part of a randomized clinical trial. The study found that, overall, there was no significant survival benefit associated with perioperative EPA. However, subgroup analyses indicated potential benefits in patients who received neoadjuvant chemotherapy (NAC) and those with nodal metastasis [107]. Further research may be needed to clarify the specific patient populations that could benefit from EPA supplementation. All these results suggest that immunonutrition can help GI cancer patients in many ways, from perioperative care to symptom reduction and immune system support (Figure 3).

4.2. Calorie Restriction and Fasting

Calorie restriction (CR) is recognized for its anti-inflammatory effects mediated by various mechanisms, demonstrating a beneficial influence on the prevention and treatment of conditions characterized by hyper-inflammatory responses. CR is a nutritional intervention that restricts energy intake by 25–30% without causing malnutrition or deprivation of essential nutrients. Since most dietary energy comes from carbohydrates, energy restriction indirectly leads to carbohydrate restriction. Accordingly, calorie restriction is assumed to regulate effector immune activities that use glucose as the primary substrate [108]. Given that glucose is the primary fuel for cancer cells, calorie restriction emerges as a promising nutritional therapy for individuals with cancer. CR holds potential in exerting anticancer effects by triggering molecular pathways that enhance cellular defenses, support DNA repair, and mitigate oxidative damage [109]. An important aspect of calorie restriction during cancer treatments is fasting, which prompts distinct responses in cancer cells compared with normal cells [110]. In normal cells, fasting leads to a reduction of proteins and enzymes related to cell growth, such as insulin-like growth factor 1 (IGF-1), mammalian target of rapamycin (mTOR), protein kinase A (PKA), and protein kinase B (PKB/AKT). This outcome induces growth arrest or reduction in healthy cells, promoting cell survival and enhancing cellular protection against chemotherapeutic agents—a phenomenon also referred to as differential stress resistance (DSR). In cancer cells, fasting induces differential stress sensitization (DSS), rendering them more susceptible to chemotherapeutic agents and promoting increased cell death [111]. The dual impact of fasting-induced autophagy in cancer underscores its potential diverse applications in cancer treatment. CR holds promise in enhancing treatment effectiveness by modulating autophagy and preserving normal cells. Studies have demonstrated that combining autophagy inhibition with calorie restriction reduces tumor growth more effectively than individual treatments [111,112]. Fasting is suggested as a procedure to halt cancer development and tumor growth by suppressing pathways that activate tumor growth and activating pathways that inhibit tumor growth in tumor cells, thus preventing disease progression [113,114]. However, it is crucial to emphasize that these outcomes were observed with prolonged fasting (>48 h). Additionally, research indicates that short-term fasting can sensitize cancer cells to chemotherapeutic agents, enhancing the efficacy of radiation and chemotherapy [115,116]. Clinical studies involving cancer patients undergoing chemotherapy have demonstrated that fasting, even in the short term, is safe and well tolerated, potentially improving treatment outcomes and enhancing QoL, although in some cases, fasting may have no notable effect [117,118,119].
Some of the positive effects of CR and fasting may be related to the impact of diet on adult stem-cell function [120]. CR enhances intestinal stem cells (ISCs) and neighboring niche cell numbers and increases stem cells’ self-renewal capacity in response to reduced mTOR signaling from Paneth cells [121]. CR decreases PI3K/AKT signaling pathways by reducing circulating insulin/IGF-1 levels and suppresses cell survival in a colon-derived human cancer cell line (SW620), accompanied by increased expression levels of forkhead box O (FOXO) target genes [121]. In addition, CR inhibits colon tumor cell (MC38) growth by regulating NF-κB activation and inflammation-related gene expression [122]. Similarly, fasting induces ISC self-renewal, mediated by peroxisome proliferator-activated receptor δ (PPARδ) triggered by the oxidation of free fatty acids released from adipose tissue. This depends on the nutrient-sensing capacity of the ISCs [120]. Further, Deng et al. showed that fasting reduced leptin-receptor-positive (Lepr+) cell numbers and, thus, serum leptin levels. This leads to a decrease in insulin-like growth factor 1 (Igf1) secreted by Lepr+ cells. As a result, the proliferation of ISCs and progenitor cells is reduced during fasting. It is noteworthy that Lepr+ mesenchymal cells (MCs) perceive dietary changes. However, no apoptotic cells were detected in MCs of intestinal crypts after fasting, indicating that Lepr+ cells decrease independently of apoptosis [123]. Unlike other stem cells, intestinal stem cells coexist with the intestinal microbiota population but live separately in their own integrity. Therefore, the relationship between the microbiota and ISCs needs to be considered. Gut microbes can be devastating, given the vital role of the long-term integrity and functionality of ISCs and progenitor cells. By causing biological damage, the GM promotes the regeneration of the epithelial layer. This leads to defense against pathogens and immunomodulatory effects [124]. Interestingly, fasting has the potential to exert immunomodulatory effects by modulating the microbiome. In mice, it was demonstrated that restricting caloric intake led to GM alterations, specifically an increase in Lactobacillus spp., believed to offer protection against invading pathogens and to lower inflammatory cytokine levels, and a decrease in Streptococcacae, known inducers of mild inflammation [125]. Additional probiotic treatment could further amplify the beneficial effects of fasting. In a pilot study involving overweight individuals, a 1-week fasting diet followed by a 6-week probiotic intervention resulted in increased GM diversity and abundance of mucin-degrading bacteria, notably Akkermansia muciniphila, with the probiotic formula bolstering specific administered gut microbial populations [126].
Although the effects of calorie restriction and fasting on cancer are promising, cancer-related clinical conditions such as malnutrition, cachexia, a possibly weakened immune system, and susceptibility to infection should be taken into account when evaluating the effectiveness of long-term fasting interventions alone in cancer treatment [110]. Therefore, to avoid adverse effects on immune function, it is crucial to implement caloric restriction and fasting interventions in cancer patients in a controlled manner and to maintain balance by maintaining adequate caloric intake.

