Effects of Dietary Intervention On Human Diseases
Effects of Dietary Intervention On Human Diseases
Effects of Dietary Intervention On Human Diseases
com/sigtrans
Diet, serving as a vital source of nutrients, exerts a profound influence on human health and disease progression. Recently, dietary
interventions have emerged as promising adjunctive treatment strategies not only for cancer but also for neurodegenerative
diseases, autoimmune diseases, cardiovascular diseases, and metabolic disorders. These interventions have demonstrated
substantial potential in modulating metabolism, disease trajectory, and therapeutic responses. Metabolic reprogramming is a
hallmark of malignant progression, and a deeper understanding of this phenomenon in tumors and its effects on immune
regulation is a significant challenge that impedes cancer eradication. Dietary intake, as a key environmental factor, can influence
tumor metabolism. Emerging evidence indicates that dietary interventions might affect the nutrient availability in tumors, thereby
increasing the efficacy of cancer treatments. However, the intricate interplay between dietary interventions and the pathogenesis of
cancer and other diseases is complex. Despite encouraging results, the mechanisms underlying diet-based therapeutic strategies
remain largely unexplored, often resulting in underutilization in disease management. In this review, we aim to illuminate the
potential effects of various dietary interventions, including calorie restriction, fasting-mimicking diet, ketogenic diet, protein
1234567890();,:
restriction diet, high-salt diet, high-fat diet, and high-fiber diet, on cancer and the aforementioned diseases. We explore the
multifaceted impacts of these dietary interventions, encompassing their immunomodulatory effects, other biological impacts, and
underlying molecular mechanisms. This review offers valuable insights into the potential application of these dietary interventions
as adjunctive therapies in disease management.
1
Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China and 2Department of Oncology, Shanghai
Medical College, Fudan University, Shanghai 200032, China
Correspondence: Yi-Zhou Jiang (yizhoujiang@fudan.edu.cn)
These authors contributed equally: Yu-Ling Xiao, Yue Gong, Ying-Jia Qi
Dietary Intervention
• Calorie restriction
Metabolic Reprogramming • Fasting or fasting-mimicking diet
• Ketogenic diet
• Glucose metabolism • Protein restriction diet
• Lipid metabolism • High-salt diet
• Amino acid metabolism • Obesity and high-fat diet
Diseases
• Cancer
TME • Neurodegenerative diseases
• Autoimmune diseases
• Cardiovascular diseases
• Metabolic disorders
Gut Microbiome
Therapy
Fig. 1 Overview of the relationship between dietary interventions and diseases. The cellular microenvironment, including the tumor
microenvironment (TME), plays a crucial role in disease biology, and diet serves as a vital source of nutrients that can influence these
microenvironments. Metabolic reprogramming, a prominent feature associated with disease progression, can affect cell metabolism and
immune function. Dietary interventions, such as caloric restriction (CR), fasting-mimicking diet (FMD), and ketogenic diet (KD), can modulate
the progression and treatment sensitivity of various diseases, including cancer. Additionally, dietary interventions can alter the composition
and functional capacity of the gut microbiome, thereby indirectly influencing the progression and treatment of diseases. These direct and
indirect effects of dietary interventions can influence metabolic reprogramming, modulate immune responses, and potentially enhance the
clinical efficacy of treatments for various diseases. This figure was created with BioRender.com
immune cells, and stromal cells, in addition to components of the As our understanding of the complex relationships between
extracellular matrix. The interplay among these constituents, along diet, metabolic reprogramming, and various diseases continues to
with the challenging environmental conditions, exerts a significant evolve, it becomes increasingly evident that dietary components
influence on the growth trajectory and progression of tumors.22 and patterns significantly influence disease risk, prevention, and
For example, oxygen levels within the TME can vary due to progression. This review delves into the unique metabolic
increased metabolic demand from rapidly proliferating tumor characteristics and nutrient availability of tumors. Furthermore,
cells, resulting in low oxygen tension, known as hypoxia, in tissues. we investigate recent evidence and emerging trends concerning
In addition, nutrient availability, including the availability of the effects of dietary interventions on both cancer and other
glucose, fatty acids, and amino acids, can vary within the TME, diseases, underscoring the potential therapeutic benefits these
impacting metabolic processes and energy production. The dietary strategies may offer to a wide range of patients (Fig. 1).
accumulation of metabolic waste products and alterations in pH
can further contribute to a hostile TME, which can impair immune
function and promote tumor progression.23 These factors, along METABOLIC CHARACTERISTICS AND NUTRIENT AVAILABILITY
with dynamic interactions within the TME, play crucial roles in IN THE TUMOR
influencing tumor proliferation and the effectiveness of antitumor Cellular metabolism encompasses a complex array of biochemical
immune responses.24 reactions that utilize specific nutrients, including carbohydrates,
Glycolysis
Ribose
etaboli
Pyruvate Lactate
Glycoly
sis
Nucleotide
Glycoly
PPP
FAS Fatty acids
FAS
FAS
biosynthesis Citrate
acid m
sis
Acetyl-CoA Fatty acids
FAO
Amino
Oxaloacetate NADH
Citrate FADH2
TCA
OXPHOS
Succinyl-CoA cycle
α-KG Electron
e
transport chain
ak
OX AO
FA upt
Amino acid and
PH
id
F
O
protein synthesis Glutamine
ac
OS
tty
Fa
OX
P
Gly HOS is
coly olys
Memory T cells FAS sis Glyc HOS MDSCs
FAO OXP
Tregs NK cells
Fig. 2 Major metabolic pathways associated with different immune cell subtypes within the tumor microenvironment (TME). Summary of the
main metabolic pathways of immune cells, highlighting the distinctive metabolic characteristics and requirements of different subsets of
immune cells. This figure was created with BioRender.com
fatty acids, and amino acids. These nutrients are the primary glycolysis, the tricarboxylic acid (TCA) cycle, oxidative phosphor-
sources for maintaining energy homeostasis and synthesizing ylation (OXPHOS), the pentose phosphate pathway (PPP), fatty
macromolecules.25 Our focus here is on cancer metabolism, which acid oxidation (FAO), fatty acid synthesis (FAS) and the amino acid
differs from that in corresponding healthy tissues in terms of metabolic pathway30 (Fig. 2).
