Next Article in Journal
New Aspect of Composition and Biological Properties of Glechoma hederacea L. Herb: Detailed Phytochemical Analysis and Evaluation of Antioxidant, Anticoagulant Activity and Toxicity in Selected Human Cells and Plasma In Vitro
Next Article in Special Issue
Oral Nutritional Supplements Reduce Body Weight Loss after Gastrectomy in Patients with Gastric Cancer: A Systematic Review and Meta-Analysis of Randomized Controlled Trials
Previous Article in Journal
Growth and Duration of Inflammation Determine Short- and Long-Term Outcome in Very-Low-Birth-Weight Infants Requiring Abdominal Surgery
Previous Article in Special Issue
Malnutrition Prevention after Allogeneic Hematopoietic Stem Cell Transplantation (alloHSCT): A Prospective Explorative Interventional Study with an Oral Polymeric Formulation Enriched with Transforming Growth Factor Beta 2 (TGF-β2)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Mediterranean Lifestyle to Contrast Low-Grade Inflammation Behavior in Cancer

by
Rosa Divella
1,*,
Graziella Marino
2,*,
Stefania Infusino
3,
Laura Lanotte
1,
Gaia Gadaleta-Caldarola
4 and
Gennaro Gadaleta-Caldarola
1
1
Unità Operativa Complessa di Oncologia, Ospedale “Mons. A. R. Dimiccoli”, Asl BT, Viale Ippocrate 15, 76121 Barletta, Italy
2
Department of Breast Unit—Centro d Riferimento Oncologico della Basilicata, IRCCS-CROB, Via Padre Pio 1, 85028 Rionero in Vulture, Italy
3
Unità Operativa Complessa di Oncologia, Ospedale “SS Annunziata”, Via Felice Migliori 1, 87100 Cosenza, Italy
4
Scienze e Tecnologie Alimentari, Università di Parma, Via Delle Scienze 59/A, 43124 Parma, Italy
*
Authors to whom correspondence should be addressed.
Nutrients 2023, 15(7), 1667; https://doi.org/10.3390/nu15071667
Submission received: 9 February 2023 / Revised: 21 March 2023 / Accepted: 25 March 2023 / Published: 29 March 2023

Abstract

:
A healthy diet and an active lifestyle are both effective ways to prevent, manage, and treat many diseases, including cancer. A healthy, well-balanced diet not only ensures that the body gets the right amount of nutrients to meet its needs, but it also lets the body get substances that protect against and/or prevent certain diseases. It is now clear that obesity is linked to long-term diseases such as heart disease, diabetes, and cancer. The main reasons for people being overweight or obese are having bad eating habits and not moving around enough. Maintaining weight in the normal range may be one of the best things to avoid cancer. It has been scientifically proven that those who perform regular physical activity are less likely to develop cancer than those who lead a sedentary lifestyle. Moving regularly not only helps to maintain a normal body weight, avoiding the effects that favor tumor growth in overweight subjects, but also makes the immune system more resistant by counteracting the growth of tumor cells. Physical activity also helps prevent cardiovascular and metabolic diseases. In this review, it is highlighted that the association between the Mediterranean diet and physical activity triggers biological mechanisms capable of counteracting the low-grade chronic inflammation found in patients with cancer. This assumes that healthy lifestyles associated with cancer therapies can improve the expectations and quality of life of cancer patients.

