Abstract
Introduction
Cancer patient’s survival has gradually improved due to immune checkpoint inhibitors (ICIs). Several studies showed a possible association between the intestinal microbiome and ICI efficacy. Strategies for modifying the composition of the gut microbiome encompass various dietary interventions, which may have distinct impacts on the outcomes of ICI-treated patients. In our systematic review, we explored how dietary habits correlate with therapeutic responses in cancer patients and cancer mouse models undergoing immunotherapy.
Methods
A systematic review was conducted using search terms: “cancer”, “immunotherapy”, “diet”, and “microbiome”, from Medline, Web of Science, Scopus, and Cochrane Library databases. The outcomes in the clinical studies were overall response rate (ORR), overall survival (OS), or progression-free survival (PFS) in human studies. In mouse studies, change in tumor size was the endpoint. The comparator attributions were questionnaire-based dietary interventions.
Results
Nineteen articles met the inclusion criteria and were included in the review (6 prospective cohort studies, 1 cross-sectional observational study, and 12 mouse studies). A consistent association was observed between high (vs. low) fiber consumption and improved therapeutic response with a pooled odds ratio of 5.79 when including all human prospective cohort studies. In mice, limited availability of methionine, cysteine, and low intake of leucine and glutamine was linked to reduced tumor progression. Combining ICIs with intermittent fasting or a fasting-mimicking diet significantly decreased tumor volume in mouse melanoma models. In humans, a higher relative abundance of short-chain fatty acid (SCFA) and lactic acid-producing bacteria—particularly Faecalibacterium prausnitzii and Akkermansia muciniphila—correlated with objective response rates (ORR). Similar microbiome alterations were observed in mouse models. Increased fiber intake enhanced ICI efficacy in mice by modulating the gut microbiome, primarily via elevated SCFA production—an effect also reflected in human studies.
Conclusion
Intermittent fasting, high fiber, and low sugar consumption are significantly associated with better ICI outcomes. The studies revealed alterations in microbiota composition linked to diet, and these findings were confirmed in animal models, regarding the production of SCFAs and lactic acid, as well as an increase in Bacteroidota/Bacillota ratio and microbial diversity.
Supplementary Information
The online version contains supplementary material available at 10.1186/s12967-025-06586-0.
Keywords: Gut microbiome, Cancer, Immunotherapy, Diet, Fiber intake, Ketogenic diet
Introduction
The diet-related gut microbiota changes might affect ICI efficacy, and there can be differences between the microbial communities of responders and non-responders. Cancer immunotherapy represents a significant advancement in patient outcomes in oncology. Various cancers have responded to immunotherapy, albeit with a need to understand the underlying mechanisms [1]. Despite its potential, there is a requirement for improving the response rate of immune checkpoint inhibition (ICI), which is currently at 15–20% in good performance status [2].
Bacteria densely inhabit the intestines and continuously interact with the immune system. The combined genetic material of bacteria and other microorganisms in this ecological community has been the subject of growing scientific interest over the last twenty years [3]. The gut microbiota also serves as a host metabolism regulator, while influencing the appropriate maturation and operation of the immune system [4]. The anti-cancer immune response might be modulated by gut microbiota through antitumor CD8 T-cell or macrophage maturation in the gut [5, 6]. Nonetheless, disruption of the intestinal microbiota due to external factors (e.g. dietary habits, antibiotic consumption), or immune system alterations may lead to widespread dissemination of detrimental microorganisms, increased vulnerability to harmful microbial infiltration, and irregular immune reactions [3, 7]. Growing evidence corroborates the involvement of the microbiome in the efficacy of cancer treatment, as evidenced by several recent studies highlighting the impact of the intestinal microbiome, particularly on the effectiveness of ICI in various forms of cancer, including melanoma, lung, and renal cancer [8–13].
Several bacterial species have been associated with improved immune checkpoint inhibitor (ICI) responses. Akkermansia muciniphila has been shown to enhance ICI efficacy by stimulating cytokine secretion via MHC Class II-restricted CD4+T cells and dendritic cells, as reported in studies by Routy et al. [9] and Lu et al. [14]. Certain Bacteroides species, including B. thetaiotaomicron and B. fragilis, have been linked to the induction of TH1 immune responses and dendritic cell maturation in tumor-draining lymph nodes, as demonstrated by earlier [15, 16]. Bifidobacterium species, such as B. breve and B. longum, have been associated with increased accumulation of antigen-specific CD8+tumor-infiltrating lymphocytes (TILs) and MHC Class II dendritic cells, as observed in multiple studies [4, 17]. Members of the Ruminococcaceae family, including R. obeum and R. bromii, have been shown to elevate levels of CD4+ and CD8+T cells, enhancing anti-tumor immunity [9, 18]. Additionally, Faecalibacterium prausnitzii has been identified as a beneficial gut bacterium in ICI responders, potentially playing a role in immunomodulation, according to findings by Frankel et al. [16] and Botticelli et al. [19].
Alterations in gut microbiota might enhance ICI-efficacy, however, less is known about dietary interventions. More data is available connected to prebiotics or probiotics that have the potential to modify gastrointestinal microbiome composition [15]. Recent research has identified associations between favorable outcomes and diets such as ketogenic, plant-based, and microbiome-supporting regimens [16]. Ongoing clinical trials investigate the impact of dietary modifications, such as enhanced fiber consumption or personalized dietary interventions, on the microbiome’s composition and its corresponding clinical consequences. An increasing array of data concerning probiotics and shifts in microbial composition might influence the efficacy of immunotherapeutic methods; still, it is vital to establish particular dietary alterations associated with changes in the microbiome and beneficial microbial entities [15].
Even though different dietary patterns -and in parallel- different microbial communities can be observed in cancer ICI responder vs. non-responder patients; the mechanistic relations remain unclear. Analysis of pre-clinical studies along with clinical trial data evaluation might help to understand the underlying biology and complex interactions between microbiota and ICI efficacy. In this systematic review, we aimed to compile the current preclinical and clinical data on nutritional factors, associated gut microbiome taxonomy, and functionality for ICI efficacy.
Methods
The systematic search was conducted according to the principles established by the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) statement [17].
Study selection
To obtain relevant publications, the scanning of Scopus, Cochrane Library, Web of Science and Medline (through the Pubmed interface) was conducted and completed in 30 March 2024. The inquiry consisted of the following terms: “diet”, “cancer”, “immunotherapy”, “microbiome”, along with their corresponding terms and connections consolidated utilizing the boolean operator OR, succeeded by multiple limitations employing the AND operator. The exact search key can be found in Supplementary file 1. The query hits generated were archived in RIS format and subsequently brought into the abstract management tool Rayyan [18]. Research articles meeting the criteria for full-text examination were acquired via the subscription of Semmelweis University and preserved within Rayyan.
Inclusion criteria
According to PICO framework, we posed the subsequent question: What are the principal dietary components that, in conjunction with the intestinal microbiome, may impact the results of immunotherapy? Based on the aforementioned scope of the study, the following inclusion criteria were prepared:
-
(I)
Diagnosis of solid cancer patients (early and advanced-stage)
-
(II)
Cancer Patients treated with immune-checkpoint blockade (anti-PD1, anti-PD-L1 and anti-CTLA4 or other ICI drugs included; neoadjuvant, adjuvant and advanced stage IIIB-IV treatments included; chemotherapy (CHT) + ICI, chemo-radiation (CRT) + ICI combinations are also allowed). Inclusion in clinical trials is permitted. The outcomes were reported in terms of response rate, overall response rate (ORR), progression-free survival (PFS), or overall survival (OS).
-
(III)
Microbiome measured in stool samples
-
(IV)
Microbial data performed by shotgun metagenomics or 16S RNA-sequencing
-
(V)
Any nutrition or diet-related information available such as interventions or questionnaires were allowed
-
(VI)
Mouse or human studies were allowed
Exclusion criteria
-
(I)
Reviews, systematic reviews, meta-analyses, editorials, correspondences, case series, case reports, commentaries, letters to editor, protocols, and conference abstracts
-
(II)
Cancer vaccination, vaccines
-
(III)
Exclusively in silico studies on open-access data
-
(IV)
Primarily probiotic treatment without dietary information
-
(V)
Specific plant extracts as intervention or unknown composition of the nutritional intervention
-
(VI)
Purely in vitro (cell line, cell culture) or ex vivo (organoid) studies
Manual selection
Two researchers (GSz and CsS) independently evaluated the abstracts according to the predetermined inclusion/exclusion criteria using a three-tiered (include/exclude/maybe) rating system. Should any disparities arise in the evaluation, the publication was subjected to a comprehensive full-text examination. During the full-text assessment, the same researchers scrutinized the publications with unrestricted access to information.
Data extraction
The consecutive information was extracted from the studies: first author name, publication year, population, tumor type, treatment, observed dietary habit, type of the available microbial data, and outcome (Table 1). In studies where the mentioned data was not assessed, we used the not assessed (NA) sign in the tables. Additional information was obtained to provide a comprehensive summary of the results of the studies (Supplementary Table 1) Information on the comparision of the response and microbial alterations in association with the different dietary interventions, alpha and beta diversity measures, Firmicutes/ Bacteroidetes ratios (F/B ratio) (Table 2), and increased and decreased abundance of certain bacterial taxa after treatment (Table 3) were extracted from articles.