nutrient levels and metabolic demands.26 Within the TME, cancer
cells can establish an immunosuppressive metabolic microenvir- Glucose metabolism
onment by depriving immune cells of vital metabolites such as Glucose serves as a vital energy source, facilitating the functioning
glucose and oxygen while also elevating the levels of mediators of immune cells. Once transported across the plasma membrane,
such as lactate and adenosine that limit the function of immune glucose is metabolically processed via three distinct pathways:
cells.27 Therefore, different subsets of immune cells undergo glycolysis, the PPP, and the TCA cycle. Glycolysis, which occurs in
metabolic reprogramming in tumors, and specific nutrients are the cytosol, transforms glucose into pyruvate and lactate,
required for these metabolic programs.28,29 Generally, the meta- simultaneously generating adenosine triphosphate (ATP). Under
bolic programs that play vital roles in immune cells include aerobic conditions, pyruvate is channeled into the TCA cycle,
Calorie restriction Breast cancer Ly6C+-expressing memory T cells (CD4+Ly6C+)↑, Ly6C+CD8+ cells↑, 118
+ + 236
Fasting-mimicking diet Breast cancer CTLA-4 Tregs↑, PD-1 Tregs↑
Myeloid cells↓, M2-like macrophages↓, PMN-MDSCs↓, M-MDSCs↓, PD-
L1+ PMN-MDSCs↓, PD-L1+ M-MDSCs↓
137
Alternate day fasting Colorectal cancer TAMs↓, M2-like macrophages↓
151
Ketogenic diet Colorectal cancer Macrophages↑, M2 to M1 TAM polarization↑
Lactate↓
147
Hopx activation↑, colonic crypt cell proliferation↓, tumor growth↓
148
Non-small cell lung cancer Per↑, AMPK activation↑, SIRT1↑, tumor cell apoptosis↑, tumor cell
growth↓
149
Neuroendocrine cancer PI3K-Akt-mTOR signaling↓, tumor growth↓
+ + + + 152
Glioma CD4 T cells↑, CD4 T cells/Tregs ratio↑, IFNγ CD8 T cells↑,
TNF+CD8+ T cells↑, IL-2+CD8+ T cells↑, cytotoxic capability of CD8+
T cells↑, IFNγ+ NK cells↑, TNF+ NK cells↑
PD-1+CD8+ T cells↓, CTLA-4+CD8+ T cells↓, IL-10+ Tregs↓
156
Colorectal cancer Tumor cell ferroptosis↑, cachexia onset↑, overall survival↓
Adrenocortical cancer
163
Dietary restriction of Prostate cancer M1-like macrophages↑, M1-like macrophages linked proteins
protein/80% methionine- (CXCL11/I-TAC, IL-1α, IL-1β, IL-12p40, M-CSF, and IL-17A) ↑, CD8+ T
restricted diet cells↑, granzyme B+CD8+ T cells↑
M2-like macrophages↓, PMN-MDSCs↓, M-MDSCs↓, M2-like
macrophages linked proteins (C-reactive protein, FGF acidic, IL-33,
leptin, and MMP9) ↓
Dietary methionine Colorectal cancer CD8+ T cells↑, GZMB+CD8+ T cells↑, IFNγ+CD8+ T cells↑ 162
166
High-protein diet Bladder cancer Urinary urea↑, intracellular deposition of ammonia↑, tumor growth↓
167
Overactivation of CRP, MCPT2, MCPT9, EPXH2, SERPING1, SRGN,
CDKN1C, CDK6, CCNB1, PCNA, BAX, MAGEB16, SERPINE1, HSPA2, and
FOS
184
High-salt diet Melanoma The expression of Tnfα, Ifnγ, and Nos2↑
Lung cancer Suppressive function of MDSCs↓
185
Breast cancer IL-12p40↑, ICAM-1↑, IFNγ↑, TNFα↑, macrophages↑, M-MDSCs
Melanoma differentiation into antitumor macrophages↑, functions of PMN-
MDSCs switch from immunosuppressive to proinflammatory and
antitumor↑, CD4+ T cells↑, CD8+ T cells↑, IFNγ+CD4+ T cells↑,
IFNγ+CD8+ T cells↑, Th17 cells↑, TNFα+ Th17 cells↑
IL-6↓, IL-10↓, GM-CSF↓, MDSCs↓, M-MDSCs↓, Tregs↓
Melanoma NK cells↑, CD107a+ NK cells↑, IFNγ+ NK cells↑, Bifidobacterium↑ 183
High-fat diet Nonalcoholic steatohepatitis and Hepatic unconventional prefoldin RPB5 interactor (URI) ↑, Th17↑, 199
hepatocarcinogenesis IL-17A↑
Neutrophil infiltration into white adipose tissue, causing insulin
resistance and release of fatty acids
Helicobacter-induced chronic Immature myeloid cells↑, CD4+ T Cells↑, IL-17A↑, granulocyte 200
diglyceride↑
CD8+ T cells↓, leukocyte/tumor cell ratio↓, CD8+ T cells/tumor cell
ratio↓, Ki67+CD8+ T cells↓, CD8+/Treg ratio↓, ICOS+CD8+ T cells↓, PD-
1+CD8+ T cells↓, GZMB+CD8+ T cells↓, fatty acid↓
PD-1highCD4+ T cells↑, PD-1highCD8+ T cells↑ 209
296
Total macrophages↑, M2-like macrophages↑
M1-like macrophages↓
212
EVs↑, YAP signaling↑, CYR61↑, M2-like macrophages↑, liver
metastasis↑
Oral squamous cell carcinoma CD45+ cells↑, myeloid cells↑, MDSCs (mainly PMN-MDSCs) ↑, CCR1+ 205
expression of Cpt1a↑
Ki67+CD8+ T cells↓
217
High-cholesterol diet Colorectal cancer IL-1β↑, IL-6↑, TNFα↑, macrophages↑, NLRP3 inflammasome
activation↑
218
Macrophages↑
IFNγ+CD8+ T cells↓
Hepatocellular carcinoma NK cells↑, NK cells↑, effector function of NK cells↑, CD8+ T cells↑ 221
222
Fish oil high-fat diet (vs. Breast cancer ROS production in TAMs↑
cocoa butter high-fat diet) TAMs↓
Furthermore, research has shown that fasting, CR, and caloric of eating patterns, including complete and voluntary deprivation
restriction mimetics (CRMs) can promote T-cell-mediated tumor of food with no restriction on drinking water.129 An FMD is based
cytotoxicity, alter NK cell function, and potentially trigger on a regimen of low-calorie and low-protein foods that mimics the
immunogenic cell death, thereby stimulating cancer immunosur- effects of fasting but induces fewer side effects. This approach
veillance pathways.119 CRMs are pharmacological agents or retains the benefits of traditional fasting methods while minimiz-
natural compounds that imitate the biochemical effects of CR by ing their potential drawbacks.130