1. Introduction

Approximately 30–35% of all malignancies are attributed to so-called lifestyle factors, which include an imbalanced diet, being overweight or obese, abusing alcohol, and inactivity [1]. Obesity, defined as abnormal excess accumulation of fat in adipose tissue, causes chronic low-grade inflammation. It is associated with a high risk of developing the four main types of non-communicable diseases (NCDs), namely, type 2 diabetes, metabolic syndrome, cardiovascular disease, and several types of cancer [2,3]. In tumors of the breast, colon, liver, and prostate, obesity is generally a predictor of poor clinical outcomes. Consequently, diet, viewed as the totality of eating habits, is a significant environmental component capable of regulating neoplastic risk either as a protective factor or as a risk factor, depending on the quality, amount, and frequency of intake of the foods that comprise it [4]. The relationship between nutrition and the immune system is crucial to both sustaining health and causing illness. When the immune system is hyperactive as a result of an unhealthy lifestyle and an imbalanced diet, a low-grade chronic inflammatory state is induced, which increases the risk of developing chronic degenerative illnesses and cancers [5,6]. Nutrition has a dual nature because it can be either a weapon of prevention and therapy or a cause of pathological events if it is primarily composed of pro-inflammatory foods consumed on a regular basis. Diets, such as the traditional Mediterranean diet (MD), and regular physical exercise are effective methods for preventing overweight, obesity, and NCDs, as well as reducing visceral fat and, as a result, the generation of pro-inflammatory cytokines and hormonal imbalances [7,8]. Moreover, the UNESCO in 2010 officially defined the MD as a cultural heritage of humanity, exalting conviviality, sensory stimulation, socialization, biodiversity, and seasonality, aspects that can reinforce the MD’s beneficial effects on wellbeing, quality of life, and healthy physical activity. Furthermore, a balanced diet is now regarded as a very effective tool for the prevention and reduction of mortality and the recurrence of oncological illnesses [9,10]. These intrinsic characteristics of the Mediterranean diet allow us to place it in a really healthy lifestyle, which is reflected in the culinary and working traditions (fishing and sheep farming) of the Mediterranean people, which require intense physical effort (far from a sedentary life) and, therefore, high energy consumption.
Physical exercise benefits cancer patients, including those receiving oncological treatments such as chemotherapy, hormone therapy, radiation, and surgery, as well as those in the rehabilitation phase and long-term survivors [11]. Physical exercise is now shown to be safe for cancer patients and must be included in treatment regimens, even though it must be tailored to the subject being treated, the kind of pathology, comorbidities, physical conditions, and effort tolerance [9]. Physical exercise, when combined with a balanced diet and a proper lifestyle (no smoking or drinking), leads to a prolongation of the expected lifespan and a decrease in the risk of chronic degenerative illnesses and tumors, which are statistically common in the adult population [12]. This review focuses attention on the importance of adopting a healthy lifestyle defined as a Mediterranean lifestyle where nutrition and physical activity become the two fundamental building blocks for achieving a good state of health and quality of life in cancer patients.

2. Physical Activity and Inflammation

Physical activity, especially when performed moderately and on a regular basis, has an anti-inflammatory effect and lowers inflammation markers [13]. Muscle contraction causes the release of proteins called myokines, some of which have local and systemic endocrine effects and are involved in lipid, glucose, and bone metabolism, and hence contribute to the anti-inflammatory effects of exercise [14,15]. In addition to aiding in weight regulation; preventing diabetes type 2, insulin resistance, and metabolic syndrome; and enhancing cognitive abilities and slowing the progression of neurodegenerative diseases such as Parkinson’s and Alzheimer’s, it plays an essential role in the body’s metabolic processes [16,17]. Additionally, it may halt the development of cancer, making it useful both for primary cancer prevention and to cancer survivors [18]. Recent research indicates that even moderate exercise on a treadmill for 20 to 30 min can stimulate the immune system and produce an anti-inflammatory response, resulting in a decrease in TNF-α (tumor necrosis factor-alpha) and a beneficial effect on low-grade chronic inflammation [19].

3. From Sedentary Lifestyle to Chronic Inflammation Status

Sedentary living is defined by excessive inactivity throughout the day and frequent sitting in front of the TV, computer, or video games; at work; and in the car [20]. Energy is expended more readily when standing due to the contraction of muscles (especially those of the lower extremities), whereas sitting has been linked to detrimental effects on lipid metabolism. Further, eating while seated is linked to weight gain, which in turn raises the risk of developing metabolic syndrome and obesity [21]. Under these conditions, the visceral adipose tissue is altered, resulting in changes in the production of steroid hormones and adipokines, metabolic disorders, and chronic subclinical inflammation. Low-grade chronic inflammation caused by elevated levels of tumor necrosis factor alpha (TNF-a), interleukin 6 (IL-6), leptin, and other inflammatory adipokines, including plasminogen activator inhibitor 1 (PAI-1), is a risk factor for the development of malignant neoplasms in people who are overweight or obese, Figure 1 [22,23,24]. In addition, this state of inflammation leads to hyperinsulinemia, which in turn promotes insulin resistance with the increased production of IGF-1 (insulin-like growth Factor-1) and hormone imbalances, including an increase in estrogens as a result of androgen conversion at the adipose tissue level, which in turn stimulates the proliferation of mammary gland and endometrial epithelial cells [25,26]. The chance of developing cancer due to these causes is elevated. The detection of C-reactive protein (PCR) in the blood has prognostic significance since it serves as an essential indication of the metabolic and inflammatory state caused by the liver in response to IL-6 emitted by adipocytes [27]. To live a healthy and long life free from diabetes, degenerative diseases, heart disease, and cancer, it is possible to make positive changes to one’s behavior (mental, physical, and nutritional) at any time.