4.3. The Role of Biotics

One promising avenue for GI cancer patients care is the modulation of the GM using biotics (probiotics, prebiotics, and synbiotics) (Figure 3). Specific probiotic strains of Bifidobacteria, Lactobacilli, E. coli, Propionibacterium, Bacillus, and Saccharomyces can beneficially modulate TLR activation by reducing the activation of MAPK and NF-κB pathways and the production of pro-inflammatory cytokines [127]. It is assumed that the Bifidobacterium longum subsp. longum GT15 strain aims to maintain normal healthy functions by responding to pro-inflammatory cytokines. In addition, overexpression of heat shock protein 20 (Hsp20), which is known to play a role in reducing inflammation, reduces TNF-α expression. In the B. longum subsp. longum GT15 strain exposed to TNF-α, the transcription of the BLGT_RS00625 gene encoding for Hsp20 increased five-fold, suggesting that this mechanism may be one of the pathways used by Bifidobacteria to reduce inflammation [128].
Moreover, a distinct group of Gram-positive bacteria can predominantly produce bacteriocins, a group of bacterial peptides, which display antimicrobial activity against other bacteria [129]. Bacteriocin producers include various genera, such as Pediococcus, Leuconostoc, Lactococcus, Enterococcus, Streptococcus, Lactobacillus, and Bifidobacterium [130,131]. Bacteriocins selectively target pathogens, but not commensal GM [132], and exert cytotoxic activity against cancer cells [133,134]. Intriguingly, some bacteriocins exhibit immunomodulatory properties, thus participating in the maintenance of a balanced crosstalk between GM and immunity [135]. Indeed, bacteriocins secreted by Bacillus subtilis were described as stimulators of innate immune response via IL-1β, IL-6, TNF-α, and NO production in both in vitro cells and mouse peritoneal macrophages [136]. The enhanced phagocytosis of macrophages correlated with the TLR4 and the NF-κB and MAPK signaling pathways [137]. Treatment of human peripheral blood mononuclear cells (PBMCs) with acidocin A, a bacteriocin, resulted in increased production of multiple cytokines and chemokines, including macrophage inflammatory protein (MIP)-1α, MIP-1β, IL-6, and TNF-α [138]. Moreover, Lactobacillus plantarum genes encoding production or secretion of bacteriocins were reported to enhance production of IL-10 over IL-12 and TNF-α induction in dendritic cells (DCs) and in PBMCs [139,140].
Other immune-modulatory functions exerted by the commensal Bacteroides fragilis are the secretion of polysaccharide A that is recognized by the heterodimer TLR2/TLR1 in cooperation with Dectin-1 and induces the cAMP response element-binding protein (CREB)-dependent expression of anti-inflammatory genes [141]. Bacteroides fragilis can also suppress the Th17 responses by promoting Tregs through TLR2 signaling [142]. Also, Lactobacillus reuteri, Lactobacillus murinus, and Helicobacter hepaticus can increase the proportion of IL-10 producing Tregs in mice [143,144,145,146,147].
A randomized, double-blind, placebo-controlled trial in CRC patients undergoing colon–rectal resection demonstrated the benefits of probiotics. Patients received a combination of Lactobacillus and Bifidobacteria strains, which included Lactobacillus acidophilus, Lactobacillus lactis, Lactobacillus casei spp., Bifidobacterium longum, Bifidobacterium bifidum, and Bifidobacterium infantis twice a day for six months. This intervention led to a decrease in pro-inflammatory cytokines and a significant reduction in post-surgical complications [148]. Another randomized controlled prospective study involving CRC patients administered a compound of eight bacterial cultures, including various Lactobacillus and Bifidobacterium strains, showed promising results. Treated patients exhibited a lower frequency of post-surgical complications, reduced operative and postoperative LOS, and a lower mortality rate in a six-month postoperative follow-up compared with untreated patients [149]. In a recent study, 100 CRC patients receiving supplementation with a probiotic containing Bifidobacterium infants, Lactobacillus acidophilus, Enterococcus faecalis, and Bacillus cereus showed dysbiosis alleviation and increased production of SCFA compared with controls [150]. A quantitative meta-analysis involving 14 studies and 1566 patients demonstrated a significant benefit of probiotics and synbiotics administration in both pre-and post-surgical care, especially reducing postoperative infections [151]. Moreover, no significant results were reported in randomized, double-blind controlled studies on patients undergoing ileostomy, performed to prevent further damage associated with anastomotic leak in CRC patients, with perioperative administration of Lactobacillus plantarum CJLP243 [152], or of a probiotic mix (Lactobacillus acidophilus DSM 24735, Lactobacillus acidophilus DSM 24735, Lactobacillus plantarum DSM 24730, Lactobacillus plantarum DSM 24730, Lactobacillus paracasei DSM 24733, Lactobacillus paracasei DSM 24733, Lactobacillus delbrueckii subsp. bulgaricus DSM 2, Bifidobacterium breve DSM 24732, Bifidobacterium longum DSM 24736 113, Bifidobacterium longum DSM 24736, Bifidobacterium infantis DSM 24737, Streptococcus thermophilus DSM 24731) [153]. More studies are needed to assess the efficacy of the treatment and subsequent inclusion of probiotics administration in a protocol before ileostomy [152]. Similar findings were observed in gastric adenocarcinoma patients undergoing radical gastrectomy, where probiotics administration reduced levels of inflammatory markers [154].
GI surgery includes several other interventions, such as hepatectomy with extrahepatic bile duct resection, esophagectomy, and pancreatoduodenectomy, which may result in bacterial translocation to mesenteric lymph nodes (MLNs) and from there to the bloodstream. Since the presence of bacteria in MLNs is directly associated with postoperative infections, it has been observed how a pre-operative administration of synbiotics can improve the intestinal microenvironment and prevent postoperative infections in esophagectomy [155]. Hepatectomy that implies extrahepatic bile duct resection and pancreatoduodenectomy is discussed below [156].
In esophageal cancer, where NAC is recommended as a standard treatment before surgery [157], it has been observed in a randomized control trial that the co-administration of synbiotics reduces the toxicity provoked by the chemotherapy treatment and prepares the intestinal environment for highly invasive surgery, with lower bacterial translocation to the MLN and to the bloodstream [158].
In more severe conditions, patients with advanced stages of esophageal cancer are also treated with prophylactic antibiotics during NAC. In a multicenter randomized study, patients undergoing a pre-operative cycle of NAC were enrolled and randomly assigned either antibiotic administration or a symbiotic administration combined with EN. In this trial, synbiotics administration with EN diminished the side effects of the chemotherapy on the intestinal tract, such as diarrhea, and resulted in an alternative treatment to antibiotics [159].
Unfortunately, synbiotics administration is not beneficial for all patients, and detecting microbiota species in patients’ guts prior to chemotherapy may also be predictive of the efficacy of a co-adjuvant synbiotics treatment [160]. As tested by Sugimoto et al. in a retrospective exploratory study, Anaerostipes hadrus and B. pseudocatenulatum may mitigate chemotherapy side effects and allow the protective role of synbiotics + EN administration during NAC [160]. Considering malignant hepatic neoplasms, hepatic resection is a standard treatment procedure in many cases, but the mortality rate of the surgery is still moderately high, about 3.5% [161]. A meta-analysis of a total of four studies involving 205 patients assessed that the pre-operative administration of prebiotics reduced postoperative infections and the need for antibiotics [162]. However, in a more recent randomized controlled trial, patients with resectable hepatocellular carcinoma administered with probiotics prior to the resection showed no beneficial results regarding bacteria translocation or post-surgical infections [163]. Finally, in pancreatic cancer patients, where the intestinal dysbiosis has been documented [164], the use of pre and probiotics could be a therapeutic approach to alleviate side effects of chemotherapy/radiotherapy, but no significant results have yet been reported. For instance, a trial involving the administration of M-20 (a biotherapeutic agent of soybean fermentation metabolites and microorganisms that reproduce the intestinal environment) to prevent cachexia in pancreatic cancer patients treated with chemotherapy is at its early stage of recruiting (NCT04600154).
While the use of probiotics is generally safe and well tolerated in the general population, their application in vulnerable subpopulations requires careful consideration of several factors, including a thoughtful probiotic selection. For this purpose, the safety and efficacy of diverse formulations used as adjunctive probiotics in oncological surgery have been assessed in various studies [165,166,167,168]. In a systematic review, Cogo et al. evaluated 21 different probiotics formulations in oncological surgery, within 36 randomized controlled trials involving 3305 participants and six nonrandomized/observational cohort studies [165]. Their findings support the belief that the effects of probiotics are specific to the product and formulation, with the most promising results obtained with the post-surgery oral supplementation of Lactobacillus acidophilus LA-5 + Lactobacillus plantarum + Bifidobacterium lactis BB-12 + Saccharomyces boulardii in CRC patients. With regards to safety, among the randomized controlled trials, 47% of patients did not furnish specific data on side effects, 25% did not experience adverse events, while 28% reported common side effects being mild and encompassing nausea and flatulence, indicating a favorable safety profile. However, a small proportion of patients (6%) reported elevated rates of specific complications with the probiotics arms, including pancreatic fistula and 30-day readmission [165]. These findings underscore the necessity for a cautious and ongoing evaluation of the safety profile of probiotics, especially in the context of surgical interventions for individuals with cancer.

5. Conclusions

In light of the intricate and multifaceted factors contributing to nutritional imbalances in GI cancer patients, effective treatments necessitate a comprehensive and multi-disciplinary approach. Immunonutrition and microbiota modulation emerge as promising avenues to enhance nutritional status, regulate immune response, promote tissue repair, and modulate the side effects of anticancer drugs. Bridging the nutritional gap by employing immunonutrients and microbiota modulators early during disease onset can help stabilize weight loss, enhance treatment tolerability, reduce the decline in performance status, prevent infections, and improve survival rates. Unfortunately, clinical practice often involves late-stage assessments, where multiple nutritional deficiencies have already surfaced, and cancer cachexia has become resistant to conventional treatments, potentially yielding contradictory results. Therefore, it is imperative to acknowledge the need for more rigorous clinical trials to thoroughly assess the impact of these interventions.

Author Contributions

Conceptualization, E.N., E.N.D., D.S., I.L. and S.M.; software, E.N.D., D.S., I.L. and S.M.; investigation, E.N., E.N.D., D.S., I.L. and S.M.; resources, E.N., E.N.D., D.S., I.L. and S.M.; writing—original draft preparation, E.N.D., D.S., I.L. and S.M.; writing—review and editing, E.N., I.L. and S.M.; visualization, E.N., E.N.D., D.S., I.L. and S.M.; supervision, E.N.; project administration, E.N. All authors have read and agreed to the published version of the manuscript.