Table 1.
Summary of included studies
| Study | Year | Population | Type of study | Tumor type | Treatment | Observed diet | Microbial data | Outcome |
|---|---|---|---|---|---|---|---|---|
| Bolte et al. | [19] | Human | Prospective cohort study | Melanoma | Ipilimumab-nivolumab combi treatment (anti PD-1) + (anti CTLA-4), or (anti PD-1/anti PD-L1) + (anti CTLA-4) | Mediterranean diet | – | ORR, PFS at 12 months |
| Pietrzak et al. | [36] | Human | Prospective cohort study | Melanoma | Nivolumab or pembrolizumab (anti PD-1) | Overall habits (protein, fiber comsumption etc..) | NGS | ORR |
| Simpson et al. | [21] | Human | Prospective cohort study | Melanoma | Ipilimumab and nivolumab combi (anti PD-1) + (anti CTLA-4) | Overall habits (protein, fiber comsumption etc..) | – | Response rate |
| Spencer et al. | [20] | Human | Prospective cohort study | Melanoma | Anti-CTLA4 + anti-PD-1 combi, or anti-PD-1 monotherapy | Fiber intake | Metagenomics | ORR (responder: CR, PR, SD: PFS >= 6 months) |
| Nomura et al. | [34] | Human | Observational cross-sectional study | Solid tumors | Pembrolizumab, nivolumab | Overall habits (protein, fiber comsumption etc..) | – | ORR, PFS |
| Golčić et al. | [24] | Human | Experimental randomised animal study | Melanoma | Pembrolizumab, nivolumab, or nivolumab + ipilimumab | Overall habits (pro-inflammatory and anti-inflammatory etc...) | Metagenomics | ORR |
| Ferrere et al. | [27] | Mouse | Experimental randomised animal study | Melanoma | Anti-CTLA4 + anti-PD-1 combi or anti-CTLA4/ anti-PD-1 monotherapy | Ketogenic diet | – | Tumor size growth |
| Boucher et al. | [25] | Mouse | Experimental randomised animal study | Melanoma, fibrosarcoma, colorectal cancer | Anti-PD-1 | Inulin-enriched diet | 16S rRNA sequencing | Tumor size growth |
| Han et al. | [28] | Mouse | Experimental randomised animal study | Colon carcinoma | Anti-PD-1 | Inulin-enriched diet | 16S rRNA sequencing | Tumor size growth |
| Zhang et al. | [1] | Mouse | Experimental randomised animal study | Colon carcinoma | Anti-PD-1 | The effect of FMT and pectin-enriched diet | – | Tumor size growth |
| Ji et al. | [52] | Mouse | Experimental randomised animal study | Colon carcinoma | Anti-PD-1 | Methionine-enriched diet | 16S rRNA sequencing, metatranscriptomics | Tumor size growth |
| Kim et al. | [45] | Mouse | Experimental randomised animal study | Glioma | Anti-PD-1 | High glucose diet | 16S rRNA sequencing | Tumor size growth |
| Udumula et al. | [2] | Mouse | Experimental randomised animal study | Epithelial ovarian cancer | Anti-PD-1 | Intermittent fasting | – | Tumor size growth |
| Tanaka et al. | [22] | Human | Prospective cohort study | Lung cancer | Anti-PD-1 (pembrolizumab) | Overall habits (protein, fiber comsumption etc..) | – | OS > 3 years, OS < 3 years |
| Cortellino et al. | [63] | Mouse | Experimental randomised animal study | Melanoma, lung cancer | Anti-PD-1, anti-PD-1, anti-PD-L1, anti-CTLA4 | Fasting | – | Tumor size growth |
| Calderón-Montaño et al. | [53] | Mouse | Experimental randomised animal study | Renal cell carcinoma | Anti-PD-1 | Vitamin, mineral and amino-acid enriched diets | – | Overall survival (developed symptoms of advanced disease and were sacrificed x days after tumor cell injection) |
| Kuehm et al. | [64] | Mouse | Experimental randomised animal study | Melanoma, colon carcinoma | Anti-PD-1, anti-CTLA-4, anti-LAG-3 | Western and fructose enriched diet | Taxon specific sequencing PCR | Tumor size growth |
| Lam et al. | [65] | Mouse | Experimental randomised animal study | Lymphoma, colon carcinoma, melanoma | Anti-PD-1, anti-PD-L1 | Western and pectin enriched diet | 16S rRNA sequencing | Tumor size growth |
| Li et al. | [66] | Mouse | Experimental randomised animal study | Melanoma, colon carcinoma | Anti-PD-1 | Inulin and mucin enriched diet | 16S rRNA sequencing | Tumor size growth |
Table 2.
Diversity metrics and F/B ratio identified in the included studies, categorized by ICI-responder groups
| Study | Alpha-diversity in responders/in the treatment group | Beta-diversity in responders/in the treatment group | Firmicutes/Bacteroides ratio in responders |
|---|---|---|---|
| Bolte et al. [19] | NA | NA | NA |
| Pietrzak et al. [36] | Decreased | Altered | Increased |
| Simpson et al. [21] | Not different | NA | NA |
| Spencer et al. [20] | Not different | NA | NA |
| Nomura et al. [34] | NA | NA | NA |
| Golčić et al. [24] | No signifincant difference between early and late responders | Increased in late responders | NA |
| Ferrere et al. [27] | NA | Altered | NA |
| Boucher et al. [25] | Altered | Altered | NA |
| Han et al. [28] | NA | NA | NA |
| Zhang et al. [1] | Increased | Altered | Increased |
| Ji et al. [52] | NA | NA | Increased |
| Kim et al. [45] | NA | NA | NA |
| Udumula et al. [2] | NA | NA | NA |
| Tanaka et al. [22] | NA | NA | NA |
| Cortellino et al. [63] | NA | NA | NA |
| Calderón-Montaño et al. [53] | NA | NA | NA |
| Kuehm et al. [64] | NA | NA | NA |
| Lam et al. [65] | NA | NA | NA |
| Li et al. [66] | NA | NA | NA |
Table 3.
Differentially abundant bacterial taxa in the included studies categorized by ICI-responder groups
| Study | Increased abundance in responders | Decreased abundance in responders | Increased abundance in non-responders | Decreased abundance in non-responders | Increased abundance in treated | Decreased abundance in treated |
|---|---|---|---|---|---|---|
| Bolte et al. [19] | NA | NA | NA | NA | NA | NA |
| Pietrzak et al. [36] | Prevotella copri, Bacteroides uniformis | NA | Faecalibacterium prausnitzii, Desulfovibrio intestinalis | NA | NA | NA |
| Simpson et al. [21] | Faecalibacterium prausnitzii, Butyricicoccus pullicaecorum, Akkermansia muciniphilia | NA | NA | Methanogenic archaea, Akkermansia muciniphilia, and Ruminococcaceae | NA | NA |
| Spencer et al. [20] | Ruminococcaceae | NA | NA | NA | NA | NA |
| Nomura et al. [34] | NA | NA | NA | NA | NA | NA |
| Golčić et al. [24] | Higher abundance of Prevotellaceae in early responders | NA | NA | NA | NA | NA |
| Ferrere et al. [27] | NA | NA | NA | NA | Akkermansia muciniphila, Ruthenibacterium lactatiformans, Pseudoflavonifractor capillosus, Eisenbergiella massiliensis | Clostridium asparagiforme, Lactobacillus caviae, Lactobacillus taiwanensis, Lactobacillus reuteri, Lactobacillus gasseri, Lactobacillus hominis, Lactobacillaceae |
| Boucher et al. [25] | NA | NA | NA | NA | Bifidobacterium (Actinobacteriota), Bifidobacterium animalis subsp. animalis | Clostridium saudiense, Clostridium disporicum, and Clostridium celatum (Firmicutes) |
| Han et al. [28] | Akkermansia, Lactobacillus, Roseburia | Oscillibater | NA | NA | NA | NA |
| Zhang et al. [1] | Ruminococcaceae, Faecalibacterium, and Holdemania | NA | NA | NA | Lactobacillaceae, Bifidobacteriaceae, Erysipelotrichaceae, and Ruminococcaceae | NA |
| Ji et al. [52] | NA | NA | NA | NA | NA | NA |
| Kim et al. [45] | NA | NA | NA | NA | Erysipelotrichaceae, Desulfovibrionaceae, AC160630_f, Rikenellaceae, Odoribacteraceae | FR888536_f,Phophyromonadaceae, Lactobacillaceae, Lachnospiraceae |
| Udumula et al. [2] | NA | NA | NA | NA | NA | NA |
| Tanaka et al. [22] | NA | NA | NA | NA | NA | NA |
| Cortellino et al. [63] | NA | NA | NA | NA | NA | NA |
| Calderón-Montaño et al. [53] | NA | NA | NA | NA | NA | NA |
| Kuehm et al. [64] | NA | NA | NA | NA | Allobaculum, Clostridium, Parabacteroides, Lactococcus, Epulopiscium, Peptostreptococcaceae, Coriobacteriaceae | Rikenellaceae, RF39, Roseburia, Turicibacter, Anaeroplasma |
| Lam et al. [65] | NA | NA | NA | NA | Enterobacteriales, Verrucomicrobiales, Betaproteobacteriales | Firmicutes, Clostridiales and Lactobacillales |
| Li et al. [66] | NA | NA | NA | NA | Actinobacteria, Bifidobacterium longum, Olsenella spp | NA |
NA not available
Meta-analysis of fiber-high vs fiber-low diets
To provide a quantitative estimate of the association between fiber-rich dietary patterns and ICI response, we performed a limited random-effects meta-analysis using four prospective cohort studies that stratified patients into responders versus non-responders. For each study, we extracted or reconstructed 2 × 2 contingency tables comparing the most extreme diet categories (e.g., highest vs. lowest fiber or SCFA intake). Odds ratios (ORs) and 95% confidence intervals were calculated individually and pooled using a DerSimonian-Laird random-effects model. Notably, the data from Bolte et al. [19] were approximated based on reported response probabilities from logistic regression outputs at extreme Mediterranean diet adherence scores and were included to provide comparative context; these estimates should be interpreted cautiously due to the absence of raw group counts.