4. Physical Activity and Cancer

Physical exercise, when properly performed in moderation and in conjunction with an anti-inflammatory diet such as the traditional Mediterranean diet, also has an essential role in preventing and fighting cancer, as has been shown by various epidemiological, observational, and meta-analysis studies [28,29]. Consistent exercise alters several biochemical markers associated with cancer cell metabolism [30]. Modulatory effects on hormones, growth factors, and cytokines are of special interest as potential pathways by which physical exercise prevents and fights cancer [31]. One of exercise’s primary effects is a decrease in plasma concentrations of the hormone estrogen. Inducing cell proliferation and progression, blocking apoptosis, and promoting angiogenesis are all ways in which estrogens promote breast carcinogenesis [32]. Several studies have revealed that menopausal women who engage in regular exercise have lower levels of free estrogen in their blood [33,34,35]. Exercise has several effects on endogenous estrogen, including weight loss, changes in serum adipokine levels, and a lower circulating insulin concentration [36,37]. The final pathway causes a rise in the hormone sex-hormone-binding globulin (SHBG), a protein that binds to estrogens and decreases their bioavailability. It is postulated that fat mass loss mediates the relationship between exercise and estrogen reduction through increased SHBG [38,39,40]. Given that fat cells produce most of the body’s endogenous estrogen after menopause, a connection between the two seems reasonable [41]. Increased circulating amounts of estrogen and/or insulin and IGF-1 may be one mechanism by which excess body fat promotes cancer development and progression in postmenopausal women (insulin-like growth factor) [42,43]. The expression of IGF-1 is greater in ER-positive tumors than in ER-negative tumors because both insulin and IGF-1 increase aromatase enzyme activity in adipose tissue, which boosts local estrogen synthesis [44,45]. As a result, an increase in adiposity is responsible for the elevated amounts of circulating estrogen seen in obese women.
Physical exercise may be used to prevent the side effects of chemotherapy and radiation, such as weariness and nausea, during treatment times [46,47]. Exercises carried out gradually and consistently may enhance certain benefits related to the tumor’s pathology and the treatments given, lower the risk of cardiovascular disease, and prevent the reappearance of neoplastic disease [48,49]. Aerobic exercise enhances cardiovascular and respiratory health as well as strength, endurance, and musculoskeletal health in cancer patients [50,51]. Exercise prevents weight gain, diminished muscular strength, and cardiotoxicity in individuals on androgen deprivation for prostate cancer [52]. Rehabilitative exercise may help with secondary lymphedema and limb mobility in women who have had axillary dissection and a mastectomy for breast cancer [53]. In fact, when more muscle groups are engaged, lymph outflow improves. Adipose and muscle tissue may undergo epigenetic changes as a result of physical exercise, which can be shown as an increase or reduction in DNA methylation [54,55]. The most interest is being shown in research on the methylation of certain genes. For instance, Nakajima’s research demonstrates that alternating aerobic exercise (three minutes of brisk walking followed by three minutes of running) affects the methylation of the ASC gene, which regulates the cytokines IL-1beta and IL-18, important for the inflammatory response [56]. The p53 tumor suppressor gene, which is suppressed in many malignancies and causes histone alterations as well as differences in the expression of microRNAs that govern protein synthesis and the regeneration of muscle mass and fiber type, is also more likely to be expressed when people are physically active [57,58,59]. It is also crucial to schedule physical activity each week. It has been shown that individuals with colon cancer who engage in at least six hours of physical activity each week have a much higher survival rate than those who engage in less than one hour of exercise each week [60,61]. A 50,000-woman study from the Albert Einstein College of Medicine in New York indicated that individuals who maintained a lifestyle in accordance with the American Cancer Society’s guidelines had a 31% lower chance of acquiring breast cancer [62]. The combined use of physical exercise and cancer treatments such as chemotherapy has positive results [63,64]. Exercise helps cancer patients even four years after diagnosis, according to May et al. In participants with a 4-year follow up, it was noted that adverse effects, such as weariness, decreased [65]. Physical exercise has a number of beneficial biological effects, some of which have been hypothesized to explain the preventative activity, including the decrease of insulin resistance, chronic inflammation, and immune system dysfunction; the reduction of fat mass in obese individuals; and the blood levels of hormones including insulin, estrogen, and growth factors [66,67,68].