Funding

This work received support from EU Twinning European project: 952583—MICAfrica—H2020-WIDESPREAD-2018-2020/H2020-WIDESPREAD-2020-5 and from the European Union Next Generation EU National Recovery and Resilience Plan, Mission 4 Component 2—Investment 1.5, THE—Tuscany Health Ecosystem, ECS00000017, CUP B83C22003920001.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed]
  2. 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] [PubMed]
  3. Hébuterne, X.; Lemarié, E.; Michallet, M.; de Montreuil, C.B.; Schneider, S.M.; Goldwasser, F. Prevalence of malnutrition and current use of nutrition support in patients with cancer. JPEN J. Parenter. Enteral Nutr. 2014, 38, 196–204. [Google Scholar] [CrossRef] [PubMed]
  4. Abraham, M.; Kordatou, Z.; Barriuso, J.; Lamarca, A.; Weaver, J.M.J.; Cipriano, C.; Papaxoinis, G.; Backen, A.; Mansoor, W. Early recognition of anorexia through patient-generated assessment predicts survival in patients with oesophagogastric cancer. PLoS ONE 2019, 14, e0224540. [Google Scholar] [CrossRef]
  5. Asakawa, A.; Fujimiya, M.; Niijima, A.; Fujino, K.; Kodama, N.; Sato, Y.; Kato, I.; Nanba, H.; Laviano, A.; Meguid, M.M.; et al. Parathyroid hormone-related protein has an anorexigenic activity via activation of hypothalamic urocortins 2 and 3. Psychoneuroendocrinology 2010, 35, 1178–1186. [Google Scholar] [CrossRef]
  6. Ezeoke, C.C.; Morley, J.E. Pathophysiology of anorexia in the cancer cachexia syndrome. J. Cachexia Sarcopenia Muscle 2015, 6, 287–302. [Google Scholar] [CrossRef]
  7. Cruz-Jentoft, A.J.; Bahat, G.; Bauer, J.; Boirie, Y.; Bruyère, O.; Cederholm, T.; Cooper, C.; Landi, F.; Rolland, Y.; Sayer, A.A.; et al. Sarcopenia: Revised European consensus on definition and diagnosis. Age Ageing 2019, 48, 16–31. [Google Scholar] [CrossRef]
  8. Haiducu, C.; Buzea, A.; Mirea, L.E.; Dan, G.A. The prevalence and the impact of sarcopenia in digestive cancers. A systematic review. Rom. J. Intern. Med. 2021, 59, 328–344. [Google Scholar] [CrossRef]
  9. Simonsen, C.; de Heer, P.; Bjerre, E.D.; Suetta, C.; Hojman, P.; Pedersen, B.K.; Svendsen, L.B.; Christensen, J.F. Sarcopenia and Postoperative Complication Risk in Gastrointestinal Surgical Oncology: A Meta-analysis. Ann. Surg. 2018, 268, 58–69. [Google Scholar] [CrossRef]
  10. Weimann, A.; Braga, M.; Carli, F.; Higashiguchi, T.; Hübner, M.; Klek, S.; Laviano, A.; Ljungqvist, O.; Lobo, D.N.; Martindale, R.G.; et al. ESPEN practical guideline: Clinical nutrition in surgery. Clin. Nutr. 2021, 40, 4745–4761. [Google Scholar] [CrossRef]
  11. Wu, J.M.; Yang, H.T.; Ho, T.W.; Shun, S.C.; Lin, M.T. Association between Interleukin-6 Levels and Perioperative Fatigue in Gastric Adenocarcinoma Patients. J. Clin. Med. 2019, 8, 543. [Google Scholar] [CrossRef]
  12. Gannavarapu, B.S.; Lau, S.K.M.; Carter, K.; Cannon, N.A.; Gao, A.; Ahn, C.; Meyer, J.J.; Sher, D.J.; Jatoi, A.; Infante, R.; et al. Prevalence and Survival Impact of Pretreatment Cancer-Associated Weight Loss: A Tool for Guiding Early Palliative Care. J. Oncol. Pract. 2018, 14, e238–e250. [Google Scholar] [CrossRef]
  13. Anker, M.S.; Holcomb, R.; Muscaritoli, M.; von Haehling, S.; Haverkamp, W.; Jatoi, A.; Morley, J.E.; Strasser, F.; Landmesser, U.; Coats, A.J.S.; et al. Orphan disease status of cancer cachexia in the USA and in the European Union: A systematic review. J. Cachexia Sarcopenia Muscle 2019, 10, 22–34. [Google Scholar] [CrossRef] [PubMed]
  14. Molfino, A.; Imbimbo, G.; Laviano, A. Current Screening Methods for the Risk or Presence of Malnutrition in Cancer Patients. Cancer Manag. Res. 2022, 14, 561–567. [Google Scholar] [CrossRef] [PubMed]
  15. Muscaritoli, M.; Arends, J.; Bachmann, P.; Baracos, V.; Barthelemy, N.; Bertz, H.; Bozzetti, F.; Hütterer, E.; Isenring, E.; Kaasa, S.; et al. ESPEN practical guideline: Clinical Nutrition in cancer. Clin. Nutr. 2021, 40, 2898–2913. [Google Scholar] [CrossRef] [PubMed]
  16. Vitaloni, M.; Caccialanza, R.; Ravasco, P.; Carrato, A.; Kapala, A.; de van der Schueren, M.; Constantinides, D.; Backman, E.; Chuter, D.; Santangelo, C.; et al. The impact of nutrition on the lives of patients with digestive cancers: A position paper. Support. Care Cancer 2022, 30, 7991–7996. [Google Scholar] [CrossRef] [PubMed]
  17. Mahendran, V.J.; Stringer, A.M.; Semple, S.J.; Song, Y.; Garg, S. Advances in the Use of Anti-inflammatory Agents to Manage Chemotherapy-induced Oral and Gastrointestinal Mucositis. Curr. Pharm. Des. 2018, 24, 1518–1532. [Google Scholar] [CrossRef]
  18. de Mooij, C.E.M.; van der Velden, W.J.F.M.; de Haan, A.F.J.; Fazel, S.; van Groningen, L.F.J.; Blijlevens, N.M.A. Grading bloodstream infection risk using citrulline as a biomarker of intestinal mucositis in patients receiving intensive therapy. Bone Marrow Transplant. 2022, 57, 1373–1381. [Google Scholar] [CrossRef]
  19. Arends, J. How to feed patients with gastrointestinal mucositis. Curr. Opin. Support. Palliat. Care 2018, 12, 168–173. [Google Scholar] [CrossRef]
  20. Kuiken, N.S.; Rings, E.H.; Havinga, R.; Groen, A.K.; Tissing, W.J. Effect of minimal enteral feeding on recovery in a methotrexate-induced gastrointestinal mucositis rat model. Support. Care Cancer 2016, 24, 1357–1364. [Google Scholar] [CrossRef]
  21. Muscaritoli, M.; Lucia, S.; Farcomeni, A.; Lorusso, V.; Saracino, V.; Barone, C.; Plastino, F.; Gori, S.; Magarotto, R.; Carteni, G.; et al. Prevalence of malnutrition in patients at first medical oncology visit: The PreMiO study. Oncotarget 2017, 8, 79884–79896. [Google Scholar] [CrossRef]
  22. Wheelwright, S.J.; Hopkinson, J.B.; Darlington, A.S.; Fitzsimmons, D.F.; Fayers, P.; Balstad, T.R.; Bredart, A.; Hammerlid, E.; Kaasa, S.; Nicolatou-Galitis, O.; et al. Development of the EORTC QLQ-CAX24, A Questionnaire for Cancer Patients with Cachexia. J. Pain. Symptom Manag. 2017, 53, 232–242. [Google Scholar] [CrossRef] [PubMed]
  23. Holland, J.C.; Alici, Y. Management of distress in cancer patients. J. Support. Oncol. 2010, 8, 4–12. [Google Scholar]
  24. Bozzetti, F.; Group, S.W. Screening the nutritional status in oncology: A preliminary report on 1000 outpatients. Support. Care Cancer 2009, 17, 279–284. [Google Scholar] [CrossRef]
  25. Muscaritoli, M.; Corsaro, E.; Molfino, A. Awareness of Cancer-Related Malnutrition and Its Management: Analysis of the Results From a Survey Conducted Among Medical Oncologists. Front. Oncol. 2021, 11, 682999. [Google Scholar] [CrossRef]
  26. Furness, K.; Silvers, M.A.; Savva, J.; Huggins, C.E.; Truby, H.; Haines, T. Long-term follow-up of the potential benefits of early nutritional intervention in adults with upper gastrointestinal cancer: A pilot randomised trial. Support. Care Cancer 2017, 25, 3587–3593. [Google Scholar] [CrossRef]
  27. Parsons, H.M.; Forte, M.L.; Abdi, H.I.; Brandt, S.; Claussen, A.M.; Wilt, T.; Klein, M.; Ester, E.; Landsteiner, A.; Shaukut, A.; et al. Nutrition as prevention for improved cancer health outcomes: A systematic literature review. JNCI Cancer Spectr. 2023, 7, pkad035. [Google Scholar] [CrossRef]
  28. Carrillo Lozano, E.; Osés Zárate, V.; Campos Del Portillo, R. Nutritional management of gastric cancer. Endocrinol. Diabetes Nutr. (Engl. Ed.) 2021, 68, 428–438. [Google Scholar] [CrossRef]
  29. Cencioni, C.; Trestini, I.; Piro, G.; Bria, E.; Tortora, G.; Carbone, C.; Spallotta, F. Gastrointestinal Cancer Patient Nutritional Management: From Specific Needs to Novel Epigenetic Dietary Approaches. Nutrients 2022, 14, 1542. [Google Scholar] [CrossRef]
  30. Poon, M.; Dennis, K.; DeAngelis, C.; Chung, H.; Stinson, J.; Zhang, L.; Bedard, G.; Popovic, M.; Lao, N.; Pulenzas, N.; et al. A prospective study of gastrointestinal radiation therapy-induced nausea and vomiting. Support. Care Cancer 2014, 22, 1493–1507. [Google Scholar] [CrossRef]
  31. Virizuela, J.A.; Camblor-Álvarez, M.; Luengo-Pérez, L.M.; Grande, E.; Álvarez-Hernández, J.; Sendrós-Madroño, M.J.; Jiménez-Fonseca, P.; Cervera-Peris, M.; Ocón-Bretón, M.J. Nutritional support and parenteral nutrition in cancer patients: An expert consensus report. Clin. Transl. Oncol. 2018, 20, 619–629. [Google Scholar] [CrossRef] [PubMed]