reducing the lysine acetylation rates of cellular proteins.120 Fasting or intake of an FMD can cause various metabolic
Examples of CRMs include hydroxycitrate (an inhibitor of ATP changes, including alterations in the systemic levels of hormones
citrate lyase), spermidine (an inhibitor of EP300 acetyl transferase and growth factors such as insulin, glucagon, growth hormone,
activity), and resveratrol (an activator of sirtuin-1 deacetylase IGF-1, glucocorticoids or adrenaline.131 In response to these
activity).121 Treatment with CRMs has been found to decrease the changes, normal cells activate protective mechanisms against
concentration of free IGF-1, promote autophagy in cancer cells, stress and toxic insults, thereby reducing their metabolic
and improve the antitumor immune response, resulting in a requirements and cell division rate. On the other hand, because
reduction in tumor growth when combined with immunogenic fasting or FMDs reduce tumor growth-promoting nutrients and
chemotherapeutics.119 CRM hydroxycitrate has been found to factors, cancer cells struggle to manage metabolite deprivation
stimulate autophagy in U2OS osteosarcoma cells in vitro, thereby and thus develop greater sensitivity to cancer therapies.132 In
increasing antitumor immunosurveillance and reducing tumor obesity-driven postmenopausal cancer mouse models, TRF was
mass in mice with autophagy-competent mutant KRAS-induced shown to delay the onset of tumors and reduce lung metastasis.
lung cancers.122 Moreover, in vitro treatment with resveratrol Moreover, TRF was found to increase systemic insulin sensitivity
inhibits mitochondrial respiration in breast cancer cell lines and decrease hyperinsulinemia. Importantly, TRF could also
through a SIRT1-dependent mechanism, diminishes the expres- restore the circadian rhythm of gene expression within tumors
sion of markers associated with breast CSCs, and promotes their while attenuating both tumor growth and insulin signal transduc-
differentiation.123 Collectively, these findings suggest that CRMs tion.133 Fasting can cause an “anti-Warburg effect” by reducing
may enhance antitumor immunosurveillance in preclinical models. aerobic glycolysis and glutaminolysis while increasing OXPHOS
Moderate physical activity, energy restriction, and their uncoupled from ATP synthesis.134 In cancer cells, OXPHOS
combination can also affect tumor growth. In fact, the combined increases reactive oxygen species (ROS) production and leads to
effects of moderate physical activity and 10% energy restriction oxidative stress, activation of p53 signaling and DNA damage,
(PA + ER) have been shown to significantly delay primary tumor particularly when combined with chemotherapy or other cancer
growth, reduce spontaneous metastases, and prolong survival. therapies.135 Therefore, the unique metabolic vulnerabilities of
These effects on tumor progression and survival are accompanied cancer cells, which differ from those of normal cells, can be
by beneficial changes in immune cell infiltrates within the strategically targeted to develop novel and effective therapeutic
microenvironment. Specifically, the PA + ER combination leads interventions. According to a recent study, the combination of
to an increase in the percentage of CD8+ T cells and a decrease in chemical treatment with an FMD reduces the expression of heme
the percentage of total MDSCs and MDSC subsets within oxygenase-1 (HO-1), which is a stress-responsive enzyme that
tumors.124 protects cancer cells against oxidative damage and apoptosis
Nevertheless, it is crucial to emphasize that there are in vivo. Interestingly, this combination treatment resulted in
established nutritional recommendations for cancer care, and upregulated HO-1 expression in normal cells. The downregulation
the weight loss or reduction in protein intake often associated of HO-1 production in cancer cells, in part, facilitated FMD-induced
with CR may conflict with these guidelines.125 These dietary chemosensitization of cancer cells by boosting CD8+ TIL-
practices could exacerbate the risk of malnutrition, sarcopenia, dependent cytotoxicity, which was possibly facilitated by
fatigue, delayed wound healing, and impaired immunity, particu- decreased Tregs.136 A separate study conducted with mouse
larly in cancer patients who are already at an increased age- models of colon cancer indicated that alternate day fasting for
associated risk for these conditions.126 Therefore, while exploring 2 weeks triggered autophagy in cancer cells, which in turn
dietary interventions for cancer treatment, the potential adverse downregulated CD73 expression. As a result, the production of
effects on overall patient health and nutritional status must be immunosuppressive adenosine in cancer cells was reduced,
carefully considered. ultimately preventing macrophages from acquiring an M2
immunosuppressive phenotype.137
Fasting or fasting-mimicking diet Clinical experiments have suggested that intake of an FMD can
In addition to CR, alternative approaches such as intermittent induce metabolic changes and increase antitumor immunity in
fasting (IF), including short-term fasting (STF), intake of an FMD, cancer patients. In fact, the final outcomes of an FMD-treated
and time-restricted feeding (TRF), which limits food consumption clinical trial (NCT03340935) demonstrated that a severely calorie-