5. Diet and Chronic Inflammation

Chronic low-grade inflammation is at the root of chronic-degenerative illnesses in the elderly and malignancies. Sedentarism, extra food calories, an improper diet, and bad food all lead to chronic inflammation [69,70,71]. Fat accumulation, particularly visceral obesity, controls the production of pro-inflammatory cytokines, which encourage the establishment of an inflammatory environment, as well as the synthesis of hormones, and the conversion of androgens into estrogen in adipose tissue, which contribute to tumor development in the endometrium, breast, and colon [72,73,74]. Additionally, diets high in processed foods and “junk food” change the gut bacterial flora, causing dysbiosis, which worsens and perpetuates the inflammatory state [75,76]. According to recent research by Zhang et al., a poor diet seems to be a major risk factor for malignant tumors. Colorectal cancer had the largest number of diagnoses and the highest percentage connected to diet (38.3%) [77]. Relevant variables were a lack of whole grains and a high intake of processed meats. Cancer was most common in males aged 45 to 64. The study’s American authors validate the statistics given by the American Association for Cancer Research, which states that food is responsible for 5% of cancers [78]. According to the latest research, a diet rich in whole grains, vegetables, fruit, fish oil, extra virgin olive oil, prebiotic fibers, spices, and a moderate intake of red wine, such as the traditional Mediterranean diet, also with the addition of spices, may modify and positively impact the immune functions with a protective and preventative activity and can maintain the microbiota in eubiosis, safeguarding the intestinal barrier [79,80]. As a result, germs and endotoxins that might lead to persistent low-grade inflammation are kept from spreading throughout the body.

6. The Mediterranean Diet (MD)

Thanks to its unusual preventative effects, the MD has attracted the attention of scientists throughout the world. The Mediterranean diet constitutes a food model that characterizes not only a lifestyle but also a culture and has been indicated as a hub for improving health, quality of life, and life span. Numerous studies have highlighted the positive correlation between MD and longevity; individuals who adhere to a nutritional style such as this have a longer life expectancy [81,82]. Moreover, the Mediterranean diet prevents many metabolic, cardiovascular, and neurodegenerative diseases; insulin resistance; and different types of cancer [83,84]. Today, cancer represents the second leading cause of death in the world, immediately after cardiovascular diseases, but its onset curve has lowered in parallel with other chronic degenerative diseases, such as diabetes and obesity. In this context, diet plays a very important role. Epidemiological and clinical studies support the association between nutrition and the development or progression of different cancer malignancies such as colon, breast, prostate, and other cancers, defining these tumors as diet-associated cancers [85,86]. If followed correctly, it may help people avoid the chronic inflammation that results from being overweight, developing metabolic syndrome, and gaining excess weight due to a poor diet. In general, the Mediterranean diet is full of healthy nutrients, including antioxidants, anti-inflammatories, and insulin sensitizers [87,88]. Within several months of its implementation, it has been linked to less inflammation and has been shown to reduce inflammatory cytokines (IL-6 and TNF-alpha) and the C-reactive protein and to enhance anti-inflammatory IL-10 [89,90]. In addition to lowering insulin levels and correcting hepatic steatosis, the MD has been shown to reverse the metabolic syndrome in a significant number of patients. It is a healthy routine that has been shown to reduce cancer risk [91,92]. This is because eating these foods together helps prevent DNA damage, cell proliferation, and the survival of cancer cells by lowering oxidative and inflammatory processes inside the body [93]. Furthermore, in the DIMENU study (Dieta Mediterranea and Nuoto), according to the data reported in the literature, serum from adolescents with high adherence to the Mediterranean diet reduced inflammation in human macrophage in vitro models, suggesting that it could have a positive impact on the prevention of chronic diseases, including cancers in adulthood [94].