  32. Davies, A.N.; Waghorn, M.; Webber, K.; Johnsen, S.; Mendis, J.; Boyle, J. A cluster randomised feasibility trial of clinically assisted hydration in cancer patients in the last days of life. Palliat. Med. 2018, 32, 733–743. [Google Scholar] [CrossRef] [PubMed]
  33. Iimura, Y.; Kurokawa, T.; Nojima, M.; Kanemoto, Y.; Yazawa, K.; Tsurita, G.; Kuroda, S. Potential thiamine deficiency and neurological symptoms in patients receiving chemotherapy for gastrointestinal cancer. Int. J. Clin. Pharmacol. Ther. 2020, 58, 139–145. [Google Scholar] [CrossRef] [PubMed]
  34. Turkiewicz, J.; Garcia, M.; Li, Z.; Surampudi, V. Micronutrient deficiencies in patients with gastrointestinal cancer: To treat or not to treat. Nutr. Clin. Pract. 2023, 38, 749–760. [Google Scholar] [CrossRef] [PubMed]
  35. Bozzetti, F. Nutritional interventions in elderly gastrointestinal cancer patients: The evidence from randomized controlled trials. Support. Care Cancer 2019, 27, 721–727. [Google Scholar] [CrossRef]
  36. Fukuda, Y.; Yamamoto, K.; Hirao, M.; Nishikawa, K.; Maeda, S.; Haraguchi, N.; Miyake, M.; Hama, N.; Miyamoto, A.; Ikeda, M.; et al. Prevalence of Malnutrition Among Gastric Cancer Patients Undergoing Gastrectomy and Optimal Preoperative Nutritional Support for Preventing Surgical Site Infections. Ann. Surg. Oncol. 2015, 22 (Suppl. S3), S778–S785. [Google Scholar] [CrossRef]
  37. Reece, L.; Hogan, S.; Allman-Farinelli, M.; Carey, S. Oral nutrition interventions in patients undergoing gastrointestinal surgery for cancer: A systematic literature review. Support. Care Cancer 2020, 28, 5673–5691. [Google Scholar] [CrossRef]
  38. Minnella, E.M.; Awasthi, R.; Loiselle, S.E.; Agnihotram, R.V.; Ferri, L.E.; Carli, F. Effect of Exercise and Nutrition Prehabilitation on Functional Capacity in Esophagogastric Cancer Surgery: A Randomized Clinical Trial. JAMA Surg. 2018, 153, 1081–1089. [Google Scholar] [CrossRef]
  39. Andersen, H.K.; Lewis, S.J.; Thomas, S. Early enteral nutrition within 24h of colorectal surgery versus later commencement of feeding for postoperative complications. Cochrane Database Syst. Rev. 2006, 4, CD004080. [Google Scholar] [CrossRef]
  40. Adiamah, A.; Skořepa, P.; Weimann, A.; Lobo, D.N. The Impact of Preoperative Immune Modulating Nutrition on Outcomes in Patients Undergoing Surgery for Gastrointestinal Cancer: A Systematic Review and Meta-analysis. Ann. Surg. 2019, 270, 247–256. [Google Scholar] [CrossRef]
  41. Ghidini, M.; Hochster, H.; Doi, T.; Van Cutsem, E.; Makris, L.; Takahashi, O.; Benhadji, K.A.; Mansoor, W. Body weight loss as a prognostic and predictive factor in previously treated patients with metastatic gastric cancer: Post hoc analyses of the randomized phase III TAGS trial. Gastric Cancer 2023, 26, 626–637. [Google Scholar] [CrossRef]
  42. Schiessel, D.L.; Vicente Cavagnari, M.A.; Mazur, C.E.; Melhem, A.R.F.; Gavarrete, D.D.; de Andrade, R.M.; Antunes, L.B.B. The Relationship between Unintentional Weight Loss, Grading System and Overall Survival in Gastric Cancer Patients. Nutr. Cancer 2022, 74, 1745–1753. [Google Scholar] [CrossRef] [PubMed]
  43. Sánchez-Lara, K.; Ugalde-Morales, E.; Motola-Kuba, D.; Green, D. Gastrointestinal symptoms and weight loss in cancer patients receiving chemotherapy. Br. J. Nutr. 2013, 109, 894–897. [Google Scholar] [CrossRef] [PubMed]
  44. Takayoshi, K.; Uchino, K.; Nakano, M.; Ikejiri, K.; Baba, E. Weight Loss During Initial Chemotherapy Predicts Survival in Patients with Advanced Gastric Cancer. Nutr. Cancer 2017, 69, 408–415. [Google Scholar] [CrossRef]
  45. Meng, Q.; Tan, S.; Jiang, Y.; Han, J.; Xi, Q.; Zhuang, Q.; Wu, G. Post-discharge oral nutritional supplements with dietary advice in patients at nutritional risk after surgery for gastric cancer: A randomized clinical trial. Clin. Nutr. 2021, 40, 40–46. [Google Scholar] [CrossRef]
  46. Zhu, M.W.; Yang, X.; Xiu, D.R.; Yang, Y.; Li, G.X.; Hu, W.G.; Wang, Z.G.; Cui, H.Y.; Wei, J.M. Effect of oral nutritional supplementation on the post-discharge nutritional status and quality of life of gastrointestinal cancer patients after surgery: A multi-center study. Asia Pac. J. Clin. Nutr. 2019, 28, 450–456. [Google Scholar] [CrossRef]
  47. Beattie, A.H.; Prach, A.T.; Baxter, J.P.; Pennington, C.R. A randomised controlled trial evaluating the use of enteral nutritional supplements postoperatively in malnourished surgical patients. Gut 2000, 46, 813–818. [Google Scholar] [CrossRef]
  48. Pędziwiatr, M.; Mavrikis, J.; Witowski, J.; Adamos, A.; Major, P.; Nowakowski, M.; Budzyński, A. Current status of enhanced recovery after surgery (ERAS) protocol in gastrointestinal surgery. Med. Oncol. 2018, 35, 95. [Google Scholar] [CrossRef]
  49. Margraf, A.; Ludwig, N.; Zarbock, A.; Rossaint, J. Systemic Inflammatory Response Syndrome After Surgery: Mechanisms and Protection. Anesth. Analg. 2020, 131, 1693–1707. [Google Scholar] [CrossRef]
  50. Eto, K.; Urashima, M.; Kosuge, M.; Ohkuma, M.; Noaki, R.; Neki, K.; Ito, D.; Takeda, Y.; Sugano, H.; Yanaga, K. Standardization of surgical procedures to reduce risk of anastomotic leakage, reoperation, and surgical site infection in colorectal cancer surgery: A retrospective cohort study of 1189 patients. Int. J. Colorectal. Dis. 2018, 33, 755–762. [Google Scholar] [CrossRef]
  51. Gül, M.O.; Akyüz, C.; Özkara, S. The effect of immunonutrition on tumor infiltrative t lymphocytes and regulatory t cells in rectal tumor patients receiving neoadjuvant chemoradiotherapy: A prospective randomized clinical study. Turk. J. Med. Sci. 2022, 52, 1058–1066. [Google Scholar] [CrossRef]
  52. Stavrou, G.; Kotzampassi, K. Gut microbiome, surgical complications and probiotics. Ann. Gastroenterol. 2017, 30, 45–53. [Google Scholar] [CrossRef]
  53. Duca, F.A.; Lam, T.K. Gut microbiota, nutrient sensing and energy balance. Diabetes Obes. Metab. 2014, 16 (Suppl. S1), 68–76. [Google Scholar] [CrossRef]
  54. Mori, H.; Verbeure, W.; Schol, J.; Carbone, F.; Tack, J. Gastrointestinal hormones and regulation of gastric emptying. Curr. Opin. Endocrinol. Diabetes Obes. 2022, 29, 191–199. [Google Scholar] [CrossRef] [PubMed]
  55. Hou, K.; Wu, Z.X.; Chen, X.Y.; Wang, J.Q.; Zhang, D.; Xiao, C.; Zhu, D.; Koya, J.B.; Wei, L.; Li, J.; et al. Microbiota in health and diseases. Signal Transduct. Target. Ther. 2022, 7, 135. [Google Scholar] [CrossRef] [PubMed]
  56. Russo, E.; Nannini, G.; Dinu, M.; Pagliai, G.; Sofi, F.; Amedei, A. Exploring the food-gut axis in immunotherapy response of cancer patients. World J. Gastroenterol. 2020, 26, 4919–4932. [Google Scholar] [CrossRef]
  57. Cheng, W.Y.; Wu, C.Y.; Yu, J. The role of gut microbiota in cancer treatment: Friend or foe? Gut 2020, 69, 1867–1876. [Google Scholar] [CrossRef] [PubMed]
  58. Wardill, H.R.; Gibson, R.J.; Van Sebille, Y.Z.; Secombe, K.R.; Coller, J.K.; White, I.A.; Manavis, J.; Hutchinson, M.R.; Staikopoulos, V.; Logan, R.M.; et al. Irinotecan-Induced Gastrointestinal Dysfunction and Pain Are Mediated by Common TLR4-Dependent Mechanisms. Mol. Cancer Ther. 2016, 15, 1376–1386. [Google Scholar] [CrossRef] [PubMed]
  59. De Almeida, C.V.; de Camargo, M.R.; Russo, E.; Amedei, A. Role of diet and gut microbiota on colorectal cancer immunomodulation. World J. Gastroenterol. 2019, 25, 151–162. [Google Scholar] [CrossRef]
  60. Russo, E.; Gloria, L.D.; Nannini, G.; Meoni, G.; Niccolai, E.; Ringressi, M.N.; Baldi, S.; Fani, R.; Tenori, L.; Taddei, A.; et al. From adenoma to CRC stages: The oral-gut microbiome axis as a source of potential microbial and metabolic biomarkers of malignancy. Neoplasia 2023, 40, 100901. [Google Scholar] [CrossRef]
  61. Fung, T.C.; Olson, C.A.; Hsiao, E.Y. Interactions between the microbiota, immune and nervous systems in health and disease. Nat. Neurosci. 2017, 20, 145–155. [Google Scholar] [CrossRef] [PubMed]
  62. Liu, T.; Sun, Z.; Yang, Z.; Qiao, X. Microbiota-derived short-chain fatty acids and modulation of host-derived peptides formation: Focused on host defense peptides. Biomed. Pharmacother. 2023, 162, 114586. [Google Scholar] [CrossRef] [PubMed]
  63. Coombes, J.L.; Powrie, F. Dendritic cells in intestinal immune regulation. Nat. Rev. Immunol. 2008, 8, 435–446. [Google Scholar] [CrossRef] [PubMed]