to a specific time window each day, are being condisered.127,128 restricted, five-day FMD regimen was well tolerated and resulted
The term “fasting” has a broad definition, encompassing a range in substantial systemic metabolic changes in patients with
Calorie restriction Breast cancer Radiotherapy Teff/Tregs ratio↑, PD-1+CD8+ T cells↑ 261
Tregs↓
122
CRM hydroxycitrate Fibrosarcomas Chemotherapy Autophagy in tumor cells↑
Colorectal cancer Tregs↓
Lung cancer
132
Fasting or fasting- Lung cancer Immunotherapy CD8/Treg ratio↑, NK cells↑
mimicking diet Tregs↓, CD19+ B cells↓, PD-1+CD8+ T cells↓, PD-1+CD4+
T cells↓
251
Pancreatic cancer Chemotherapy Levels of equilibrative nucleoside transporter (hENT1) in
tumor cells↑
Levels of ribonucleotide reductase M1 (RRM1) in tumor
cells↓
252
Breast cancer Chemotherapy ROS in tumor cells↑
+ + + + + 236
Immunotherapy T cells↑, CD8 T cells↑, GZMB CD8 T cell↑, Ki67 CD8
T cells↑, γδ T cells↑, GZMB+ γδ T cells↑, Ki67+ γδ T cells↑,
Ki67+FOXP3-CD4+ T cells↑, OX40+FOXP3-CD4+ T cells↑,
PD-1+FOXP3-CD4+ T cells↑, Ki67+ Tregs↑, OX40+ Tregs↑,
PD-L1+ PMN-MDSCs↑, macrophages↑, PD-L1+
macrophages↑, ToxintCD8+ T cells↑, ToxintPD-
1intCD39lowCD8+ T cells↑
PMN-MDSCs↓, M2-like macrophages↓
265
Endocrine therapy Circulating IGF1, insulin, and leptin levels↓, AKT-mTOR
signaling↓
Breast cancer Chemotherapy CD8+ T cells↑, CD3+ T cells↑, granzyme-B↑ 136
258
Neuroblastoma Chemotherapy Tumor burden↓
259
Chemotherapy Serine, glutamine and glycine↑
Tumor blood-vessel density and intratumoral
hemorrhage↓, serum levels of essential amino acids↓
260
Pancreatic cancer Chemotherapy Tumor NADH levels↑
263
Radiotherapy Oxidative stress in tumor cells↑
268
PI3K inhibitors Hyperglycemia↓, insulin secretion↓, mTORC1 signaling↓
262
Lung cancer Radiation or radio- Oxidative stress in tumor cells↑
chemotherapy
Dietary restriction of Prostate cancer Immunotherapy CD8+ T cells↑, CD8+ T cells/M1-like ratio↑, CD8+ T cells/M2- 163
Melanoma
Melanoma Immunotherapy PD-1+ NK cells↓ 183
Calorie restriction NCT01819233 Surgery and radiotherapy Stage 0-I breast cancer Completed
NCT02792270 Pre-operative radiotherapy Sarcoma Unknown
NCT01802346 Chemotherapy Breast or prostate cancer Recruiting
Cyclic, 5-day calorie restriction NCT05703997 Atezolizumab Small cell lung cancer Not yet
recruiting
Calorie restriction and exercise NCT03131024 Anthracycline-containing Breast cancer Completed
chemotherapy
Intermittent calorie restriction and NCT05359848 Chemotherapy Cancer Recruiting
plant-based diet
Low-carbohydrate diet NCT02149459 Radiotherapy with or without Recurrent brain cancer Unknown
metformin
Very low carbohydrate diet NCT04035096 High dose intravenous vitamin C Stage IV colon cancer with KRAS and Unknown
BRAF mutation
Intermittent fasting NCT01175837 Chemotherapy Cancer Completed
NCT02607826 Chemotherapy Solid tumors Unknown
NCT06015087 Chemotherapy Breast cancer Recruiting
NCT01304251 Chemotherapy Breast cancer Completed
NCT05722288 Radiotherapy and/or chemotherapy Prostate, cervical or rectal cancer Recruiting
and/or hormone therapy
NCT04247464 Chemotherapy Colorectal cancer Enrolling
by
invitation
Nightly fasting NCT05023967 Chemotherapy and metformin Early breast cancer Recruiting
Prolonged nightly fasting NCT05083416 Immunotherapy Advanced head & neck cancer Active, not
recruiting
Alternate day fasting NCT05990426 Chemotherapy Endometrial, ovarian, fallopian tube or Not yet
primary peritoneal cancer recruiting
5:2 intermittent fasting NCT05861362 Radiotherapy Breast Cancer Completed
Time restricted eating with or without NCT05259410 Chemotherapy Breast Cancer Recruiting
Mediterranian diet
Intermittent fasting or vegan diet NCT03162289 Chemotherapy Gynecological cancer Active, not
recruiting
Intermittent fasting or Mediterranian NCT02710721 Chemotherapy with or without Advanced metastatic prostate cancer Completed
diet hormone therapy
Fasting-mimicking diet NCT03595540 Active cancer treatment Cancer Completed
NCT03709147 Metformin hydrochloride, cisplatin, Advanced LKB1-inactive lung Unknown
carboplatin, pemetrexed, adenocarcinoma
pembrolizumab
NCT05763992 Chemoimmunotherapy Triple-negative breast cancer Recruiting
NCT05503108 Neoadjuvant chemotherapy (ddAC, T) HR+, HER2- breast cancer Recruiting
NCT02126449 Neoadjuvant chemotherapy (AC>T) HER2- breast cancer Completed
NCT04248998 Neoadjuvant chemotherapy (AC>T) Triple-negative breast cancer Active, not
with or without metformin recruiting
NCT03340935 Standard cancer treatment Malignancies with the exception of Completed
small cell neuroendocrine tumors
NCT05921149 Carboplatin and paclitaxel Advanced or recurrent ovarian, Not yet
fallopian tube and primary peritoneal recruiting
cancer
Ketogenic diet NCT05119010 Nivolumab and ipilimumab Metastatic renal cell carcinoma Recruiting
NCT05938322 Neoadjuvant radiotherapy Locally advanced nonmetastatic rectal Not yet
adenocarcinoma recruiting
NCT04316520 Nivolumab + ipilimumab, Metastatic renal cell carcinoma Recruiting
pembrolizumab + axitinib, sunitinib or
pazopanib
NCT03962647 Letrozole Early-stage ER+, HER2- breast cancer Active, not
recruiting
NCT03535701 Paclitaxel Stage IV breast cancer Completed
NCT05234502 Neoadjuvant chemotherapy (AC>T) Breast cancer Not yet
recruiting