7. Role of an Anti-Inflammatory and Calorie-Restricted Diet in Cancer Prevention

The typical Mediterranean diet can help avoid cancer. To achieve this objective, it is recommended to follow the diet following the suggestions below and illustrated in the model of the Mediterranean diet pyramid illustrated in Figure 2.

The Typical Mediterranean Diet Recommendations to Follow

(1)
Consume whole grains every day, three portions a day of vegetables, two portions a day of fruit, and legumes 3–4 times a week.
(2)
Limit the consumption of red meat, avoid processed meats such as preserved meats, cold cuts, and frankfurters.
(3)
Stay away from items high on the glycemic index, such as white bread, white rice, and simple sweets. Dinner carbs are acidifying and may raise blood sugar and insulin levels, so it is best to avoid them.
(4)
Fruit is best eaten on an empty stomach, either first thing in the morning or late in the afternoon and never toward the conclusion of a meal, especially one that is high in carbs. Do not eat fruit salad since it contains ingredients with wildly varying pH levels, many of which might lead to stomach and bowel issues.
(5)
Foods high in acidity and polyamines should be avoided (vegetables of the nightshade family, oranges, tangerines, mandarin oranges).
(6)
Stay away from packaged foods with hydrogenated polyunsaturated fatty acids (trans), which cause high cholesterol and inflammation throughout the body, which damages cells. Trans fats can be found in bakery products such as industrial bread, cookies, and pastries, as well as in ready-to-eat meals and French fries.
(7)
Food supplements should not be taken unless they have been properly tested in clinical trials.
(8)
Observe a moderate calorie restriction while making sure that all essential nutrients are in the diet. This is to make sure that the nutritional status is not affected (for example 1–2 days a week).
(9)
Weight should be managed in a healthy way; one should “stay trim” and monitor one’s waist circumference (waist size), which should be no more than 80–88 cm in women and 94–102 cm in males.
(10)
Even more so as you get older, it is important to get your body moving every day; try going for a brisk 30 min walk, walking 10,000 steps, or spending an hour at the gym.
Numerous studies suggest that calorie restriction decreases plasma insulin, glucose levels, sex hormones, and inflammatory cytokines; increases detoxifying enzymes; decreases oxidative stress; and increases adiponectin, an anti-inflammatory protein produced by adipocytes [95,96,97]. When the diet contains all the required nutrients, 25–30% less calories in the diet lengthens animal life and minimizes the occurrence of cancers [98]. In fact, it has been shown that calorie restriction stimulates the AMPK (AMP-activated protein kinase) protein, which decreases the production of mTOR (mammalian target of rapamycin), one of the major oncogenes and regulators of cell growth and proliferation [99,100]. When caloric restriction is accompanied by a drop in animal protein, plasma levels of IGF-1 decline, which, in conjunction with insulin, activates the PI3K–Akt–mTORC signaling pathway [101]. As an alternative to mitochondrial respiration, the latter may stimulate cell growth and aerobic glycolysis, which constitutes the primary energy source of cancer cells. Aerobic glycolysis generates lactic acid and acidifies the milieu in which tumors thrive; the acidity increases tumor dissemination by boosting angiogenesis through VEGF (vascular endothelial growth factor) [102]. It has been shown that even brief periods of fasting (a couple of days a week) lower blood sugar, insulin, and IGF-1 levels and enhance the efficacy of oncological therapies [103,104]. By blocking mTOR, calorie restriction is in fact able to limit cancer cells’ DNA repair capabilities. In the days before chemotherapy, calorie restriction might be prescribed to cancer patients. In conclusion, the key points of physical exercise and a healthy Mediterranean diet are given below:
  • Modulates insulin levels, reduces insulin resistance and insulin-like growth factor (IGF-1).
  • Fights against weight gain and obesity.
  • Decreases the production of inflammatory adipokines and leptin (mitogenic factor) while increasing the production of adiponectin (pro-apoptotic factor) by adipocytes with anti-inflammatory and antitumor action.
  • Reduces plasma levels of estrogens, which are involved in the growth of breast and endometrial cancer cells.
  • Contributes to the anti-inflammatory action with the muscular release of anti-inflammatory myokines whose protective effect in many tumors has been demonstrated.
  • Increases intestinal motility, thereby reducing the contact time of the intestinal mucosa with carcinogenic compounds.
  • Modifies the composition and metabolic profile of the microbiota, exerting a protective action in inflammatory bowel diseases.