Results
A sum of 1155 research papers were evaluated in our comprehensive exploration of Medline, Web of Science, Scopus, and Cochrane Library databases. The procedure of selecting studies is illustrated in Fig. 1. Before the screening, 386 duplicated records were removed from the list, and another 19, due to the absence of the DOI code. Upon completion of the abstract screening process, 833 abstracts were deemed irrelevant and subsequently excluded. Afterwards, 322 studies underwent full-text screening, of which 293 were excluded for the same reason of irrelevance or not meeting inclusion and/or exclusion criteria. Of the 29 residual studies, 10 were disregarded due to the ineligibility of the applied intervention. Ultimately, 19 studies met the selection criteria and were incorporated into the systematic review.
Fig. 1.
PRISMA flow chart of the used search methodology for identifying relevant papers. A systematic search of Web of Science, Medline, Cochrane Library, and Scopus was conducted to identify studies in which the immune function influential effect of dietary interventions were assessed in immune checkpoint inhibitor-treated mice and men. 19 studies underwent assessment based on predetermined eligibility criteria, with 10 being deemed unsuitable due to inappropriate intervention
Quality assessment
The Newcastle–Ottawa scale (NOS) was employed to quantitatively evaluate the methodological quality of the 7 prospective cohort studies included. The evaluation was based on the Coding Manual for case–control and control studies, which assessed the study groups’ selection, comparability, and exposure/outcome aspects. None of the studies were identified as having a high risk of bias (0–5) according to the NOS criteria Supplementary Table 2.
The quality assessment of the animal studies was performed by the Systematic Review Center for Laboratory Aminal Experimentation (SYRCLE) risk of bias assessment tools. The findings regarding the attribution of bias across each domain of SYRCLE’s assessment tool are illustrated in Fig. 2. It is important to highlight that none of the studies sufficiently clarified whether proper blinding was applied regarding the caregivers’ or investigators’ awareness of the specific interventions administered to each animal throughout the research, or whether the outcome assessors were blinded or not. Also, all studies exhibited an absence of selective outcome reporting and did not manifest any additional discernible issues that might contribute to a heightened risk of bias.
Fig. 2.
The potential for bias within the specific animal studies has been taken into consideration. The SYRCLE’s Risk of Bias tool was employed to evaluate the subsequent domains: 1, Was the allocation sequence adequately generated and applied? 2, Were the groups similar at baseline, or were they adjusted for confounders in the analysis? 3, Was the allocation to the different groups adequately concealed? 4, Were the animals randomly housed during the experiment? 5, Were the caregivers and/or investigators blinded from knowledge of which intervention each animal received during the experiment? 6, Were animals selected at random for outcome assessment? 7, Was the outcome assessor-blinded? 8, Were incomplete outcome data adequately addressed? 9, Are reports of the study free of selective outcome reporting? 10, Was the study apparently free of other problems that could result in high risk of bias? Abbreviations and symbols: Red circle: high bias, Blue circle: Not evaluated, Yellow circle: Unknown bias, Green circle: Low bias
Study characteristics
The study characteristics are shown in Table 1. The used articles included 6 prospective cohort studies, and 1 observational cross-sectional study and 12 experimental randomized animal studies. The observed tumor types were melanoma (n = 6 human and n = 5 mouse), non-small cell lung cancer (NSCLC) in humans (n = 1), various types of solid tumors including head and neck cancer (n = 1), gastrointestinal cancer (n = 1), urothelial cancer (n = 1), glioma (n = 1), epithelial ovarian cancer (n = 2), colon carcinoma (n = 7 mouse), lymphoma (n = 1), renal cell carcinoma (n = 1 human and n = 1 mouse), and sarcoma (n = 1 human and n = 1 mouse). The applied tumor cell lines in mouse models are presented in Table 1. All of the study subjects were immune-checkpoint inhibitor (ICI) treated, only in the Pietrzak et al. [36] study observed 10 healthy individuals.
The study subjects (both humans and mice) were treated with anti-PD-1, anti-PDL-1, or anti-CTLA-4 inhibitors in monotherapy or combination therapy setup. In the case of human studies, information related to dietary habits were assessed by food frequency questionnaires. Some studies employed custom-made surveys, others used broadly accepted questionnaires to get more reliable and comparable results like EPIC-Norfolk FFQ [19] or National Cancer Institute Dietary Screener Questionnaire [20]. Food Frequency Questionnaire (FQ18N) developed by Wageningen University and Research, Cancer Council Victoria Dietary Questionnaire for Epidemiological Studies [21] and brief-type self-administered diet history questionnaire (BDHQ) [22]. From the surveys, calculations were made for macronutrient, micronutrient, and calorie consumption with a subsequent comparison of the findings between responders and non-responders, early or late responders, as well as long-term versus short-term survivors. The outcomes were reported in terms of overall response rate (ORR), progression-free survival (PFS), or overall survival (OS). In the case of pre-clinical studies, distinctions in microbial composition were evaluated through fecal sample sequencing, including 16S (n = 7) and shotgun (n = 2) and response assessment of tumor volume via caliper-based diameter measurements. Subcutaneous (n = 9) or orthotopic (n = 3) mice tumor models were used in the studies included in our systematic review.
High versus low fiber intake was observed in the relation of prolonged survival in the human studies. The alternate Mediterranean diet score (aMED), which measures adherence to a traditional Mediterranean diet marked by high consumption of vegetables, legumes, nuts, fruits, whole grains, and fish and low intake of red and processed meats, was used to evaluate dietary patterns in the study by Bolte et al. [19]. Furthermore, the percentage of plant-derived versus animal-derived foods was assessed using the original plant-based diet index (oPDI), which was further divided into two categories: the unhealthy plant-based diet index (u-PDI), which includes less nutrient-dense plant-based items like juices and refined grains, and the healthy plant-based diet index (h-PDI), which prioritizes unprocessed plant-based foods. Different thresholds have been used in different studies to determine high fiber consumption. While Simpson et al. [21] cited the Australian Dietary Guidelines, which suggest 30 g of fiber per day for males and 25 g per day for women, Pietrzak et al. [36] defined high fiber consumption as 20 g or more per day. Similarly, the criterion for a high fiber intake was set at 20 g per day by Spencer et al. [20]. Frequent use of foods high in fiber, such as green vegetables, cabbage, and mushrooms, was linked to higher levels of fecal short-chain fatty acids (SCFAs), according to Nomura et al. [34]. In the study by Golčić et al. [24], a high fiber intake was determined by 25 g daily, whereas a low intake is 22 g. Tanaka et al. [22], however, noted no significant difference in survival outcomes between individuals consuming 8.8 g per day and those consuming 11 g per day of fiber in both short- and long-term follow-ups. Table 4. lists Food frequency questionnaires (FFQs) used in human studies.
Table 4.
Types of FFQs used in the included studies
| Study | Questionnaire type |
|---|---|
| Bolte et al. [19] | EPIC-Norfolk FFQ |
| Pietrzak et al. [36] | Unique questionnaire evaluating fat, meat, fermented food, fruit, grain, salt, alcohol consumption, antibiotic, proton pump inhibitor usage |
| Simpson et al. [21] | Cancer Council Victoria Dietary Questionnaire for Epidemiological Studies (DQES v3.2) and Food Frequency Questionnaire (FQ18N) developed by Wageningen University and Research |
| Spencer et al. [20] | National Cancer Institute Dietary Screener Questionnaire |
| Nomura et al. [34] | Unique questionnaire evaluating beef, pork, chicken, fish, beans, green vegetables, cabbage, potato, radish, pumpkin, mushroom, seaweed, fruit, and yogurt consumption |
| Golčić et al. [24] | Croatian, Danish and American food composition database Danish and the Phenol-Explorer 3.0 database were used |
| Ferrere et al. [27] | Mouse study |
| Boucher et al. [25] | Mouse study |
| Han et al. [28] | Mouse study |
| Zhang et al. [1] | Mouse study |
| Ji et al. [52] | Mouse study |
| Kim et al. [45] | Mouse study |
| Udumula et al. [2] | Mouse study |
| Tanaka et al. [22] | Brief-type self-administered diet history questionnaire and DHQ Support Center software |
| Cortellino et al. [63] | Mouse study |
| Calderón-Montaño et al. [53] | Mouse study |
| Kuehm et al. [64] | Mouse study |
| Lam et al. [65] | Mouse study |
| Li et al. [66] | Mouse study |
In mouse studies, various interventions were applied to reveal the response modifier effect of different dietary interventions. High fiber consumption was observed as inulin, pectin, and mucin supplementation which can have a similar effect as fruit and vegetable consumption in human patients [23]. High sugar intake was modeled as a high-glucose, fructose, and high sugar plus high fat-containing “western type” diet, which can be compared with ketogenic diet which is based on low carbohydrate consumption. The parallelized dietary interventions are shown in Fig. 3.