Vitamin B12 and folic acid NCT02679443 Chemotherapy Non-small cell lung cancer Completed
supplementation NCT00609518 Pemetrexed and dexamethasone Non-small cell lung cancer Completed
NCT00216099 Pemetrexed Hormone refractory prostate cancer Completed
Oral vitamin D NCT03467789 Radiotherapy Basal cell carcinoma Recruiting
NCT04864431 Chemotherapy Epithelial ovarian cancer Recruiting
NCT02603757 Chemotherapy Colorectal cancer Completed
NCT04091178 Chemotherapy Breast cancer Completed
NCT04677816 Chemotherapy Triple-negative breast cancer Recruiting
NCT03331562 Pembrolizumab Metastatic pancreatic ductal Completed
adenocarcinoma
Oral vitamin D and Omega-3 NCT05331807 Chemotherapy Breast cancer Recruiting
Oral vitamin E NCT03613389 Chemotherapy Pediatric cancer Unknown
NCT00363129 Chemotherapy Cancer Completed
Oral vitamin E and Hydrogen-rich NCT04713332 Radiotherapy Rectal cancer Unknown
water
Antioxidant-deficient diet NCT00486304 Chemotherapy and radiotherapy Oropharyngeal cancer Completed
Anti-inflammatory diet NCT03994055 Chemo-radiotherapy and Cervical cancer Active, not
brachytherapy recruiting
Low copper diet NCT00003751 Radiotherapy and penicillamine Glioblastoma multiforme Completed
Paleolithic diet and exercise NCT04574323 Radiotherapy Breast cancer Completed
Low-residue diet NCT00258401 Radiotherapy Uterine, cervical or prostate cancer Completed
KD has also shown promise in supporting the effectiveness of host. The constituents of the gut microbiome and their interac-
phosphatidylinositol 3 kinase (PI3K) inhibitors and overcoming tions with the host immune system can impact the development
drug resistance in various mouse cancer models, including of tumors and carcinogenesis.271 Various dietary patterns have
pancreatic, bladder, endometrial, and breast cancer models, as been found to significantly influence the composition and
well as acute myeloid leukemia.145 KD appears to enhance this functionality of the gut microbiome.272,273 It is through these
effectiveness by decreasing hyperglycemia and reducing insulin changes in the gut microbiome that dietary patterns can indirectly
secretion, actions correlated with a decrease in mTORC1 signaling influence the outcomes of cancer patients.274
within the tumor.268 In recent early studies, several interventional strategies, ranging
Finally, the combination of serine deprivation and biguanide from dietary interventions to fecal microbiome transplant (FMT)
treatment, such as phenformin and metformin, can lead to and prebiotic, probiotic and antibiotic treatments, have shown
metabolic stress in cancer cells. This stress arises from the forced promise in altering the composition or functional capacity of the
upregulation of glycolysis due to the biguanide-induced reduction gut microbiome.275 Two prospective cohort studies have sug-
in OXPHOS. Under conditions of serine deficiency, this stress may gested that diet-related inflammation can alter the gut micro-
exceed the metabolic flexibility of cancer cells, leading to their biome, leading to the development of CRC by suppressing
potential death and, consequently, enhanced anticancer adaptive antitumor immune responses.276,277 Other prospective
effects.269 cohort studies have revealed the associations between prudent
In summary, these findings underscore the potential of dietary diets (rich in whole grains and dietary fiber) and Western diets
interventions to modulate the therapeutic landscape of cancer (rich in red and processed meat, refined grains, and desserts) with
treatment, enhancing the effectiveness of drugs and potentially CRC risk and indicated that the effect of these diets may differ
overcoming resistance mechanisms. However, it should be viewed based on the presence of Fusobacterium nucleatum in tumor
with cautious optimism. The biological plausibility of diet tissue.278,279 Specifically, these studies showed that, compared
modifying treatment efficacy and resistance is compelling; with a Western diet, adhering to a long-term prudent diet is
however, the translation of this concept into clinical practice associated with a reduced risk of F. nucleatum-positive CRC;
requires rigorous validation. It is critical to remain grounded in however, it does not appear to mitigate the risk of F. nucleatum-
evidence-based medicine, recognizing that dietary strategies are negative CRC.278 A recent study investigated the impact of the gut
adjuncts, not replacements, for established therapeutic regimens. microbiota and dietary patterns on the response to ICIs in patients
Further exploration and clinical validation are necessary to fully with melanoma. The present study revealed that patients with
understand these interactions and to integrate dietary strategies microbiomes dominated by the Ruminococcaceae family had
into standard cancer care effectively and safely. greater response rates than did those with microbiomes
dominated by the Bacteroidaceae family. Furthermore, another
finding revealed that a poor response was associated with
DIET CHANGES THE GUT MICROBIOME IN CONJUNCTION WITH decreased intake of fiber and omega-3 fatty acids.280 These
ANTITUMOR EFFECTS AND CANCER TREATMENT results suggest that dietary interventions may be promising for
The gut microbiome encompasses the genetic makeup of all improving cancer treatment outcomes.