8. Conclusions

The American Institute for Cancer Research and the World Cancer Research Fund estimate that following appropriate dietary guidelines, engaging in consistent daily physical exercise, and maintaining a healthy weight would prevent 30–40% of all cancers. Additionally, the same treatment might lower mortality from pancreatic, prostate, colorectal, breast, and other cancers by up to 50%. When it concerns prostate cancer, dietary and lifestyle modifications not only have the obvious effect of preventing the neoplasm but also of regulating its behavior in the development stages, either by slowing down the evolution of the tumor when it is already present or by preventing its return after a drastic surgery [105,106]. From a physiological point of view, physical activity improves insulin sensitivity, lowers visceral body fat by bringing the adipocytes back into a state of balance in the production of adipokines favoring anti-inflammatory ones, and lowers estrogen levels, as well as inflammation markers such as CRP, which together facilitate cancer formation. Physical exercise may lower the probability of developing cancer, Friedenreich et al. reports a 25% reduction in the risk of breast cancer, cancerous cell proliferation, and the side effects of adjuvant therapy [107]. High-intensity workouts carried out for a short period of time have also been shown to benefit patients with different forms of cancer [108]. In conclusion, it seems adequate to engage in 150 to 300 min per week of aerobic physical activity of moderate intensity, or 75 to 150 min of vigorous and intensive activity, in order to gain a health advantage. The combination of a balanced diet with an active lifestyle is an effective strategy for the prevention, management, and treatment of several illnesses [109]. A healthy and balanced diet not only ensures an appropriate supply of nutrients capable of meeting the body’s demands but also permits the incorporation of chemicals that play a protective and/or preventative function against specific pathological disorders. A diet abundant in fruit, vegetables, legumes, fatty fish with a high content of omega-3 fatty acids, and extra virgin olive oil; a balanced ratio between omega-6 and omega-3 fatty acids; the daily intake of foods containing physiologically active herbal components; and the use of fish oil supplements all show preventative activities [110,111].
Dietary and lifestyle interventions may result in functional alterations of the endocrine system, a decrease in inflammatory events, and regulation of genetic transcription [112,113,114]. Considering that cancer is a disease caused by genetic abnormalities that come from damage to cellular DNA, this component is crucial. It has been established that various lines of intervention have beneficial effects not only on the cardiovascular system but also on tumor pathology, slowing the evolution of tumors. The main focuses of cancer supportive care are on enhancing survival, motivating patients to take action, and lessening the burden of treatment-related side effects. Cancer treatment plans should always incorporate complementary measures, such as nutrition and exercise. International guidelines and scientific evidence show that a well-planned diet and exercise program may reduce or eliminate many of the negative consequences of both the ailment and its treatment. Despite a growing amount of evidence, sports and physical therapy and nutritional therapy are seldom included into standard cancer treatment plans. Interprofessional communication is crucial for incorporating dietary and physical interventions, as well as sports therapy, into cancer care.