Fig. 3.
Associations of dietary interventions in mouse and human studies. Mouse studies highlight the underlying biology between specific dietary interventions and therapeutic responses to ICI. Fiber intake (marked with green) in human studies was presented in animal studies as inulin, mucin and pectin intake. Those mentioned molecules are prebiotics. Sugar consumption was categorized in human studies as carbohydrate intake, while in animal studies, the glucose, and fructose intake was observed as well as high sugar-, and fat-containing western type versus ketogenic diet
Microbial taxonomy, diversity, and nutritional aspects
The overall alterations in the microbial communities were observed in 7 alpha and beta diversity studies (Fig. 4). Three human studies [20, 21, 24] did not find significant differences between alpha diversity of responder and non-responder patients, however Pietrzak et al. [36] reported significantly decreased alpha diversity in responders (Fig. 4). Beta diversity was reported to be increased in Golčić et al. [24]. The study evaluated both Jaccard distance and Bray–Curtis dissimilarity between the groups. It was observed in the study by Pietrzak et al. [36], that the beta diversity is significantly different (p = 0.003) defined by permutational multivariate analysis of variance in responder patients compared to non-responders. In animal studies, alpha, and beta diversity were reported after fiber supplementation [25, 26]. Altered beta diversity was observed following high fiber and ketogenic diet [25–27].
Fig. 4.
Bacterial diversity changes in patients with better outcomes after specific dietary interventions in the identified prospective cohort-, and observational cross-sectional studies
The article by Pietrzak et al. [36], and Simpson et al. [21]. described Faecalibacterium prausnitzii as an indicator of response, and they also highlighted the importance of a fiber-rich diet. In the study of Simpson et al. [21] Akkermansia muciniphila reported to be enriched in responders, as well as in a preclinical experiments, where mice were treated with a ketogenic diet [27], and inulin [28].
Observing the metabolic potential of the identified microbes in the context of therapeutic response, four SCFA producers and four lactic acid producer bacteria were identified as favorable. SCFA producers identified in responder patients were the Faecalibacterium prausnitzii [29]. Bacteroides uniformis [30] and Butyricicoccus pullicaecorum [1] species in addition to Clostridium disporicum [31]. An elevated presence of lactic acid producers were also reported, where Ruthenibacterium lactatiformans [32] after ketogenic diet, Bifidobacterium animalis subsp. animalis after inulin supplementation [33], and Clostridium disporicum [34] after inulin treatment were identified apart from the decreased abundance of Turicibacter [35] after high sugar and fat-containing nourishment.
Comparison of the outcomes
The human studies used questionnaires to examine the nutritional differences according to response. In most of the human studies, PFS (n = 3) and ORR (n = 6) were assessed, the minority of studies (n = 2) observed the discrepancies between early and late responders. Besides response rate, n = 1 study evaluated the disease control rate (DCR).
In the studies by Bolte et al. [19] and Simpson et al. [21], a statistically significant distinction was observed between responders and non-responders (p = 0.02, False Discovery Rate [FDR]: 0.032) following high fiber consumption. Increased PFS was reported, with p = 0.03 and p = 0.01, respectively, linked to adequate (above median/mean) intake of fiber and mushrooms, as demonstrated by Spencer et al. [20] and Nomura et al. [34]. A substantial disparity was detected in the consumption of flavones (p = 0.027), and sugars (p = 0.04) in association with late versus early response, as reported in the study by Golčić et al. [24]. Likewise, a significant increase in responders was noted by Tanaka et al. [22] concerning increased seafood- (p = 0.045), and low sugar consumption (p = 0.031).
Bolte et al. [19] identified, that individuals with elevated fiber or unsaturated fat (characteristic of a Mediterranean dietary regimen) intake exhibited superior outcomes, with 46–49% attaining progression-free survival at the 12-month mark (PFS12) and 58–59% achieving objective response rates (ORR). Similarly, Pietrzak et al. [36] showed that individuals who responded favorably to ICI therapy consumed a higher fiber-containing diet, with reduced dairy intake, and elevated levels of SCFAs in their serum. Parallel to that, in the study by Nomura et al. [34], it was indicated, that patients with various cancers demonstrated higher fecal SCFA concentrations, associated with fiber intake, and also achieved a superior overall response rate (28.8%) and DCR (46.1%). After anti-PD-1 therapy, Simpson et al. [21] observed that melanoma patients exhibited favorable pathological responses, and had increased consumption of fiber and omega-3 fatty acids. In the study conducted by Spencer et al. [20] responder melanoma patients had elevated fiber intake, with 193 out of 293 patients (65.87%) achieving a favorable therapeutic outcome. Golčić et al. [24] showed, that individuals categorized as late responders to immunotherapy exhibited higher consumption of flavonoids, vitamin D, and anthocyanins, alongside reduced intake of sweets and saturated fats, thereby suggesting that particular nutrients may influence the timing and effectiveness of treatment. Tanaka et al. [22] revealed that long-term survivors (OS > 3 years) of NSCLC had increased consumption of seafood and eicosapentaenoic acid, which were positively correlated with increased OS.
Across the entirety of the human studies, fiber, unsaturated fats, SCFA production, and specific nutrients such as omega-3 fatty acids and flavonoids were consistently associated with improved responses to immunotherapy, thereby improving survival (Fig. 5), indicating that dietary modifications could potentially play a pivotal role in augmenting treatment outcomes. The study by Ferrere et al. [27] revealed that adherence to a ketogenic diet markedly inhibited tumor proliferation, resulting in a significant decrease in tumor size and an extension of overall survival in 60% of mice injected with melanoma. Parallel to this, Kuehm et al. [64] documented that mice subjected to a high-glucose, Western-style diet exhibited diminished efficacy in immunotherapy and larger tumor sizes compared to control cohorts.
Fig. 5.
Associations of dietary habits and ICI efficiency in human and mouse data. Increased outcome in the association of a certain dietary habit was marked with green color. Decreased outcome with the association of a certain dietary habit was marked with red color
It was reported by Zhang et al. [1], that the intake of pectin substantially diminished tumor volume within a colorectal cancer model, thereby further substantiating the association between dietary fiber and enhanced outcomes after immunotherapy. In a similar vein, Boucher et al. [25], and Han et al. [28] showed that the supplementation of inulin decelerated tumor growth and also stimulated anti-tumor immune responses, with an inverse correlation observed between tumor size and the prevalence of advantageous microbiota such as Akkermansia and Lactobacillus. Calderón-Montaño et al. [53] demonstrated that particular restrictions on amino acids, safeguarded non-malignant cells and resulted in a diminished tumor burden and enhanced survival rates in mice receiving anti-PD1 immunotherapy. On the contrary, study by Ji et al. [52] reported that the dietary restriction of methionine resulted in increased colon tumor volumes.
In contrast, fasting-associated interventions consistently yielded beneficial results. Udumula et al. [2] documented significantly reduced tumor volumes in mice subjected to intermittent fasting, while Cortellino et al. [63] established that a diet, mimicking fasting-induced tumor regression, was characterized by a notable reduction in tumor size relative to standard chow-consuming mice. Collectively, these studies suggest that various dietary modifications can exert a potential influence on tumor growth (Fig. 5), with specific diets such as ketogenic, fasting, and high-fiber exhibiting considerable potential in reducing tumor size and augmenting the efficacy of immunotherapy. Patients with better survival exhibited longer PFS, and OS compared to patients with decreased survival. The unanimous finding of the studies comparing responder and non-responder patients is that the high amount of fiber consumption in the form of fruits and vegetables, as well as fish increased survival [19–21, 36]. It was also observed by Nomura et al. [34], that higher fecal SCFA content is associated with longer PFS, which can be a consequence of high fiber consumption. Observing the differences between long-term and short-term survivors, there was no detected difference in protein, fat, carbohydrate, fiber, and fatty acid intake overall, however long term-survivors consumed less sugar according to Tanaka et al. [22] and Golčić et al. [24].
Quantitative synthesis of high-fiber diet impact on ICI response
We selected the four prospective human cohort studies for a partial meta-analysis, because they uniquely evaluated a clinically relevant binary outcome—response versus non-response to ICI—in relation to diet. Among the various dietary exposures assessed, the most comparable and clinically meaningful distinction across studies was the contrast between high versus low fiber intake. This dichotomy directly reflects dietary quality with known immunomodulatory potential via gut microbiota modulation. In addition to directly reported fiber intake [21, 36], two studies used established proxies for high fiber consumption: circulating short-chain fatty acid (SCFA) levels [34] and adherence to a Mediterranean diet [19], both of which reflect increased fiber intake and microbial fermentation. All included studies reported response outcomes in a form suitable for pooled analysis, enabling estimation of the effect size of fiber-rich versus fiber-poor diets across diverse populations and cancer types. The central research question addressed is whether high fiber intake—or its validated dietary or microbial surrogates—is consistently associated with improved ICI efficacy. This is clinically relevant as dietary modification is a feasible, low-cost, and scalable intervention that may improve immunotherapy outcomes and guide future interventional strategies.