species within the gut, such as bacteria, viruses, yeasts, Accumulating data suggest that alterations in the gut micro-
protozoans, fungi, and archaea, and can be affected by a range biome primarily contribute to the progression, prognosis, and
of internal and external factors.270 The gut microbiota plays a treatment of cancer, primarily through interactions with the
significant role in influencing the health and disease status of the immune system. Metabolites produced by the microbiota play
macrophage
polarization
crosstalk of
NK cells and DCs
anti-PD-1
IFNγ+CD8+ T cells efficacy
Akkermansia muciniphila
b
ketone bodies tolerogenic c-di-AMP
High-fiber Diet
bacteria
KD Tcf1+PD-1+CD8+ T cells
β-HB immunogenic
bacteria
Bifidobacteria
butyrate-producing
pectin bacteria
T cells infiltration
T cells activation
ICB efficacy
intestinal Th17 cells
c d e
SCFAs-producing bacteria Lactobacillus reuteri
HSD HFD Tryptophan
Bacteroides fragilis Bifidobacterium
Bacteroides
colon
intratumor localization CRC progression
carcinogenesis
chemotherapy
gastric M2 TAM chemotherapy
NK cells resistance Treg cells ICB efficacy IFNγ+CD8+ T cells
carcinogenesis recruitment efficacy
Fig. 3 Mechanisms by which diet modulates antitumor effects and cancer treatment via modulation of the gut microbiome. a Calorie restriction (CR)
elevates IFNγ+CD8+ T cells in the tumor microenvironment (TME) by enriching Bifidobacterium bifidum and increasing acetate levels. b Ketogenic diet
(KD) induces a shift from tolerogenic (Lactobacilli spp., Clostridium asparagiforme) toward immunogenic bacteria (such as Akkermansia muciniphila)
driven by host production of ketone bodies, of which β-HB selectively inhibits the growth of bifidobacteria, resulting in KD-associated decreases in
intestinal Th17 cell levels. c High-salt diet (HSD) increases the abundance of Bifidobacterium and leads to intratumoral localization of Bifidobacterium,
further enhancing NK cell functions and tumor regression. HSD decreases the expression of IL-17A and iNOS and inhibits inflammation, which
reduces enterotoxigenic Bacteroides fragilis (ETBF)-promoted colon carcinogenesis. HSD exacerbates Helicobacter pylori infection and promotes gastric
carcinogenesis. d High-fat diet (HFD), through augmentation of queuosine-producing gut bacteria, can incite chemotherapy resistance in pancreatic
cancer patients. HFD reduces SCFA-producing bacteria and SCFA production, leading to decreased levels of short-chain fatty acids (SCFAs) that
activate the MCP-1/CCR2 axis, which promotes M2 TAM recruitment and polarization, ultimately contributing to colorectal cancer (CRC) progression.