Author Contributions

Writing—review and editing, R.D.; visualization G.M., S.I. and G.G.-C. (Gaia Gadaleta-Caldarola); supervision, G.G.-C. (Gennaro Gadaleta-Caldarola) and L.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The study did not report any data.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ng, R.; Sutradhar, R.; Yao, Z.; Wodchis, W.P.; Rosella, L.C. Smoking, drinking, diet and physical activity-modifiable lifestyle risk factors and their associations with age to first chronic disease. Int. J. Epidemiol. 2020, 49, 113–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Marshall, K.; Beaden, P.; Durrani, H.; Tang, K.; Mogilevskii, R.; Bhutta, Z. The role of the private sector in noncommunicable disease prevention and management in low- and middle-income countries: A series of systematic reviews and thematic syntheses. Int. J. Qual. Stud. Health Well-Being 2023, 18, 2156099. [Google Scholar] [CrossRef] [PubMed]
  3. Cheah, Y.K.; Lim, K.K.; Ismail, H.; Mohd Yusoff, M.F.; Kee, C.C. Can the association between hypertension and physical activity be moderated by age? J. Taibah Univ. Med. Sci. 2023, 18, 844–854. [Google Scholar] [CrossRef] [PubMed]
  4. Gherasim, A.; Arhire, L.I.; Niță, O.; Popa, A.D.; Graur, M.; Mihalache, L. The relationship between lifestyle components and dietary patterns. Proc. Nutr. Soc. 2020, 79, 311–323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Soldati, L.; Di Renzo, L.; Jirillo, E.; Ascierto, P.A.; Marincola, F.M.; De Lorenzo, A. The influence of diet on anti-cancer immune responsiveness. J. Transl. Med. 2018, 16, 75. [Google Scholar] [CrossRef]
  6. Childs, C.E.; Calder, P.C.; Miles, E.A. Diet and Immune Function. Nutrients 2019, 11, 1933. [Google Scholar] [CrossRef] [Green Version]
  7. Calcaterra, V.; Verduci, E.; Vandoni, M.; Rossi, V.; Fiore, G.; Massini, G.; Berardo, C.; Gatti, A.; Baldassarre, P.; Bianchi, A.; et al. The Effect of Healthy Lifestyle Strategies on the Management of Insulin Resistance in Children and Adolescents with Obesity: A Narrative Review. Nutrients 2022, 14, 4692. [Google Scholar] [CrossRef]
  8. Sood, S.; Feehan, J.; Itsiopoulos, C.; Wilson, K.; Plebanski, M.; Scott, D.; Hebert, J.R.; Shivappa, N.; Mousa, A.; George, E.S.; et al. Higher Adherence to a Mediterranean Diet Is Associated with Improved Insulin Sensitivity and Selected Markers of Inflammation in Individuals Who Are Overweight and Obese without Diabetes. Nutrients 2022, 14, 4437. [Google Scholar] [CrossRef]
  9. United Nations Educational, Scientific, and Cultural Organization (UNESCO). Representative List of the Intangible Cultural Heritage of Humanity. Available online: http://www.unesco.org/culture/ich/index.php (accessed on 8 October 2015).
  10. Dominguez, L.J.; Veronese, N.; Di Bella, G.; Cusumano, C.; Parisi, A.; Tagliaferri, F.; Ciriminna, S.; Barbagallo, M. Mediterranean diet in the management and prevention of obesity. Exp. Gerontol. 2023, 174, 112121. [Google Scholar] [CrossRef]
  11. Iolascon, G.; Di Pietro, G.; Gimigliano, F.; Mauro, G.L.; Moretti, A.; Giamattei, M.T.; Ortolani, S.; Tarantino, U.; Brandi, M.L. Physical exercise and sarcopenia in older people: Position paper of the Italian Society of Orthopaedics and Medicine (OrtoMed). Clin. Cases Miner. Bone Metab. 2014, 11, 215–221. [Google Scholar] [CrossRef] [Green Version]
  12. Cerqueira, É.; Marinho, D.A.; Neiva, H.P.; Lourenço, O. Inflammatory Effects of High and Moderate Intensity Exercise-A Systematic Review. Front. Physiol. 2020, 10, 1550. [Google Scholar] [CrossRef] [Green Version]
  13. Hoffmann, C.; Weigert, C. Skeletal Muscle as an Endocrine Organ: The Role of Myokines in Exercise Adaptations. Cold Spring Harb. Perspect. Med. 2017, 7, a029793. [Google Scholar] [CrossRef] [Green Version]