  64. Stockinger, B.; Shah, K.; Wincent, E. AHR in the intestinal microenvironment: Safeguarding barrier function. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 559–570. [Google Scholar] [CrossRef] [PubMed]
  65. Baldi, S.; Menicatti, M.; Nannini, G.; Niccolai, E.; Russo, E.; Ricci, F.; Pallecchi, M.; Romano, F.; Pedone, M.; Poli, G.; et al. Free Fatty Acids Signature in Human Intestinal Disorders: Significant Association between Butyric Acid and Celiac Disease. Nutrients 2021, 13, 742. [Google Scholar] [CrossRef]
  66. Niccolai, E.; Di Pilato, V.; Nannini, G.; Baldi, S.; Russo, E.; Zucchi, E.; Martinelli, I.; Menicatti, M.; Bartolucci, G.; Mandrioli, J.; et al. The Gut Microbiota-Immunity Axis in ALS: A Role in Deciphering Disease Heterogeneity? Biomedicines 2021, 9, 753. [Google Scholar] [CrossRef]
  67. Celiberto, L.S.; Graef, F.A.; Healey, G.R.; Bosman, E.S.; Jacobson, K.; Sly, L.M.; Vallance, B.A. Inflammatory bowel disease and immunonutrition: Novel therapeutic approaches through modulation of diet and the gut microbiome. Immunology 2018, 155, 36–52. [Google Scholar] [CrossRef]
  68. Klimesova, K.; Kverka, M.; Zakostelska, Z.; Hudcovic, T.; Hrncir, T.; Stepankova, R.; Rossmann, P.; Ridl, J.; Kostovcik, M.; Mrazek, J.; et al. Altered gut microbiota promotes colitis-associated cancer in IL-1 receptor-associated kinase M-deficient mice. Inflamm. Bowel Dis. 2013, 19, 1266–1277. [Google Scholar] [CrossRef]
  69. Iida, N.; Dzutsev, A.; Stewart, C.A.; Smith, L.; Bouladoux, N.; Weingarten, R.A.; Molina, D.A.; Salcedo, R.; Back, T.; Cramer, S.; et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 2013, 342, 967–970. [Google Scholar] [CrossRef]
  70. Daillère, R.; Vétizou, M.; Waldschmitt, N.; Yamazaki, T.; Isnard, C.; Poirier-Colame, V.; Duong, C.P.M.; Flament, C.; Lepage, P.; Roberti, M.P.; et al. Enterococcus hirae and Barnesiella intestinihominis Facilitate Cyclophosphamide-Induced Therapeutic Immunomodulatory Effects. Immunity 2016, 45, 931–943. [Google Scholar] [CrossRef]
  71. Teng, H.; Wang, Y.; Sui, X.; Fan, J.; Li, S.; Lei, X.; Shi, C.; Sun, W.; Song, M.; Wang, H.; et al. Gut microbiota-mediated nucleotide synthesis attenuates the response to neoadjuvant chemoradiotherapy in rectal cancer. Cancer Cell 2023, 41, 124–138.e6. [Google Scholar] [CrossRef] [PubMed]
  72. Liu, Y.; Baba, Y.; Ishimoto, T.; Gu, X.; Zhang, J.; Nomoto, D.; Okadome, K.; Baba, H.; Qiu, P. Gut microbiome in gastrointestinal cancer: A friend or foe? Int. J. Biol. Sci. 2022, 18, 4101–4117. [Google Scholar] [CrossRef]
  73. Liu, X.; Chen, Y.; Zhang, S.; Dong, L. Gut microbiota-mediated immunomodulation in tumor. J. Exp. Clin. Cancer Res. 2021, 40, 221. [Google Scholar] [CrossRef] [PubMed]
  74. Sánchez-Guillén, L.; Arroyo, A. Immunonutrition in patients with colon cancer. Immunotherapy 2020, 12, 5–8. [Google Scholar] [CrossRef]
  75. Challine, A.; Rives-Lange, C.; Danoussou, D.; Katsahian, S.; Ait Boudaoud, A.; Gaujoux, S.; Dousset, B.; Carette, C.; Lazzati, A.; Czernichow, S. Impact of Oral Immunonutrition on Postoperative Morbidity in Digestive Oncologic Surgery: A Nation-wide Cohort Study. Ann. Surg. 2021, 273, 725–731. [Google Scholar] [CrossRef]
  76. Ren, W.; Chen, S.; Yin, J.; Duan, J.; Li, T.; Liu, G.; Feng, Z.; Tan, B.; Yin, Y.; Wu, G. Dietary Arginine Supplementation of Mice Alters the Microbial Population and Activates Intestinal Innate Immunity. J. Nutr. 2014, 144, 988–995. [Google Scholar] [CrossRef]
  77. Wu, M.; Xiao, H.; Shao, F.; Tan, B.; Hu, S. Arginine accelerates intestinal health through cytokines and intestinal microbiota. Int. Immunopharmacol. 2020, 81, 106029. [Google Scholar] [CrossRef]
  78. Singh, K.; Gobert, A.P.; Coburn, L.A.; Barry, D.P.; Allaman, M.; Asim, M.; Luis, P.B.; Schneider, C.; Milne, G.L.; Boone, H.H.; et al. Dietary Arginine Regulates Severity of Experimental Colitis and Affects the Colonic Microbiome. Front. Cell Infect. Microbiol. 2019, 9, 66. [Google Scholar] [CrossRef]
  79. Singh, K.; Coburn, L.A.; Barry, D.P.; Boucher, J.L.; Chaturvedi, R.; Wilson, K.T. L-arginine uptake by cationic amino acid transporter 2 is essential for colonic epithelial cell restitution. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 302, G1061–G1073. [Google Scholar] [CrossRef]
  80. Watson, H.; Mitra, S.; Croden, F.C.; Taylor, M.; Wood, H.M.; Perry, S.L.; Spencer, J.A.; Quirke, P.; Toogood, G.J.; Lawton, C.L.; et al. A randomised trial of the effect of omega-3 polyunsaturated fatty acid supplements on the human intestinal microbiota. Gut 2018, 67, 1974–1983. [Google Scholar] [CrossRef]
  81. Rifkin, S.B.; Sen, A.; Turgeon, D.K.; Chan, R.; Ruffin, M.T.; Brenner, D.E.; Schloss, P.D.; Djuric, Z. Increased Akkermansia abundance is associated with increased colonic mucosal ω-3 fatty acids and decreased colonic mucosal PGE2 concentrations following healthy dietary pattern interventions. MedRxiv 2021, 21264748. [Google Scholar] [CrossRef]
  82. Roussel, C.; Anunciação Braga Guebara, S.; Plante, P.L.; Desjardins, Y.; Di Marzo, V.; Silvestri, C. Short-term supplementation with ω-3 polyunsaturated fatty acids modulates primarily mucolytic species from the gut luminal mucin niche in a human fermentation system. Gut Microbes 2022, 14, 2120344. [Google Scholar] [CrossRef] [PubMed]
  83. Horigome, A.; Okubo, R.; Hamazaki, K.; Kinoshita, T.; Katsumata, N.; Uezono, Y.; Xiao, J.Z.; Matsuoka, Y.J. Association between blood omega-3 polyunsaturated fatty acids and the gut microbiota among breast cancer survivors. Benef. Microbes 2019, 10, 751–758. [Google Scholar] [CrossRef]
  84. Shen, J.; Dai, S.; Li, Z.; Dai, W.; Hong, J.; Huang, J.; Chen, J. Effect of Enteral Immunonutrition in Patients Undergoing Surgery for Gastrointestinal Cancer: An Updated Systematic Review and Meta-Analysis. Front. Nutr. 2022, 9, 941975. [Google Scholar] [CrossRef] [PubMed]
  85. Moya, P.; Miranda, E.; Soriano-Irigaray, L.; Arroyo, A.; Aguilar, M.D.; Bellón, M.; Muñoz, J.L.; Candela, F.; Calpena, R. Perioperative immunonutrition in normo-nourished patients undergoing laparoscopic colorectal resection. Surg. Endosc. 2016, 30, 4946–4953. [Google Scholar] [CrossRef] [PubMed]
  86. Horie, H.; Okada, M.; Kojima, M.; Nagai, H. Favorable effects of preoperative enteral immunonutrition on a surgical site infection in patients with colorectal cancer without malnutrition. Surg. Today 2006, 36, 1063–1068. [Google Scholar] [CrossRef] [PubMed]
  87. 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]
  88. Probst, P.; Ohmann, S.; Klaiber, U.; Hüttner, F.J.; Billeter, A.T.; Ulrich, A.; Büchler, M.W.; Diener, M.K. Meta-analysis of immunonutrition in major abdominal surgery. Br. J. Surg. 2017, 104, 1594–1608. [Google Scholar] [CrossRef]
  89. Serrano, P.E.; Parpia, S.; Simunovic, M.; Duceppe, E.; Pinto-Sanchez, M.I.; Bhandari, M.; Levine, M. Perioperative optimization with nutritional supplements in patients undergoing gastrointestinal surgery for cancer: A randomized, placebo-controlled feasibility clinical trial. Surgery 2022, 172, 670–676. [Google Scholar] [CrossRef]
  90. Franceschilli, M.; Siragusa, L.; Usai, V.; Dhimolea, S.; Pirozzi, B.; Sibio, S.; Di Carlo, S. Immunonutrition reduces complications rate and length of stay after laparoscopic total gastrectomy: A single unit retrospective study. Discov. Oncol. 2022, 13, 62. [Google Scholar] [CrossRef]
  91. Klek, S.; Szybinski, P.; Szczepanek, K. Perioperative immunonutrition in surgical cancer patients: A summary of a decade of research. World J. Surg. 2014, 38, 803–812. [Google Scholar] [CrossRef]
  92. Farreras, N.; Artigas, V.; Cardona, D.; Rius, X.; Trias, M.; González, J.A. Effect of early postoperative enteral immunonutrition on wound healing in patients undergoing surgery for gastric cancer. Clin. Nutr. 2005, 24, 55–65. [Google Scholar] [CrossRef] [PubMed]
  93. Li, X.K.; Cong, Z.Z.; Wu, W.J.; Xu, Y.; Zhou, H.; Wang, G.M.; Qiang, Y.; Luo, L.G.; Shen, Y. Enteral immunonutrition versus enteral nutrition for patients undergoing esophagectomy: A randomized controlled trial. Ann. Palliat. Med. 2021, 10, 1351–1361. [Google Scholar] [CrossRef]
  94. Ma, C.; Tsai, H.; Su, W.; Sun, L.; Shih, Y.; Wang, J. Combination of arginine, glutamine, and omega-3 fatty acid supplements for perioperative enteral nutrition in surgical patients with gastric adenocarcinoma or gastrointestinal stromal tumor (GIST): A prospective, randomized, double-blind study. J. Postgrad. Med. 2018, 64, 155–163. [Google Scholar] [CrossRef] [PubMed]