A random-effects meta-analysis was conducted using four prospective human studies that compared the likelihood of response to ICI between patients with fiber-rich or Mediterranean-like diets and those with fiber-poor diets (Fig. 6A). The most diet-extreme groups were selected from each study, and 2 × 2 contingency tables were constructed to calculate odds ratios (ORs) for responder versus non-responder outcomes. Data from Bolte et al. were simulated based on response probabilities due to lack of raw counts. The pooled analysis yielded a summary OR of 5.79 (95% CI: 2.45–13.68), indicating a significantly higher likelihood of ICI response among individuals adhering to fiber-rich diets. Study contributions to the summary effect were highest for Pietrzak et al. (32.5%) and Simpson et al. (28.4%), while Bolte et al., based on estimated data, contributed only 18.2% (Fig. 6B). Despite heterogeneity in dietary metrics and populations, all studies demonstrated directionally consistent results favoring dietary fiber as a supportive factor for ICI efficacy.
Fig. 6.
Meta-analysis of outcomes from prospective cohort studies. Forest plot showing odds ratios (log scale) for response to ICIs in patients with high- vs. low-fiber diets across four prospective studies. ORR = objective response rate; path. response = pathological response. B Relative study weights in the pooled random-effects meta-analysis based on inverse-variance estimation. Simulated odds from Bolte et al. [19] were included with caution due to estimated input values, contributing 17.5% to the overall summary effect
Discussion
Recently, anti-cancer ICI therapy outcomes have been linked to microbiota [37, 38]. Nutritional habits might influence the gut microbiome, which holds the potential to alter taxa abundance, diversity, and functionality. The advantageous impacts of consuming low sugar or ketogenic diet along with high dietary fiber arise from its physical, immunomodulatory, and prebiotic characteristics [39, 40]. These benefits are commonly linked to the increased production of SCFAs, which play a pivotal role in the modulation of immunological tolerance through both lymphocytes and myeloid cells and by decreasing tumor necrosis factor (TNF)-α, interferon (IFN)-γ, interleukin (IL)-17,10,6 and IL13 production. Short-chain fatty acids (SCFAs) modulate immune tolerance by influencing lymphocyte and myeloid cell function and reducing pro-inflammatory cytokines such as TNF-α, IFN-γ, IL-17, IL-10, IL-6, and IL-13 [41]. SCFAs impact chemotaxis, phagocytosis, ROS production, cellular proliferation, and have anti-inflammatory, antitumorigenic, and antimicrobial properties. They enhance epithelial integrity [46], activate the NLRP3 inflammasome to induce IL-18 secretion, promote neutrophil recruitment, and support FOXP3+ regulatory T cell differentiation. Butyrate acts via G protein-coupled receptor activation and histone deacetylase inhibition, increasing tight junction and antimicrobial peptide expression [47]. SCFA producers function as significant agents in the conservation of gastrointestinal homeostasis through the enhancement of epithelial integrity and the improvement of mucosal immunity, whereas current investigations suggest that they are also indispensable for the regulation of systemic immunity [41].
In the present systematic review, we aimed to summarize the current evidence on clinical and preclinical data on nutritional habits, gut microbiota taxonomy, diversity, and ICI treatment outcomes. The majority of studies showed that fiber-rich diet associated with favorable outcomes in ICI-treated patients [42, 43, 45]. The observed human studies [19–22, 36] in agreement, also highlighted the importance and beneficial effects of fruit and vegetable consumption, Nomura et al. [34] also emphasizing the significant beneficial effect of mushroom intake in responders. However, Russo et al. [43] reported increased bacterial diversity parallel to fiber consumption, Pietrzak et al. [36] observed decreased alpha-, and altered beta-diversity in responders in the case of elevanted fiber consumption. Analyzing the results of precilincal mouse studies, Ferrere et al. [27], Boucher et al. [25] and Zhang et al. [1] reported altered beta-diversity after ketogenic, and fiber-enriched diet. Following high fiber-containing diet, Zhang et al. [1] and Boucher et al. [25] reported increased and altered alpha-diversity. The identified results underpin the hypothesis of the correlation between high-fiber diet and beneficial effect on ICI outcome. Furthermore, the quantitative synthesis suggested that high fiber intake—or its validated dietary and microbial proxies—was consistently associated with improved response to ICIs. The pooled effect estimate (OR = 5.79) and directionally aligned results across studies shows the clinical relevance of diet as a modifiable factor in immunotherapy outcomes. Despite some heterogeneity and one study relying on estimated data, the findings support dietary fiber as a low-cost, scalable strategy to enhance ICI efficacy and justify prospective interventional trials.
Cancer alters the body’s utilization of amino acids. Tumors rely on amino acids as a fuel source and source of antioxidants to regulate the generation of harmful reactive oxygen species. Amino acids play a significant role in modifying the epigenetic information to either upregulate or downregulate the activity of genes associated with tumors [46]. Dietary addition or restriction of certain amino acids were examined by several studies [47–51]. In the included studies, the impact of restricting dietary methionine [52] and the influence of changing the amino acid composition of the diet [53] were also investigated. The immunostimulatory effect of methionine in immunocompetent mice was evidenced by the production of H2S by microbes according to Ji et al. [52] and Calderón-Montaño et al. [53]. The Authors reported, that methionine restriction alone is not sufficient to enhance anti-cancer activity, the composition of other amino acids and nutrients are also important. Altogether, amino acid composition of diet has a significant effect on anti-cancer immunity, in which processes intestinal microbiome has a crucial role.
Epidemiological data has continuously established a correlation between sugar consumption with elevated occurrences of obesity, metabolic syndrome, and diabetes, all of which represent predisposing factors for cancer [44]. In a colorectal cancer model, high sugar diet exhibited a promotion of tumor growth, despite the absence of metabolic alterations like diabetes [45]. In the study by Golčić et al. [24], it was observed, that patients who exhibited complete remission consumed smaller amounts of sweets. The detrimental effect of elevated sugar intake was confirmed by Kuehm et al. [64] by observing that Western-diet impaired the response to ICI therapy. Microbial differences between Western-diet and normal groups were also identified. These results are also supported by Ferrere et al. [27], namely that the ketogenic diet led to alterations in the composition of the gut microbiota, in both mice and humans following carbohydrate-restricted dietary interventions resulting in enhanced anti-neoplastic effect of T cells in the tumor microenvironment. In contrast, Kim et al. [45] observed that short-term high glucose supplementation enhanced anti-tumor immunity in a glioblastoma multiforme (GBM) mouse model. There is a difference in the tumor microenvironment between immunologically “hot” tumors, like melanoma, colon cancer, NSCLC, and “cold” tumors like GBM, which can have an impact of the different behavior of the mentioned tumor after glucose supplementation [45].
Modulating the gut microbiota is a promising strategy to enhance the efficacy of immune checkpoint inhibitors (ICIs), with three primary approaches under investigation: fecal microbiota transplantation (FMT), probiotics, and dietary interventions. While probiotics have shown potential in preclinical and observational studies, clinical trials have yielded inconclusive results, and non-personalized use may reduce microbial diversity, potentially impairing ICI outcomes [9, 12]. FMT, by contrast, has demonstrated clinical benefit in refractory cancers by increasing microbial diversity and enriching beneficial taxa associated with improved immunotherapeutic responses [54] However, due to its invasiveness, dietary interventions offer a more practical, non-invasive alternative. Diets rich in fiber and low in sugar may mimic the microbiota-modulating effects of FMT and enhance ICI efficacy [9, 57]. Reflecting this potential, several clinical trials (NCT04645680, NCT05356182, NCT06236360, NCT04866810, NCT06671613) are evaluating dietary strategies, particularly in non-small cell lung cancer and melanoma. Although most remain ongoing, the increasing focus on dietary modulation underscores its appeal due to its safety, cost-effectiveness, and accessibility for improving immune responses in oncology patients.
Another conclusion from studies in the field is that fasting and ketogenic diets may offer immunotherapeutic benefits, their application in cancer patients—especially those with cachexia or treatment-related side effects—requires caution [39, 55]. Cachexia, characterized by severe weight and muscle loss, may be worsened by restrictive diets. Arends and colleagues emphasize the importance of nutritional support and advise against further nutrient restriction in malnourished patients [56]. Adherence to intermittent fasting or ketogenic diets is also challenged by appetite loss and treatment side effects. Klement and colleagues highlight the potential of ketogenic diets in oncology but note adherence difficulties due to their restrictive nature [57]. These findings are also in line with recent perspectives on the interplay between diet, the microbiome, and immune modulation in cancer [58, 59]. Future studies should evaluate the long-term safety, metabolic effects, and feasibility of such diets in cancer patients and explore personalized dietary strategies balancing efficacy with nutritional adequacy.
Diverse food FFQs introduce systematic measurement bias, often leading to over- or underestimation of dietary intake and reduced cross-study comparability. Self-reported data are prone to response and social desirability bias, increasing misclassification and compromising dietary exposure precision [60, 61]. Errors also arise from inaccurate portion size estimation and recall difficulties. Differences in FFQ design and validation across populations further limit accuracy when applied to diverse cohorts. These limitations challenge FFQs’ ability to detect diet–disease associations, raising concerns about their reliability in epidemiological research [62].