High bile salt hydrolase (BSH) enzyme activity in an HFD mouse model activates the β-catenin/CCL28 axis, further inducing immunosuppressive Tregs
and accelerating CRC progression. e Dietary intake rich in tryptophan stimulates certain Bacteroides to produce the metabolite indole-3-acetic acid (3-
IAA). Increased levels of 3-IAA enhance the efficacy of chemotherapy treatment. Dietary intake rich in tryptophan, through the action of the probiotic
Lactobacillus reuteri (Lr), leads to the production of the metabolite indole-3-aldehyde (I3A). This metabolite promotes the production of IFNγ from
CD8+ T cells, thereby enhancing antitumor immunity and the efficacy of immune checkpoint inhibitors (ICIs). f High-fiber diet enriches Akkermansia
muciniphila which produces the microbiota-derived STING agonist c-di-AMP, inducing type I interferon (IFN-I) production by intratumoural
monocytes, resulting in various TME modulation pathways, including reprogramming of mononuclear phagocytes into immunostimulatory
monocytes and DCs, promoting macrophage polarization toward an antitumor phenotype and stimulating crosstalk between NK cells and DCs,
further enhancing the therapeutic effect of immunotherapy. Dietary fiber inulin can enhance the effectiveness of anti-PD-1 therapy by increasing the
abundance of beneficial commensal microbes (e.g., Akkermansia, Lactobacillus and Roseburia) and SCFAs, further increasing the number of stem-like T-
cell factor-1 (Tcf1)+PD-1+CD8+ T cells numbers. Dietary fiber pectin can improve the effectiveness of anti-PD-1 therapy by increasing the abundance
of butyrate-producing bacteria, further promoting T-cell infiltration and activation in the TME. This figure was created with BioRender.com
GABA
hippocampal catalase
Akkermansia gamma-glutamyl
GABA
Parabacteroides amino acids
KD
Bifidobacteria TNF
β-HB
β-HB
NLRP3 inflammasome activation
microglial activation
Firmicutes
Tenericutes
Opisthokonta
FMD Proteobacteria loss of dopaminergic neuron
propionic acid glial cells TNF-α and IL-1β
isobutyric acid
butyric acid
valeric acid TyrRS
tyrosine sirtuin pathway
amyloid-β protein
CR
BDNF, HSP70
NANA
naive CD4+ T cells
HFD CD4+ TEMs
Tregs
amyloid-β protein
MD phosphorylated tau
Fig. 4 Impact of different diets on neurodegenerative diseases. The ketogenic diet (KD) can enhance inhibitory neurotransmission and anti-
inflammatory effects in epilepsy, influence the gut microbiota, and elevate beneficial metabolites. KD is particularly beneficial for treating
pediatric drug-resistant epilepsy with elevated specific Bifidobacteria and TNF. In Alzheimer’s disease (AD) and Parkinson’s disease (PD), KD
could counteract decreased β-HB levels, inhibit the NLRP3 inflammasome, reduce pathology, and alleviate symptoms by inhibiting microglial
activation. Fasting mimicking diet (FMD) enhances the gut microbiota composition and metabolites, inhibiting neuroinflammation. This
results in the attenuated loss of dopaminergic neurons in the substantia nigra in patients with PD. Caloric restriction (CR) may prevent AD by
lowering serum tyrosine levels, reversing the exhaustion of tyrosyl-tRNA synthetase (TyrRS), and upregulating the sirtuin pathway, which
attenuates the amyloidogenic processing of amyloid-β protein precursor (APP). Dietary restriction can increase brain-derived neurotrophic
factor (BDNF) and chaperone heat-shock protein-70 (HSP70) levels in the striatum and cortex, which are relevant to Huntington’s disease (HD).
High-fat diet (HFD) can accelerate recognition-memory impairment in an AD mouse model by increasing blood N-acetylneuraminic acid
(NANA) levels, leading to systemic immune exhaustion. Conversely, the Mediterranean diet (MD) may protect against memory decline and
mediotemporal atrophy by lowering amyloid-β protein and phosphorylated tau levels, reducing AD risk. This figure was created with
BioRender.com
Prevotella copri
fumarate, succinate
RA SLE
IL-1β
TGFβ signalling
TNFα
IL-6
PGE2 myelin-debris clearance
in microglia
IBD MS
+ antibiotics
Fig. 5 Impact of different diets on autoimmune diseases. Extravirgin olive oil (EVOO) can reduce joint inflammation and degradation in
rheumatoid arthritis (RA) due to its phenolic compounds. However, the protective effects of a high-fiber diet can be reversed by Prevotella
copri colonization, which promotes proinflammatory responses. Fish oil supplementation can suppress proinflammatory cytokines and
cartilage degradation, improving RA outcomes. Vitamin D can inhibit the proliferation, differentiation, and function of B and T cells,
potentially reducing inflammatory cytokine expression in systemic lupus erythematosus (SLE) patients. A diet low in fermentable
oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) can alleviate gut symptoms in quiescent inflammatory bowel
disease (IBD) patients, possibly by regulating the immune response through reducing fecal microbial abundance. However, a high-fat diet
(HFD) can exacerbate pre-IBD inflammation by impairing epithelial mitochondrial bioenergetics and triggering microbiota disruptions,
especially when combined with antibiotics. High salt diet (HSD) can exacerbate autoimmune conditions such as multiple sclerosis (MS) by
promoting the induction of pathogenic Th17 cells. Intermittent fasting (IF) can improve MS by reducing the number of IL-17-producing T cells,
increasing the number of Tregs in the gut, and enhancing antioxidative microbial metabolic pathways. However, the Western diet can impair
myelin-debris clearance in microglia, hindering lesion recovery after demyelination and potentially contributing to MS induction. This figure
was created with BioRender.com
patterns other than single nutrients as supplementary treatments IBD treatment according to clinical guidelines.364 Obesity is a risk
for SLE still require further investigation.363 factor for IBD, especially for CD.365 As a potential trigger of obesity,
Ulcerative colitis (UC) and Crohn’s disease (CD) are the two HFD, together with antibiotics, exacerbates inflammation in pre-
major clinical phenotypes of IBD. Dietary management and IBDs by impairing epithelial mitochondrial bioenergetics and
microbiota modulation have been clinically recommended for triggering microbiota disruptions in mouse models.366 However,
metabolic health and providing a potential treatment strategy for levels and reducing hepatic steatosis in patients with NAFLD,
obesity and its related metabolic disorders.410 possibly by downregulating hepatic inflammatory pathways,
Cohort studies have demonstrated that healthy diets, including modifying lipogenic gene expression and increasing levels of
the Portfolio diet, DASH diet, and MD, are associated with a autophagy.427,428
decreased risk of T2DM.411–413 The promotion of SCFA-producing PCOS features a series of metabolic irregularities, mainly
bacteria induced by dietary fibers observed in T2DM patients androgen excess and ovarian dysfunction. A meta-analysis
suggests the potential value of fiber supplementation in clinical showed that women with PCOS have a lower overall diet quality
practice.414 In addition, increased fiber consumption is associated with higher cholesterol, lower magnesium and lower zinc
with decreased insulin resistance, the mechanism of which mainly intake.429 Dietary modification with lower caloric intake to achieve
includes the gut microbiota and associated molecules.415,416 IF is weight loss is recommended as a first-line therapy for managing
an effective strategy for controlling weight and increasing insulin PCOS, and higher supplementary nutrient intake, including
sensitivity in patients with diabetes and can also improve vitamin D, chromium and ω-3, may also benefit patients suffering
cardiometabolic outcomes.417,418 The every-other-day fasting from PCOS.430 MD, KD and their combination can all lead to
(EODF) regimen selectively stimulates beige fat development significant improvements in body weight, metabolic function and
within white adipose tissue and shifts the gut microbiota ovulatory dysfunction in PCOS patients.431–433 In addition, IF may
composition in experimental models, explaining the mechanism be beneficial for treating anovulatory PCOS by reducing body fat
through which IF ameliorates obesity, insulin resistance, and and improving menstruation, hyperandrogenemia, insulin resis-
hepatic steatosis.419 KD has therapeutic effects on glycemia, lipid tance and chronic inflammation.434 CR may also improve weight
control, and weight reduction in T2DM patients.420 However, KD and metabolic disorders in patients with PCOS, alone or in
may contribute to decreased sensitivity to peripheral insulin and combination with supplementation.435 However, the exact
impaired glucose tolerance by upregulating insulin receptors, as mechanisms of these dietary interventions remain unclear and
determined by previous studies, which contradicts clinical need further exploration.