  95. Bozzetti, F.; Gianotti, L.; Braga, M.; Di Carlo, V.; Mariani, L. Postoperative complications in gastrointestinal cancer patients: The joint role of the nutritional status and the nutritional support. Clin. Nutr. 2007, 26, 698–709. [Google Scholar] [CrossRef]
  96. Beltrán Chaidez, Y.L.; Reyes Barretero, D.Y.; Flores Merino, M.V.; Jaimes Alpizar, E.; de Anda Torres, V.Y.; Domínguez García, M.V. Effect of parenteral glutamine in patients with gastrointestinal cancer undergoing surgery. Nutr. Hosp. 2019, 36, 5–12. [Google Scholar] [CrossRef] [PubMed]
  97. Lu, C.Y.; Shih, Y.L.; Sun, L.C.; Chuang, J.F.; Ma, C.J.; Chen, F.M.; Wu, D.C.; Hsieh, J.S.; Wang, J.Y. The inflammatory modulation effect of glutamine-enriched total parenteral nutrition in postoperative gastrointestinal cancer patients. Am. Surg. 2011, 77, 59–64. [Google Scholar] [CrossRef]
  98. Sun, J.; Wang, H.; Hu, H. Glutamine for chemotherapy induced diarrhea: A meta-analysis. Asia Pac. J. Clin. Nutr. 2012, 21, 380–385. [Google Scholar]
  99. Jolfaie, N.R.; Mirzaie, S.; Ghiasvand, R.; Askari, G.; Miraghajani, M. The effect of glutamine intake on complications of colorectal and colon cancer treatment: A systematic review. J. Res. Med. Sci. 2015, 20, 910–918. [Google Scholar] [CrossRef]
  100. 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]
  101. Granci, V.; Cai, F.; Lecumberri, E.; Clerc, A.; Dupertuis, Y.M.; Pichard, C. Colon cancer cell chemosensitisation by fish oil emulsion involves apoptotic mitochondria pathway. Br. J. Nutr. 2013, 109, 1188–1195. [Google Scholar] [CrossRef] [PubMed]
  102. De Carlo, F.; Witte, T.R.; Hardman, W.E.; Claudio, P.P. Omega-3 eicosapentaenoic acid decreases CD133 colon cancer stem-like cell marker expression while increasing sensitivity to chemotherapy. PLoS ONE 2013, 8, e69760. [Google Scholar] [CrossRef] [PubMed]
  103. Fenton, J.I.; McCaskey, S.J. Curcumin and docosahexaenoic acid block insulin-induced colon carcinoma cell proliferation. Prostaglandins Leukot. Essent. Fat. Acids 2013, 88, 219–226. [Google Scholar] [CrossRef] [PubMed]
  104. Fasano, E.; Serini, S.; Piccioni, E.; Toesca, A.; Monego, G.; Cittadini, A.R.; Ranelletti, F.O.; Calviello, G. DHA induces apoptosis by altering the expression and cellular location of GRP78 in colon cancer cell lines. Biochim. Biophys. Acta 2012, 1822, 1762–1772. [Google Scholar] [CrossRef]
  105. Bruera, E.; Strasser, F.; Palmer, J.L.; Willey, J.; Calder, K.; Amyotte, G.; Baracos, V. Effect of fish oil on appetite and other symptoms in patients with advanced cancer and anorexia/cachexia: A double-blind, placebo-controlled study. J. Clin. Oncol. 2003, 21, 129–134. [Google Scholar] [CrossRef] [PubMed]
  106. Read, J.A.; Beale, P.J.; Volker, D.H.; Smith, N.; Childs, A.; Clarke, S.J. Nutrition intervention using an eicosapentaenoic acid (EPA)-containing supplement in patients with advanced colorectal cancer. Effects on nutritional and inflammatory status: A phase II trial. Support. Care Cancer 2007, 15, 301–307. [Google Scholar] [CrossRef]
  107. Aoyama, T.; Yoshikawa, T.; Ida, S.; Cho, H.; Sakamaki, K.; Ito, Y.; Fujitani, K.; Takiguchi, N.; Kawashima, Y.; Nishikawa, K.; et al. Effects of perioperative Eicosapentaenoic acid-enriched oral nutritional supplement on lean body mass after total gastrectomy for gastric cancer. J. Cancer 2019, 10, 1070–1076. [Google Scholar] [CrossRef]
  108. Dübüş, E.N.; Lamminpää, I.; Nannini, G.; Niccolai, E. Nourishing Immunity and Combatting Neuroinflammation: The Power of Immunonutrition and The Microbiome. Front. Biosci. 2023, 28, 178. [Google Scholar] [CrossRef]
  109. Vidoni, C.; Ferraresi, A.; Esposito, A.; Maheshwari, C.; Dhanasekaran, D.N.; Mollace, V.; Isidoro, C. Calorie Restriction for Cancer Prevention and Therapy: Mechanisms, Expectations, and Efficacy. J. Cancer Prev. 2021, 26, 224–236. [Google Scholar] [CrossRef]
  110. Longo, V.D.; Mattson, M.P. Fasting: Molecular mechanisms and clinical applications. Cell Metab. 2014, 19, 181–192. [Google Scholar] [CrossRef]
  111. Buono, R.; Longo, V.D. Starvation, Stress Resistance, and Cancer. Trends Endocrinol. Metab. 2018, 29, 271–280. [Google Scholar] [CrossRef] [PubMed]
  112. Lashinger, L.M.; O’Flanagan, C.H.; Dunlap, S.M.; Rasmussen, A.J.; Sweeney, S.; Guo, J.Y.; Lodi, A.; Tiziani, S.; White, E.; Hursting, S.D. Starving cancer from the outside and inside: Separate and combined effects of calorie restriction and autophagy inhibition on Ras-driven tumors. Cancer Metab. 2016, 4, 18. [Google Scholar] [CrossRef]
  113. Chen, X.; Lin, X.; Li, M. Comprehensive modulation of tumor progression and regression with periodic fasting and refeeding circles via boosting IGFBP-3 loops and NK responses. Endocrinology 2012, 153, 4622–4632. [Google Scholar] [CrossRef] [PubMed]
  114. Sun, P.; Wang, H.; He, Z.; Chen, X.; Wu, Q.; Chen, W.; Sun, Z.; Weng, M.; Zhu, M.; Ma, D.; et al. Fasting inhibits colorectal cancer growth by reducing M2 polarization of tumor-associated macrophages. Oncotarget 2017, 8, 74649–74660. [Google Scholar] [CrossRef] [PubMed]
  115. Lee, C.; Raffaghello, L.; Brandhorst, S.; Safdie, F.M.; Bianchi, G.; Martin-Montalvo, A.; Pistoia, V.; Wei, M.; Hwang, S.; Merlino, A.; et al. Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy. Sci. Transl. Med. 2012, 4, 124ra127. [Google Scholar] [CrossRef]
  116. Safdie, F.; Brandhorst, S.; Wei, M.; Wang, W.; Lee, C.; Hwang, S.; Conti, P.S.; Chen, T.C.; Longo, V.D. Fasting enhances the response of glioma to chemo- and radiotherapy. PLoS ONE 2012, 7, e44603. [Google Scholar] [CrossRef]
  117. Dorff, T.B.; Groshen, S.; Garcia, A.; Shah, M.; Tsao-Wei, D.; Pham, H.; Cheng, C.-W.; Brandhorst, S.; Cohen, P.; Wei, M.; et al. Safety and feasibility of fasting in combination with platinum-based chemotherapy. BMC Cancer 2016, 16, 360. [Google Scholar] [CrossRef]
  118. Safdie, F.M.; Dorff, T.; Quinn, D.; Fontana, L.; Wei, M.; Lee, C.; Cohen, P.; Longo, V.D. Fasting and cancer treatment in humans: A case series report. Aging (Albany NY) 2009, 1, 988–1007. [Google Scholar] [CrossRef]
  119. de Groot, S.; Vreeswijk, M.P.; Welters, M.J.; Gravesteijn, G.; Boei, J.J.; Jochems, A.; Houtsma, D.; Putter, H.; van der Hoeven, J.J.; Nortier, J.W.; et al. The effects of short-term fasting on tolerance to (neo) adjuvant chemotherapy in HER2-negative breast cancer patients: A randomized pilot study. BMC Cancer 2015, 15, 652. [Google Scholar] [CrossRef]
  120. Messina-Pacheco, J.; Gregorieff, A. A gut feeling: Diet-sensing mesenchymal cells regulate intestinal stem cell function. Cell Res. 2022, 32, 605–606. [Google Scholar] [CrossRef]
  121. Calibasi-Kocal, G.; Mashinchian, O.; Basbinar, Y.; Ellidokuz, E.; Cheng, C.W.; Yilmaz, Ö.H. Nutritional Control of Intestinal Stem Cells in Homeostasis and Tumorigenesis. Trends Endocrinol. Metab. 2021, 32, 20–35. [Google Scholar] [CrossRef] [PubMed]
  122. Harvey, A.E.; Lashinger, L.M.; Otto, G.; Nunez, N.P.; Hursting, S.D. Decreased systemic IGF-1 in response to calorie restriction modulates murine tumor cell growth, nuclear factor-κB activation, and inflammation-related gene expression. Mol. Carcinog. 2013, 52, 997–1006. [Google Scholar] [CrossRef]
  123. Deng, M.; Guerrero-Juarez, C.F.; Sheng, X.; Xu, J.; Wu, X.; Yao, K.; Li, M.; Yang, X.; Li, G.; Xiao, J.; et al. Lepr+ mesenchymal cells sense diet to modulate intestinal stem/progenitor cells via Leptin–Igf1 axis. Cell Res. 2022, 32, 670–686. [Google Scholar] [CrossRef] [PubMed]
  124. Ahmad Sophien, A.N.; Jusop, A.S.; Tye, G.J.; Tan, Y.F.; Wan Kamarul Zaman, W.S.; Nordin, F. Intestinal stem cells and gut microbiota therapeutics: Hype or hope? Front. Med. 2023, 10, 1195374. [Google Scholar] [CrossRef] [PubMed]
  125. Zhang, C.; Li, S.; Yang, L.; Huang, P.; Li, W.; Wang, S.; Zhao, G.; Zhang, M.; Pang, X.; Yan, Z.; et al. Structural modulation of gut microbiota in life-long calorie-restricted mice. Nat. Commun. 2013, 4, 2163. [Google Scholar] [CrossRef]