The included studies had varying primary scopes: human studies predominantly examined fecal microbiome differences between ICI responders and non-responders or assessed serum fatty acid content in blood plasma. Dietary information obtained via food frequency questionnaires was secondary scope to these main focuses. In addition, differences in FFQs introduce measurement errors, response bias, portion size variability, and population-specific inaccuracies, potentially limiting their reliability in detecting diet-disease associations, and cross-study comparison. Additionally, outcomes were often evaluated and reported as categorical rather than continuous variables, limiting data comparison or detailed analysis. Mouse studies varied in dietary interventions; the caregivers and/or investigators were not blinded, nor were animals selected at random for outcome assessment, making challenging comparisons across studies. More research is needed, specifically on how distinct dietary interventions influence immune checkpoint inhibitor (ICI) efficacy.
Conclusions
Intermittent fasting, high fiber, and low sugar consumption are significantly associated with better ICI outcomes. Clinical prospective cohort studies revealed alterations in microbiota composition linked to diet. The alterations in microbiota composition linked to diet appear to be confirmed in animal models, particularly regarding the production of SCFAs and lactic acid, as well as an increase in Firmicutes/Bacteroidetes ratio modifications in the overall microbial diversity. Further prospective studies are needed to accelerate microbiota therapeutics in the field.
Supplementary Information
Abbreviations
- CHT
Chemotherapy
- CRT
Chemoradiotherapy
- IO
Immuno-oncology
- IT
Immunotherapy
- ICI
Immune checkpoint inhibitors
- SCFA
Short-chain fatty acid
- F/B ratio
Firmicutes/ Bacteroidetes ratio
- NA
Not assessed
- PFS
Progression-free survival
- ORR
Overall response rate
- PFS12
PFS at 12 months
- RECIST
Response evaluation criteria
- NSCLC
Non-small cell lung cancer
- GMB
Glioblastoma multiforme
- TNF-α
Tumor necrosis factor
- IFN-γ
Interferon gamma
- DCR
Disease control rate
Author contributions
CS: Conceptualization, Data curation, Investigation, Methodology, Visualization, Writing—original draft, Writing—review & editing. DD: Data curation, Formal analysis, Methodology, Software, Supervision, Visualization, Writing—original draft, Writing—review & editing. MH: Formal analysis, Methodology, Project administration, Validation, Visualization, Writing—original draft, Writing—review & editing. SG: Methodology, Visualization, Writing—original draft, Writing—review & editing. ZL: Conceptualization, Formal analysis, Funding acquisition, Investigation, Project administration, Resources, Supervision, Validation, Writing—original draft, Writing—review & editing.
Funding
Open access funding provided by Semmelweis University. ZL acknowledges funding from the Hungarian National Research, Development, and Innovation Office (OTKA-FK, #146775). DD acknowledges funding from the Hungarian National Research, Development and Innovation Office (OTKA-PD, #142287) DD was also supported by the Bolyai Research Scholarship of the Hungarian Academy of Sciences.
Data availability
This was a systematic review, where no data were generated or deposited by the authors. All data is accessible in the included articles.
Declarations
Competing interests
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- 1.Zhang Y, Zhang Z. The history and advances in cancer immunotherapy: understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell Mol Immunol. 2020;17(8):807–21. 10.1038/s41423-020-0488-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Udumula MP, Singh H, Rashid F, Poisson L, Tiwari N, Dimitrova I, Hijaz M, Gogoi R, Swenor M, Munkarah A, Giri S, Rattan R. Intermittent fasting induced ketogenesis inhibits mouse epithelial ovarian cancer by promoting antitumor T cell response. iScience. 2023;26(10):107839. 10.1016/j.isci.2023.107839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Zheng D, Liwinski T, Elinav E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020;30(6):492–506. 10.1038/s41422-020-0332-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Matson V, Chervin CS, Gajewski TF. Cancer and the microbiome—influence of the commensal microbiota on cancer, immune responses, and immunotherapy. Gastroenterology. 2020;160(2):600–13. 10.1053/j.gastro.2020.11.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kim J, Lee HK. The role of gut microbiota in modulating tumor growth and anticancer agent efficacy. Molecules Cells. 2021; 44(5): 356–62. 10.14348/molcells.2021.0032. [DOI] [PMC free article] [PubMed]
- 6.Dora D, Weiss GJ, Megyesfalvi Z, Gállfy G, Dulka E, Kerpel-Fronius A, Berta J, Moldvay J, Dome B, Lohinai Z. Computed tomography-based quantitative texture analysis and gut microbial community signatures predict survival in non-small cell lung cancer. Cancers. 2023;15(20):5091. 10.3390/cancers15205091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Dora D, Szőcs E, Soós Á, Halasy V, Somodi C, Mihucz A, Rostás M, Mógor F, Lohinai Z, Nagy N. From bench to bedside: an interdisciplinary journey through the gut-lung axis with insights into lung cancer and immunotherapy. Front Immunol. 2024;15:1434804. 10.3389/fimmu.2024.1434804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Dora D, Ligeti B, Kovacs T, Revisnyei P, Galffy G, Dulka E, Krizsán D, Kalcsevszki R, Megyesfalvi Z, Dome B, Weiss GJ, Lohinai Z. Non-small cell lung cancer patients treated with anti-PD1 immunotherapy show distinct microbial signatures and metabolic pathways according to progression-free survival and PD-L1 status. Oncoimmunology. 2023;12(1):2204746. 10.1080/2162402X.2023.2204746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, Fluckiger A, Messaoudene M, Rauber C, Roberti MP, Fidelle M, Flament C, Poirier-Colame V, Opolon P, Klein C, Iribarren K, Mondragón L, Jacquelot N, Qu B, Ferrere G, Clémenson C, Mezquita L, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–7. 10.1126/science.aan3706. [DOI] [PubMed] [Google Scholar]
- 10.Fluckiger A, Daillère R, Sassi M, Sixt BS, Liu P, Loos F, Richard C, Rabu C, Alou MT, Goubet AG, Lemaitre F, Ferrere G, Derosa L, Duong CPM, Messaoudene M, Gagné A, Joubert P, De Sordi L, Debarbieux L, Simon S, Scarlata CM, Ayyoub M, et al. Cross-reactivity between tumor MHC class I-restricted antigens and an enterococcal bacteriophage. Science. 2020;369(6506):936–42. 10.1126/science.aax0701. [DOI] [PubMed] [Google Scholar]
- 11.Zitvogel L, Ma Y, Raoult D, Kroemer G, Gajewski TF. The microbiome in cancer immunotherapy: diagnostic tools and therapeutic strategies. Science. 2018;359(6382):1366–70. 10.1126/science.aar6918. [DOI] [PubMed] [Google Scholar]
- 12.Gopalakrishnan V, Helmink BA, Spencer CN, Reuben A, Wargo JA. The influence of the gut microbiome on cancer, immunity, and cancer immunotherapy. Cancer Cell. 2018;33(4):570–80. 10.1016/j.ccell.2018.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Dora D, Kiraly P, Somodi C, Ligeti B, Dulka E, Galffy G, Lohinai Z. Gut metatranscriptomics based de novo assembly reveals microbial signatures predicting immunotherapy outcomes in non-small cell lung cancer. J Transl Med. 2024;22:1044. 10.1186/s12967-024-05835-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Lu Y, Yuan X, Wang M, He Z, Li H, Wang J, Li Q. Gut microbiota influence immunotherapy responses: mechanisms and therapeutic strategies. J Hematol Oncol. 2022;15(1):47. 10.1186/s13045-022-01273-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Chilakapati SR, Ricciuti J, Zsiros E. Microbiome and cancer immunotherapy. Curr Opin Biotechnol. 2020;65:114–7. 10.1016/j.copbio.2020.02.007. [DOI] [PubMed] [Google Scholar]
- 16.Kian N, Behrouzieh S, Razi S, Rezaei N. Diet influences immunotherapy outcomes in cancer patients: a literature review. Nutr Cancer. 2023;75(2):415–29. 10.1080/01635581.2022.2133151. [DOI] [PubMed] [Google Scholar]