findings.421 While the potential of dietary interventions to influence
NAFLD features hepatic steatosis or adiposity with a potential systemic diseases of the whole body is supported by various
risk of developing into inflammation, fibrosis, and cancer. MD, as studies, a critical outlook reveals the necessity for more rigorous,
the most recommended dietary pattern for NAFLD, can reduce long-term clinical trials to validate these findings. It is essential to
liver steatosis and improve insulin sensitivity even without weight approach these interventions with caution, considering individual
loss in an insulin-resistant population.422 Reduced liver fat may be differences and the intricate balance of potential benefits against
associated with ameliorated inflammation induced by antiox- nutritional deficiencies or other risks.
idants, low glycemic response induced by dietary fiber, and
improved hepatic lipid metabolism.423 KD is more clinically
meaningful for glycemic control in individuals with T2DM and CONCLUSIONS AND PERSPECTIVES
NAFLD than low-calorie diet or high-carbohydrate, low-fat (HCLF) Our review provides compelling evidence that dietary interven-
diet.424,425 Mechanistically, ketone bodies may modulate inflam- tions, including calorie restriction, fasting or FMD, KD, protein
mation and fibrosis in hepatic cells.426 IF alone or combined with restriction diet, HSD, HFD, and high-fiber diet, have substantial
exercise is effective at lowering intrahepatic triglyceride (IHTG) potential for modulating metabolism, redirecting disease
High
diet -fib
h-fat er d
iet
Hig
Calorie intake
Ev
ng
JNK, IKK
er
di
y-
inflammation
fee
ot
he
ted
r-d
&inflammation bacteria
tric
ay
res
adjusted rhythmic
fast
creatine-mediated beige fat
Time
thermogenesis
ing
Obesity T2DM
PVT dysfunction gut microbiota
alteration
daytime sleepiness
glucose tolerance
Fructose ov
weight
O
H
HO
blood lipid
er-in
OH
et
CH2
c di
OH
take
inflammation
eni
metabolic function hepatic fibrosis
tog
ovulatory dysfunction
Ke
weight IHTG
hepatic steaosis
Ca
lo
lipogenic gene
ie
r
re autophagy
st
ric
tio
n
ise
exerc
Intermittent-fasting&
Fig. 7 Impact of different diets on metabolic disorders. High-fat diet (HFD) can directly increase caloric intake, induce inflammatory mediators
such as JNK and IκB kinase (IKK) to promote hypothalamic inflammation, and contribute to adipose tissue hypoxia and inflammation, which all
lead to the development of obesity and/or insulin resistance. Over-intake of fructose can also increase caloric intake and induce obesity by
impairing hepatic insulin sensitivity. However, time-restricted feeding (TRF) with equivalent caloric intake from HFD can adjust various
signaling pathways and rhythmic creatine-mediated thermogenesis and reverse excessive daytime sleepiness induced by paraventricular
thalamic nucleus (PVT) dysfunction, resulting in a protective effect on HFD-induced obesity. High-fiber diet can reduce inflammation and
insulin resistance by influencing the gut microbiota and associated molecules, for instance, SCFA-producing bacteria. Every-other-day fasting
(EODF) regimen can also shift the gut microbiota composition and stimulate beige fat development within white adipose tissue to inhibit
insulin resistance. Ketogenic diet (KD) is clinically beneficial for the glycemic control of type 2 diabetes mellitus (T2DM) and nonalcoholic fatty
liver disease (NAFLD). However, in experimental models, KD can decrease sensitivity to peripheral insulin by upregulating insulin receptors.
Intermittent fasting (IF) alone or combined with exercise can reduce intrahepatic triglyceride (IHTG) levels and hepatic steatosis in NAFLD
patients by downregulating hepatic inflammatory pathways, modifying lipogenic gene expression and inducing autophagy. Calorie restriction
(CR) can be effective at reducing weight loss and reversing ovulatory/metabolic dysfunction in polycystic ovarian syndrome (PCOS) patients.
This figure was created with BioRender.com
progression, and enhancing therapeutic responses. These findings Despite compelling evidence, the potential impact of dietary
highlight the pivotal role of diet, an important environmental interventions on disease treatment, particularly cancer treatment,
factor, in influencing tumor metabolism and the course of various is not fully understood.436 The latest American Society of Clinical
diseases, such as cancer, neurodegenerative diseases, autoim- Oncology (ASCO) guidelines suggest that “there is currently
mune diseases, CVD, and metabolic disorders. insufficient evidence to recommend for or against dietary