  126. Remely, M.; Hippe, B.; Geretschlaeger, I.; Stegmayer, S.; Hoefinger, I.; Haslberger, A. Increased gut microbiota diversity and abundance of Faecalibacterium prausnitzii and Akkermansia after fasting: A pilot study. Wien. Klin. Wochenschr. 2015, 127, 394–398. [Google Scholar] [CrossRef]
  127. Plaza-Diaz, J.; Gomez-Llorente, C.; Fontana, L.; Gil, A. Modulation of immunity and inflammatory gene expression in the gut, in inflammatory diseases of the gut and in the liver by probiotics. World J. Gastroenterol. 2014, 20, 15632–15649. [Google Scholar] [CrossRef]
  128. Veselovsky, V.A.; Dyachkova, M.S.; Menyaylo, E.A.; Polyaeva, P.S.; Olekhnovich, E.I.; Shitikov, E.A.; Bespiatykh, D.A.; Semashko, T.A.; Kasianov, A.S.; Ilina, E.N.; et al. Gene Networks Underlying the Resistance of Bifidobacterium longum to Inflammatory Factors. Front. Immunol. 2020, 11, 595877. [Google Scholar] [CrossRef]
  129. Zimina, M.; Babich, O.; Prosekov, A.; Sukhikh, S.; Ivanova, S.; Shevchenko, M.; Noskova, S. Overview of Global Trends in Classification, Methods of Preparation and Application of Bacteriocins. Antibiotics 2020, 9, 553. [Google Scholar] [CrossRef]
  130. Fugaban, J.I.I.; Vazquez Bucheli, J.E.; Park, Y.J.; Suh, D.H.; Jung, E.S.; Franco, B.; Ivanova, I.V.; Holzapfel, W.H.; Todorov, S.D. Antimicrobial properties of Pediococcus acidilactici and Pediococcus pentosaceus isolated from silage. J. Appl. Microbiol. 2022, 132, 311–330. [Google Scholar] [CrossRef]
  131. Savinova, T.; Brzhozovskaya, E.; Alyabieva, N.; Lazareva, A.; Shagin, D.; Mayanskiy, N. Multiple-Drug Resistant Nasopharyngeal Streptococcus pneumoniae Isolated in Russia: Serotypes, Antimicrobial Susceptibility, and Molecular Characterization of the Emergent Serotype 13/ST2754 Lineage. Microb. Drug Resist. 2022, 28, 39–47. [Google Scholar] [CrossRef] [PubMed]
  132. Simons, A.; Alhanout, K.; Duval, R.E. Bacteriocins, Antimicrobial Peptides from Bacterial Origin: Overview of Their Biology and Their Impact against Multidrug-Resistant Bacteria. Microorganisms 2020, 8, 639. [Google Scholar] [CrossRef]
  133. Kaur, S. Bacteriocins as Potential Anticancer Agents. Front. Pharmacol. 2015, 6, 272. [Google Scholar] [CrossRef]
  134. Cesa-Luna, C.; Alatorre-Cruz, J.M.; Carreño-López, R.; Quintero-Hernández, V.; Baez, A. Emerging Applications of Bacteriocins as Antimicrobials, Anticancer Drugs, and Modulators of The Gastrointestinal Microbiota. Pol. J. Microbiol. 2021, 70, 143–159. [Google Scholar] [CrossRef]
  135. Huang, F.; Teng, K.; Liu, Y.; Cao, Y.; Wang, T.; Ma, C.; Zhang, J.; Zhong, J. Bacteriocins: Potential for Human Health. Oxidative Med. Cell. Longev. 2021, 2021, 5518825. [Google Scholar] [CrossRef]
  136. Wang, S.; Ye, Q.; Wang, K.; Zeng, X.; Huang, S.; Yu, H.; Ge, Q.; Qi, D.; Qiao, S. Enhancement of Macrophage Function by the Antimicrobial Peptide Sublancin Protects Mice from Methicillin-Resistant Staphylococcus aureus. J. Immunol. Res. 2019, 2019, 3979352. [Google Scholar] [CrossRef]
  137. Wang, S.; Wang, Q.; Zeng, X.; Ye, Q.; Huang, S.; Yu, H.; Yang, T.; Qiao, S. Use of the Antimicrobial Peptide Sublancin with Combined Antibacterial and Immunomodulatory Activities To Protect against Methicillin-Resistant Staphylococcus aureus Infection in Mice. J. Agric. Food Chem. 2017, 65, 8595–8605. [Google Scholar] [CrossRef]
  138. Antoshina, D.V.; Balandin, S.V.; Bogdanov, I.V.; Vershinina, M.A.; Sheremeteva, E.V.; Toropygin, I.Y.; Finkina, E.I.; Ovchinnikova, T.V. Antimicrobial Activity and Immunomodulatory Properties of Acidocin A, the Pediocin-like Bacteriocin with the Non-Canonical Structure. Membranes 2022, 12, 1253. [Google Scholar] [CrossRef]
  139. Meijerink, M.; van Hemert, S.; Taverne, N.; Wels, M.; de Vos, P.; Bron, P.A.; Savelkoul, H.F.; van Bilsen, J.; Kleerebezem, M.; Wells, J.M. Identification of genetic loci in Lactobacillus plantarum that modulate the immune response of dendritic cells using comparative genome hybridization. PLoS ONE 2010, 5, e10632. [Google Scholar] [CrossRef]
  140. van Hemert, S.; Meijerink, M.; Molenaar, D.; Bron, P.A.; de Vos, P.; Kleerebezem, M.; Wells, J.M.; Marco, M.L. Identification of Lactobacillus plantarum genes modulating the cytokine response of human peripheral blood mononuclear cells. BMC Microbiol. 2010, 10, 293. [Google Scholar] [CrossRef]
  141. Erturk-Hasdemir, D.; Oh, S.F.; Okan, N.A.; Stefanetti, G.; Gazzaniga, F.S.; Seeberger, P.H.; Plevy, S.E.; Kasper, D.L. Symbionts exploit complex signaling to educate the immune system. Proc. Natl. Acad. Sci. USA 2019, 116, 26157–26166. [Google Scholar] [CrossRef] [PubMed]
  142. Round, J.L.; Lee, S.M.; Li, J.; Tran, G.; Jabri, B.; Chatila, T.A.; Mazmanian, S.K. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science 2011, 332, 974–977. [Google Scholar] [CrossRef]
  143. Di Giacinto, C.; Marinaro, M.; Sanchez, M.; Strober, W.; Boirivant, M. Probiotics ameliorate recurrent Th1-mediated murine colitis by inducing IL-10 and IL-10-dependent TGF-beta-bearing regulatory cells. J. Immunol. 2005, 174, 3237–3246. [Google Scholar] [CrossRef]
  144. Karimi, K.; Inman, M.D.; Bienenstock, J.; Forsythe, P. Lactobacillus reuteri-induced regulatory T cells protect against an allergic airway response in mice. Am. J. Respir. Crit. Care Med. 2009, 179, 186–193. [Google Scholar] [CrossRef]
  145. Tang, C.; Kamiya, T.; Liu, Y.; Kadoki, M.; Kakuta, S.; Oshima, K.; Hattori, M.; Takeshita, K.; Kanai, T.; Saijo, S.; et al. Inhibition of Dectin-1 Signaling Ameliorates Colitis by Inducing Lactobacillus-Mediated Regulatory T Cell Expansion in the Intestine. Cell Host Microbe 2015, 18, 183–197. [Google Scholar] [CrossRef]
  146. Kullberg, M.C.; Jankovic, D.; Gorelick, P.L.; Caspar, P.; Letterio, J.J.; Cheever, A.W.; Sher, A. Bacteria-triggered CD4(+) T regulatory cells suppress Helicobacter hepaticus-induced colitis. J. Exp. Med. 2002, 196, 505–515. [Google Scholar] [CrossRef]
  147. Shen, Y.; Giardino Torchia, M.L.; Lawson, G.W.; Karp, C.L.; Ashwell, J.D.; Mazmanian, S.K. Outer membrane vesicles of a human commensal mediate immune regulation and disease protection. Cell Host Microbe 2012, 12, 509–520. [Google Scholar] [CrossRef]
  148. 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]
  149. 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]
  150. Huang, F.; Li, S.; Chen, W.; Han, Y.; Yao, Y.; Yang, L.; Li, Q.; Xiao, Q.; Wei, J.; Liu, Z.; et al. Postoperative Probiotics Administration Attenuates Gastrointestinal Complications and Gut Microbiota Dysbiosis Caused by Chemotherapy in Colorectal Cancer Patients. Nutrients 2023, 15, 356. [Google Scholar] [CrossRef]
  151. Chen, Y.; Qi, A.; Teng, D.; Li, S.; Yan, Y.; Hu, S.; Du, X. Probiotics and synbiotics for preventing postoperative infectious complications in colorectal cancer patients: A systematic review and meta-analysis. Tech. Coloproctol. 2022, 26, 425–436. [Google Scholar] [CrossRef] [PubMed]
  152. Yoon, B.J.; Oh, H.K.; Lee, J.; Cho, J.R.; Kim, M.J.; Kim, D.W.; Kang, S.B. Effects of probiotics on bowel function restoration following ileostomy closure in rectal cancer patients: A randomized controlled trial. Colorectal Dis. 2021, 23, 901–910. [Google Scholar] [CrossRef]
  153. Rodríguez-Padilla, Á.; Morales-Martín, G.; Pérez-Quintero, R.; Gómez-Salgado, J.; Balongo-García, R.; Ruiz-Frutos, C. Postoperative Ileus after Stimulation with Probiotics before Ileostomy Closure. Nutrients 2021, 13, 626. [Google Scholar] [CrossRef] [PubMed]
  154. Liu, C.; Ng, S.-K.; Ding, Y.; Lin, Y.; Liu, W.; Wong, S.H.; Sung, J.J.-Y.; Yu, J. Meta-analysis of mucosal microbiota reveals universal microbial signatures and dysbiosis in gastric carcinogenesis. Oncogene 2022, 41, 3599–3610. [Google Scholar] [CrossRef]
  155. Yokoyama, Y.; Nishigaki, E.; Abe, T.; Fukaya, M.; Asahara, T.; Nomoto, K.; Nagino, M. Randomized clinical trial of the effect of perioperative synbiotics versus no synbiotics on bacterial translocation after oesophagectomy. Br. J. Surg. 2014, 101, 189–199. [Google Scholar] [CrossRef] [PubMed]
  156. Yokoyama, Y.; Miyake, T.; Kokuryo, T.; Asahara, T.; Nomoto, K.; Nagino, M. Effect of Perioperative Synbiotic Treatment on Bacterial Translocation and Postoperative Infectious Complications after Pancreatoduodenectomy. Dig. Surg. 2016, 33, 220–229. [Google Scholar] [CrossRef]