- 17.Page MJ, Moher D, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, Shamseer L, Tetzlaff JM, Akl EA, Brennan SE, Chou R, Glanville J, Grimshaw JM, Hróbjartsson A, Lalu MM, Li T, Loder EW, Mayo-Wilson E, McDonald S, McGuinness LA, Stewart LA, Thomas J, et al. PRISMA 2020 explanation and elaboration: updated guidance and exemplars for reporting systematic reviews. BMJ. 2021;372:n160. 10.1136/bmj.n160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ouzzani M, Hammady H, Fedorowicz Z, Elmagarmid A. Rayyan-a web and mobile app for systematic reviews. Syst Rev. 2016;5(1):210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bolte LA, Lee KA, Björk JR, Leeming ER, Campmans-Kuijpers MJ, de Haan JJ, Vila AV, Maltez-Thomas A, Segata N, Board R, Harries M, Lorigan P, de Vries EG, Nathan P, Fehrmann R, Bataille V, Spector TD, Hospers GA, Weersma RK. Association of a Mediterranean diet with outcomes for patients treated with immune checkpoint blockade for advanced melanoma. JAMA Oncol. 2023;9(5):705–9. 10.1001/jamaoncol.2022.7753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Spencer CN, McQuade JL, Gopalakrishnan V, McCulloch JA, Vetizou M, Cogdill AP, Khan MAW, Zhang X, White MG, Peterson CB, Wong MC, Morad G, Rodgers T, Badger JH, Helmink BA, Andrews MC, Rodrigues RR, Morgun A, Kim YS, Roszik J, Hoffman KL, Zheng J, et al. Dietary fiber and probiotics influence the gut microbiome and melanoma immunotherapy response. Science. 2021;374(6575):1632–40. 10.1126/science.aaz7015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Simpson RC, Shanahan ER, Batten M, Reijers ILM, Read M, Silva IP, Versluis JM, Ribeiro R, Angelatos AS, Tan J, Adhikari C, Menzies AM, Saw RPM, Gonzalez M, Shannon KF, Spillane AJ, Velickovic R, Lazar AJ, Damania AV, Mishra AK, Chelvanambi M, Banerjee A, et al. Diet-driven microbial ecology underpins associations between cancer immunotherapy outcomes and the gut microbiome. Nat Med. 2022;28(11):2344–52. 10.1038/s41591-022-01965-2. [DOI] [PubMed] [Google Scholar]
- 22.Tanaka I, Yano Y, Mori M, Manabe S, Fukuo K. Impact of serum eicosapentaenoic acid/arachidonic acid ratio on overall survival in lung cancer patients treated with pembrolizumab: a pilot study. Sci Rep. 2024;14:1384. 10.1038/s41598-024-51967-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Desai MS, Seekatz AM, Koropatkin NM, Kamada N, Hickey CA, Wolter M, Pudlo NA, Kitamoto S, Terrapon N, Muller A, Young VB, Henrissat B, Wilmes P, Stappenbeck TS, Núñez G, Martens EC. A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell. 2016;167(5):1339-1353.e21. 10.1016/j.cell.2016.10.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Golčić M, Simetić L, Herceg D, Blažičević K, Kenđel Jovanović G, Dražić I, Belančić A, Skočibušić N, Palčevski D, Rubinić I, Vlahović-Palčevski V, Majnarić T, Dobrila-Dintinjana R, Pleština S. Analysis of the gut microbiome and dietary habits in metastatic melanoma patients with a complete and sustained response to immunotherapy. Cancers. 2023;15(11):3052. 10.3390/cancers15113052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Boucher E, Plazy C, Richard ML, Suau A, Mangin I, Cornet M, Aldebert D, Toussaint B, Hannani D. Inulin prebiotic reinforces host cancer immunosurveillance via ɣδ T cell activation. Front Immunol. 2023;14:1104224. 10.3389/fimmu.2023.1104224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Zhang SL, Mao YQ, Zhang ZY, Li ZM, Kong CY, Chen HL, Cai PR, Han B, Ye T, Wang LS. Pectin supplement significantly enhanced the anti-PD-1 efficacy in tumor-bearing mice humanized with gut microbiota from patients with colorectal cancer. Theranostics. 2021;11(9):4155–70. 10.7150/thno.54476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Ferrere G, Tidjani Alou M, Liu P, Goubet AG, Fidelle M, Kepp O, Durand S, Iebba V, Fluckiger A, Daillère R, Thelemaque C, Grajeda-Iglesias C, Alves Costa Silva C, Aprahamian F, Lefevre D, Zhao L, Ryffel B, Colomba E, Arnedos M, Drubay D, Rauber C, Raoult D, Asnicar F, Spector T, Segata N, Derosa L, Kroemer G, Zitvogel L. Ketogenic diet and ketone bodies enhance the anticancer effects of PD-1 blockade. JCI Insight. 2021;6(2): e145207. 10.1172/jci.insight.145207. [DOI] [PMC free article] [PubMed]
- 28.Han K, Nam J, Xu J, Sun X, Huang X, Animasahun O, Achreja A, Jeon JH, Pursley B, Kamada N, Chen GY, Nagrath D, Moon JJ. Generation of systemic antitumour immunity via the in situ modulation of the gut microbiome by an orally administered inulin gel. Nat Biomed Eng. 2021;5(11):1377–88. 10.1038/s41551-021-00749-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Parsaei M, Sarafraz N, Moaddab SY, Ebrahimzadeh LH. The importance of Faecalibacterium prausnitzii in human health and diseases. New Microbes New Infections. 2021;43:100928. 10.1016/j.nmni.2021.100928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Dai W, Zhang J, Chen L, Yu J, Zhang J, Yin H, Shang Q, Yu G. Discovery of Bacteroides uniformis F18–22 as a safe and novel probiotic bacterium for the treatment of ulcerative colitis from the healthy human colon. Int J Mol Sci. 2023;24(19):14669. 10.3390/ijms241914669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Horn N. Clostridium disporicum sp. nov., a saccharolytic species able to form two spores per cell, isolated from a rat cecum. Int J Syst Evol Microbiol. 1987;37(4):398–402. 10.1099/00207713-37-4-398. [Google Scholar]
- 32.Shkoporov AN, Chaplin AV, Shcherbakova VA, Suzina NE, Kafarskaia LI, Bozhenko VK, Efimov BA. Ruthenibacterium lactatiformans gen. nov., sp. nov., an anaerobic, lactate-producing member of the family Ruminococcaceae isolated from human faeces. Int J Syst Evol Microbiol. 2016;66(8):3041–9. 10.1099/ijsem.0.001143. [DOI] [PubMed] [Google Scholar]
- 33.Bunesova V, Killer J, Javurkova B, Vlkova E, Tejnecky V, Musilova S, Rada V. Diversity of the subspecies Bifidobacterium animalis subsp. lactis. Anaerobe. 2017;44:40–7. 10.1016/j.anaerobe.2017.01.006. [DOI] [PubMed] [Google Scholar]
- 34.Nomura M, Nagatomo R, Doi K, Shimizu J, Baba K, Saito T, Matsumoto S, Inoue K, Muto M. Association of short-chain fatty acids in the gut microbiome with clinical response to treatment with nivolumab or pembrolizumab in patients with solid cancer tumors. JAMA Netw Open. 2020;3(4):e202895. 10.1001/jamanetworkopen.2020.2895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Bosshard PP, Zbinden R, Altwegg M. Turicibacter sanguinis gen nov., sp. nov., a novel anaerobic, Gram-positive bacterium. Int J Syst Evol Microbiol. 2002;52(Pt 4):1263–6. 10.1099/00207713-52-4-1263. [DOI] [PubMed] [Google Scholar]
- 36.Pietrzak B, Tomela K, Olejnik-Schmidt A, Galus Ł, Mackiewicz J, Kaczmarek M, Mackiewicz A, Schmidt M. A clinical outcome of the anti-PD-1 therapy of melanoma in polish patients is mediated by population-specific gut microbiome composition. Cancers. 2022;14(21):5369. 10.3390/cancers14215369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Szczyrek M, Bitkowska P, Chunowski P, Czuchryta P, Krawczyk P, Milanowski J. Diet, microbiome, and cancer immunotherapy—a comprehensive review. Nutrients. 2021;13(7):2217. 10.3390/nu13072217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Zitvogel L, Kroemer G. Boosting the immunotherapy response by nutritional interventions. J Clin Invest. 2022;132(11). 10.1172/JCI161483. [DOI] [PMC free article] [PubMed]
- 39.Golonko A, Pienkowski T, Swislocka R, Orzechowska S, Marszalek K, Szczerbinski L, Swiergiel AH, Lewandowski W. Dietary factors and their influence on immunotherapy strategies in oncology: a comprehensive review. Cell Death Dis. 2024;15(4):254. 10.1038/s41419-024-06641-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Slavin J. Fiber and prebiotics: mechanisms and health benefits. Nutrients. 2013;5(4):1417–35. 10.3390/nu5041417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Porbahaie M, Hummel A, Saouadogo H, Coelho RML, Savelkoul HFJ, Teodorowicz M, van Neerven RJJ. Short-chain fatty acids inhibit the activation of T lymphocytes and myeloid cells and induce innate immune tolerance. Benef Microbes. 2023;14(4):401–19. 10.1163/18762891-20220113. [DOI] [PubMed] [Google Scholar]
- 42.Malczewski AB, Ketheesan N, Coward JIG, Navarro S. Front Immunol. 2021;12:624434. 10.3389/fimmu.2021.624434. [DOI] [PMC free article] [PubMed]
- 43.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(33):4919–32. 10.3748/wjg.v26.i33.4919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Makarem N, Bandera EV, Nicholson JM, Parekh N. Consumption of sugars, sugary foods, and sugary beverages in relation to cancer risk: a systematic review of longitudinal studies. Annu Rev Nutr. 2018;38:17–39. 10.1146/annur. [DOI] [PubMed] [Google Scholar]
- 45.Kim J, Kim Y, La J, Park WH, Kim HJ, Park SH, Ku KB, Kang BH, Lim J, Kwon MS, Lee HK. Supplementation with a high-glucose drink stimulates anti-tumor immune responses to glioblastoma via gut microbiota modulation. Cell Rep. 2023;42(10):113220. 10.1016/j.celrep.2023.113220. [DOI] [PubMed] [Google Scholar]
- 46.Lieu EL, Nguyen T, Rhyne S, Kim J. Amino acids in cancer. Exp Mol Med. 2020;52(1):15–30. 10.1038/s12276-020-0375-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Maddocks ODK, Athineos D, Cheung EC, Lee P, Zhang T, van den Broek NJF, Mackay GM, Labuschagne CF, Gay D, Kruiswijk F, Blagih J, Vincent DF, Campbell KJ, Ceteci F, Sansom OJ, Blyth K, Vousden KH. Modulating the therapeutic response of tumours to dietary serine and glycine starvation. Nature. 2017;544(7650):372–76. 10.1038/nature22056. Erratum in: Nature. 2017;548(7665):122. 10.1038/nature23471.