  157. Nakamura, K.; Kato, K.; Igaki, H.; Ito, Y.; Mizusawa, J.; Ando, N.; Udagawa, H.; Tsubosa, Y.; Daiko, H.; Hironaka, S.; et al. Three-arm phase III trial comparing cisplatin plus 5-FU (CF) versus docetaxel, cisplatin plus 5-FU (DCF) versus radiotherapy with CF (CF-RT) as preoperative therapy for locally advanced esophageal cancer (JCOG1109, NExT study). Jpn. J. Clin. Oncol. 2013, 43, 752–755. [Google Scholar] [CrossRef]
  158. Fukaya, M.; Yokoyama, Y.; Usui, H.; Fujieda, H.; Sakatoku, Y.; Takahashi, T.; Miyata, K.; Niikura, M.; Sugimoto, T.; Asahara, T.; et al. Impact of synbiotics treatment on bacteremia induced during neoadjuvant chemotherapy for esophageal cancer: A randomised controlled trial. Clin. Nutr. 2021, 40, 5781–5791. [Google Scholar] [CrossRef]
  159. Motoori, M.; Sugimura, K.; Tanaka, K.; Shiraishi, O.; Kimura, Y.; Miyata, H.; Yamasaki, M.; Makino, T.; Miyazaki, Y.; Iwama, M.; et al. Comparison of synbiotics combined with enteral nutrition and prophylactic antibiotics as supportive care in patients with esophageal cancer undergoing neoadjuvant chemotherapy: A multicenter randomized study. Clin. Nutr. 2022, 41, 1112–1121. [Google Scholar] [CrossRef]
  160. Sugimoto, T.; Atobe, S.; Kado, Y.; Takahashi, A.; Motoori, M.; Sugimura, K.; Miyata, H.; Yano, M.; Tanaka, K.; Doki, Y.; et al. Gut microbiota associated with the mitigation effect of synbiotics on adverse events of neoadjuvant chemotherapy in patients with esophageal cancer: A retrospective exploratory study. J. Med. Microbiol. 2023, 72, 001723. [Google Scholar] [CrossRef]
  161. Xourafas, D.; Pawlik, T.M.; Cloyd, J.M. Early Morbidity and Mortality after Minimally Invasive Liver Resection for Hepatocellular Carcinoma: A Propensity-Score Matched Comparison with Open Resection. J. Gastrointest. Surg. 2019, 23, 1435–1442. [Google Scholar] [CrossRef] [PubMed]
  162. Gan, Y.; Su, S.; Li, B.; Fang, C. Efficacy of Probiotics and Prebiotics in Prevention of Infectious Complications Following Hepatic Resections: Systematic Review and Meta-Analysis. J. Gastrointestin Liver Dis. 2019, 28, 205–211. [Google Scholar] [CrossRef] [PubMed]
  163. Roussel, E.; Brasse-Lagnel, C.; Tuech, J.J.; Montialoux, H.; Papet, E.; Tortajada, P.; Bekri, S.; Schwarz, L. Influence of Probiotics Administration Before Liver Resection in Patients with Liver Disease: A Randomized Controlled Trial. World J. Surg. 2022, 46, 656–665. [Google Scholar] [CrossRef]
  164. Zhou, W.; Zhang, D.; Li, Z.; Jiang, H.; Li, J.; Ren, R.; Gao, X.; Wang, X.; Wang, W.; Yang, Y. The fecal microbiota of patients with pancreatic ductal adenocarcinoma and autoimmune pancreatitis characterized by metagenomic sequencing. J. Transl. Med. 2021, 19, 215. [Google Scholar] [CrossRef] [PubMed]
  165. Cogo, E.; Elsayed, M.; Liang, V.; Cooley, K.; Guerin, C.; Psihogios, A.; Papadogianis, P. Probiotics Evaluation in Oncological Surgery: A Systematic Review of 36 Randomized Controlled Trials Assessing 21 Diverse Formulations. Curr. Oncol. 2021, 28, 5192–5214. [Google Scholar] [CrossRef]
  166. Ishibashi, N.; Yamazaki, S. Probiotics and safety. Am. J. Clin. Nutr. 2001, 73, 465s–470s. [Google Scholar] [CrossRef] [PubMed]
  167. Sanders, M.E.; Akkermans, L.M.; Haller, D.; Hammerman, C.; Heimbach, J.; Hörmannsperger, G.; Huys, G.; Levy, D.D.; Lutgendorff, F.; Mack, D.; et al. Safety assessment of probiotics for human use. Gut Microbes 2010, 1, 164–185. [Google Scholar] [CrossRef]
  168. 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]
Figure 1. Underlying causes of malnutrition in GI cancers. Nutritional alterations are due to various factors: reduction in energy intake and poor physical activity; consequences of chemoradiotherapy such as nausea, vomiting, taste changes, and cancer-related causes such as systemic inflammation and difficulty absorbing nutrients (due to destruction of the GI architecture). Alterations in the GM composition may be attributed to all three factors described (image created with Biorender.com).
Figure 1. Underlying causes of malnutrition in GI cancers. Nutritional alterations are due to various factors: reduction in energy intake and poor physical activity; consequences of chemoradiotherapy such as nausea, vomiting, taste changes, and cancer-related causes such as systemic inflammation and difficulty absorbing nutrients (due to destruction of the GI architecture). Alterations in the GM composition may be attributed to all three factors described (image created with Biorender.com).
Nutrients 15 04408 g001
Figure 2. Nutritional Care Workflow for GI Cancer Surgery Patients. (1) The first step in planning nutritional care for surgical cancer patients involves screening and assessment of nutritional status. (2) This is followed by a nutrition diagnosis that commonly includes identifying conditions such as malnutrition, cachexia, or sarcopenia in cancer patients. (3) At least, the appropriate nutrition therapy (enteral nutrition, parental nutrition, oral nutritional supplement, or nutrition counseling) should be determined. (4) Postoperatively and during the discharge period, patients are closely monitored, and it is beneficial to reevaluate their nutritional status (image created with Biorender.com).
Figure 2. Nutritional Care Workflow for GI Cancer Surgery Patients. (1) The first step in planning nutritional care for surgical cancer patients involves screening and assessment of nutritional status. (2) This is followed by a nutrition diagnosis that commonly includes identifying conditions such as malnutrition, cachexia, or sarcopenia in cancer patients. (3) At least, the appropriate nutrition therapy (enteral nutrition, parental nutrition, oral nutritional supplement, or nutrition counseling) should be determined. (4) Postoperatively and during the discharge period, patients are closely monitored, and it is beneficial to reevaluate their nutritional status (image created with Biorender.com).
Nutrients 15 04408 g002
Figure 3. Overview of the potential benefits of immunonutrition and microbiota modulation in gastrointestinal cancer patient care. The combined effect of nutritional intervention and gut microbiota modulation can comprehensively improve patient care by reducing infection complications, shortening the hospital stay, improving the overall nutritional status, ameliorating the host immune response, and enhancing the effects of conventional anticancer treatments. At the intracellular level, an antiproliferative effect, an increase in apoptosis, and the inhibition of cancer invasion after nutritional and microbiota modulation interventions were described (image created with Biorender.com).
Figure 3. Overview of the potential benefits of immunonutrition and microbiota modulation in gastrointestinal cancer patient care. The combined effect of nutritional intervention and gut microbiota modulation can comprehensively improve patient care by reducing infection complications, shortening the hospital stay, improving the overall nutritional status, ameliorating the host immune response, and enhancing the effects of conventional anticancer treatments. At the intracellular level, an antiproliferative effect, an increase in apoptosis, and the inhibition of cancer invasion after nutritional and microbiota modulation interventions were described (image created with Biorender.com).
Nutrients 15 04408 g003
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

Martinelli, S.; Lamminpää, I.; Dübüş, E.N.; Sarıkaya, D.; Niccolai, E. Synergistic Strategies for Gastrointestinal Cancer Care: Unveiling the Benefits of Immunonutrition and Microbiota Modulation. Nutrients 2023, 15, 4408. https://doi.org/10.3390/nu15204408

AMA Style

Martinelli S, Lamminpää I, Dübüş EN, Sarıkaya D, Niccolai E. Synergistic Strategies for Gastrointestinal Cancer Care: Unveiling the Benefits of Immunonutrition and Microbiota Modulation. Nutrients. 2023; 15(20):4408. https://doi.org/10.3390/nu15204408

Chicago/Turabian Style

Martinelli, Serena, Ingrid Lamminpää, Eda Nur Dübüş, Dilara Sarıkaya, and Elena Niccolai. 2023. "Synergistic Strategies for Gastrointestinal Cancer Care: Unveiling the Benefits of Immunonutrition and Microbiota Modulation" Nutrients 15, no. 20: 4408. https://doi.org/10.3390/nu15204408

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