- 48.Sullivan MR, Mattaini KR, Dennstedt EA, Nguyen AA, Reilly MF, Meeth K, Muir A, Darnell AM, Bosenberg MW, Lewis CA, Vander Heiden MG. Increased serine synthesis provides an advantage for tumors arising in tissues where serine levels are limiting. Cell Metab. 2019;29(6):1410-1421.e4. 10.1016/j.cmet.2019.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Katada C, Fukazawa S, Sugawara M, Sakamoto Y, Takahashi K, Takahashi A, Watanabe A, Wada T, Ishido K, Furue Y, Harada H, Hosoda K, Yamashita K, Hiki N, Sato T, Ichikawa T, Shichiri M, Tanabe S, Koizumi W. Randomized study of prevention of gastrointestinal toxicities by nutritional support using an amino acid-rich elemental diet during chemotherapy in patients with esophageal cancer (KDOG 1101). Esophagus. 2021;18(2):296–305. 10.1007/s10388-020-00787-w. [DOI] [PubMed] [Google Scholar]
- 50.Ishikawa T, Yasuda T, Doi T, Okayama T, Sakamoto N, Gen Y, Dohi O, Yoshida N, Kamada K, Uchiyama K, Handa O, Takagi T, Konishi H, Yagi N, Kokura S, Naito Y, Itoh Y. The amino acid-rich elemental diet Elental® preserves lean body mass during chemo- or chemoradiotherapy for esophageal cancer. Oncol Rep. 2016;36(2):1093–100. 10.3892/or.2016.4877. [DOI] [PubMed]
- 51.Peña Vivas JDC, Orduz Arena AC, Alonso García A, Carrascal Gordillo CF, Martínez Gutiérrez R, Rodríguez-Acosta Caballero C, Fernández Freije I, Paino Martínez AB, Belloso Cuesta T, Juan Rijo G, Calleja FA. Clinical, functional, and nutritional efficacy of a glutamine-enriched oligomeric diet in patients with rectal cancer. Nutr Cancer. 2024;76(1):128–36. 10.1080/01635581.2023.2286698. [DOI] [PubMed] [Google Scholar]
- 52.Ji M, Xu X, Xu Q, Hsiao YC, Martin C, Ukraintseva S, Popov V, Arbeev KG, Randall TA, Wu X, Garcia-Peterson LM, Liu J, Xu X, Andrea Azcarate-Peril M, Wan Y, Yashin AI, Anantharaman K, Lu K, Li JL, Shats I, Li X. Methionine restriction-induced sulfur deficiency impairs antitumour immunity partially through gut microbiota. Nat Metab. 2023;5(9):1526–43. 10.1038/s42255-023-00854-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Calderón-Montaño JM, Guillén-Mancina E, Jiménez-Alonso JJ, Jiménez-González V, Burgos-Morón E, Mate A, Pérez-Guerrero MC, López-Lázaro M. Manipulation of amino acid levels with artificial diets induces a marked anticancer activity in mice with renal cell carcinoma. Int J Mol Sci. 2022;23(24):16132. 10.3390/ijms232416132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kim Y, Kim G, Kim S, Cho B, Kim SY, Do EJ, Bae DJ, Kim S, Kweon MN, Song JS, Park SH, Hwang SW, Kim MN, Kim Y, Min K, Kim SH, Adams MD, Lee C, Park H, Park SR. Fecal microbiota transplantation improves anti-PD-1 inhibitor efficacy in unresectable or metastatic solid cancers refractory to anti-PD-1 inhibitor. Cell Host Microbe. 2024;32(8):1380-1393.e9. 10.1016/j.chom.2024.06.010. [DOI] [PubMed] [Google Scholar]
- 55.Ferrer M, Mourikis N, Davidson EE, Kleeman SO, Zaccaria M, Habel J, Rubino R, Gao Q, Flint TR, Young L, Connell CM, Lukey MJ, Goncalves MD, White EP, Venkitaraman AR, Janowitz T. Ketogenic diet promotes tumor ferroptosis but induces relative corticosterone deficiency that accelerates cachexia. Cell Metab. 2023;35(7):1147-1162.e7. 10.1016/j.cmet.2023.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Arends J, Baracos V, Bertz H, Bozzetti F, Calder PC, Deutz NEP, Erickson N, Laviano A, Lisanti MP, Lobo DN, McMillan DC, Muscaritoli M, Ockenga J, Pirlich M, Strasser F, de van der Schueren M, Van Gossum A, Vaupel P, Weimann A. ESPEN expert group recommendations for action against cancer-related malnutrition. Clin Nutr. 2017;36(5):1187–96. 10.1016/j.clnu.2017.06.017. [DOI] [PubMed]
- 57.Klement RJ, Brehm N, Sweeney RA. Ketogenic diets in medical oncology: a systematic review with focus on clinical outcomes. Med Oncol. 2020;37(2):14. 10.1007/s12032-020-1337-2. [DOI] [PubMed] [Google Scholar]
- 58.Ağagündüz D, Cocozza E, Cemali Ö, Bayazıt AD, Nanì MF, Cerqua I, Morgillo F, Saygılı SK, Berni Canani R, Amero P, Capasso R. Understanding the role of the gut microbiome in gastrointestinal cancer: a review. Front Pharmacol. 2023;14:1130562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Şahin TÖ, Yılmaz B, Yeşilyurt N, Cicia D, Szymanowska A, Amero P, Ağagündüz D, Capasso R. Recent insights into the nutritional immunomodulation of cancer-related microRNAs. Phytother Res. 2023;37(10):4375–97. [DOI] [PubMed] [Google Scholar]
- 60.Popoola AA, Frediani JK, Hartman TJ, Paynabar K. Mitigating underreported error in food frequency questionnaire data using a supervised machine learning method and error adjustment algorithm. BMC Med Inform Decis Mak. 2023;23(1):178. 10.1186/s12911-023-02262-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Cheng Z, Shuai P, Qiao Q, Li T. Validity and reliability of a simplified food frequency questionnaire: a cross sectional study among physical health examination adults in southwest region of China. Nutr J. 2020;19(1):114. 10.1186/s12937-020-00630-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Kipnis V, Subar AF, Midthune D, Freedman LS, Ballard-Barbash R, Troiano RP, Bingham S, Schoeller DA, Schatzkin A, Carroll RJ. Structure of dietary measurement error: results of the OPEN biomarker study. Am J Epidemiol. 2003;158(1):14–21; discussion 22–6. 10.1093/aje/kwg091 [DOI] [PubMed]
- 63.Cortellino S, Quagliariello V, Delfanti G, Blaževitš O, Chiodoni C, Maurea N, Di Mauro A, Tatangelo F, Pisati F, Shmahala A, Lazzeri S, Spagnolo V, Visco E, Tripodo C, Casorati G, Dellabona P, Longo VD. Fasting mimicking diet in mice delays cancer growth and reduces immunotherapy-associated cardiovascular and systemic side effects. Nat Commun. 2023;14(1):5529. 10.1038/s41467-023-41066-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Kuehm LM, Khojandi N, Piening A, Klevorn LE, Geraud SC, McLaughlin NR, Griffett K, Burris TP, Pyles KD, Nelson AM, Preuss ML, Bockerstett KA, Donlin MJ, McCommis KS, DiPaolo RJ, Teague RM. Fructose promotes cytoprotection in melanoma tumors and resistance to immunotherapy. Cancer Immunol Res. 2021;9(2):227–38. 10.1158/2326-6066.CIR-20-0396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Lam KC, Araya RE, Huang A, Chen Q, Di Modica M, Rodrigues RR, Lopès A, Johnson SB, Schwarz B, Bohrnsen E, Cogdill AP, Bosio CM, Wargo JA, Lee MP, Goldszmid RS. Microbiota triggers STING-type I IFN-dependent monocyte reprogramming of the tumor microenvironment. Cell. 2021;184(21):5338–56.e21. 10.1016/j.cell.2021.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Li Y, Elmén L, Segota I, Xian Y, Tinoco R, Feng Y, Fujita Y, Segura Muñoz RR, Schmaltz R, Bradley LM, Ramer-Tait A, Zarecki R, Long T, Peterson SN, Ronai ZA. Prebiotic-induced anti-tumor immunity attenuates tumor growth. Cell Rep. 2020;30(6):1753–66.e6. 10.1016/j.celrep.2020.01.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
This was a systematic review, where no data were generated or deposited by the authors. All data is accessible in the included articles.