  14. Leal, L.G.; Lopes, M.A.; Batista, M.L., Jr. Physical Exercise-Induced Myokines and Muscle-Adipose Tissue Crosstalk: A Review of Current Knowledge and the Implications for Health and Metabolic Diseases. Front. Physiol. 2018, 9, 1307. [Google Scholar] [CrossRef]
  15. Zhao, R.R.; O’Sullivan, A.J.; Fiatarone Singh, M.A. Exercise or physical activity and cognitive function in adults with type 2 diabetes, insulin resistance or impaired glucose tolerance: A systematic review. Eur. Rev. Aging Phys. Act. 2018, 15, 1. [Google Scholar] [CrossRef] [Green Version]
  16. García-Muñoz, C.; González-García, P.; Casuso-Holgado, M.J.; Martínez-Calderón, J.; Heredia-Rizo, A.M. Are movement-based mindful exercises (QIGONG, TAI CHI, AND YOGA) beneficial for stroke and Parkinson’s disease? A scoping review. Complement. Ther. Med. 2023, 72, 102912. [Google Scholar] [CrossRef]
  17. Mills, R.C., 3rd. Breast Cancer Survivors, Common Markers of Inflammation, and Exercise: A Narrative Review. Breast Cancer 2017, 11, 1178223417743976. [Google Scholar] [CrossRef]
  18. Dimitrov, S.; Hulteng, E.; Hong, S. Inflammation and exercise: Inhibition of monocytic intracellular TNF production by acute exercise via β2-adrenergic activation. Brain Behav. Immun. 2017, 61, 60–68. [Google Scholar] [CrossRef]
  19. Caprara, G. Mediterranean-Type Dietary Pattern and Physical Activity: The Winning Combination to Counteract the Rising Burden of Non-Communicable Diseases (NCDs). Nutrients 2021, 13, 429. [Google Scholar] [CrossRef]
  20. Park, J.H.; Moon, J.H.; Kim, H.J.; Kong, M.H.; Oh, Y.H. Sedentary Lifestyle: Overview of Updated Evidence of Potential Health Risks. Korean J. Fam. Med. 2020, 41, 365–373. [Google Scholar] [CrossRef]
  21. Hill, J.O.; Wyatt, H.R.; Peters, J.C. The Importance of Energy Balance. Eur. Endocrinol. 2013, 9, 111–115. [Google Scholar] [CrossRef]
  22. Czaja-Stolc, S.; Potrykus, M.; Kaska, Ł.; Malgorzewicz, S. Pro-Inflammatory Profile of Adipokines in Obesity. Encyclopedia. Available online: https://encyclopedia.pub/entry/21914 (accessed on 7 February 2023).
  23. Ragino, Y.I.; Stakhneva, E.M.; Polonskaya, Y.V.; Kashtanova, E.V. The Role of Secretory Activity Molecules of Visceral Adipocytes in Abdominal Obesity in the Development of Cardiovascular Disease: A Review. Biomolecules 2020, 10, 374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Chaplin, A.; Rodriguez, R.M.; Segura-Sampedro, J.J.; Ochogavía-Seguí, A.; Romaguera, D.; Barceló-Coblijn, G. Insights behind the Relationship between Colorectal Cancer and Obesity: Is Visceral Adipose Tissue the Missing Link? Int. J. Mol. Sci. 2022, 23, 13128. [Google Scholar] [CrossRef] [PubMed]
  25. Sidorkiewicz, I.; Jóźwik, M.; Niemira, M.; Krętowski, A. Insulin Resistance and Endometrial Cancer: Emerging Role for microRNA. Cancers 2020, 12, 2559. [Google Scholar] [CrossRef] [PubMed]
  26. Cohen, D.H.; LeRoith, D. Obesity, type 2 diabetes, and cancer: The insulin and IGF connection. Endocr.-Relat. Cancer 2023, 19, F27–F45. [Google Scholar] [CrossRef]
  27. Menzel, A.; Samouda, H.; Dohet, F.; Loap, S.; Ellulu, M.S.; Bohn, T. Common and Novel Markers for Measuring Inflammation and Oxidative Stress Ex Vivo in Research and Clinical Practice—Which to Use Regarding Disease Outcomes? Antioxidants 2021, 10, 414. [Google Scholar] [CrossRef]
  28. Vallis, J.; Wang, P.P. The Role of Diet and Lifestyle in Colorectal Cancer Incidence and Survival. In Gastrointestinal Cancers; Morgado-Diaz, J.A., Ed.; Exon Publications: Brisbane, Australia, 2022; Chapter 2. [Google Scholar]
  29. Shahid, R.; Haq, I.U.; Mahnoor; Awan, K.A.; Iqbal, M.J.; Munir, H.; Saeed, I. Diet and lifestyle modifications for effective management of polycystic ovarian syndrome (PCOS). J. Food Biochem. 2022, 46, e14117. [Google Scholar] [CrossRef]
  30. Hoedjes, M.; Nijman, I.; Hinnen, C. Psychosocial Determinants of Lifestyle Change after a Cancer Diagnosis: A Systematic Review of the Literature. Cancers 2022, 14, 2026. [Google Scholar] [CrossRef]
  31. Thomas, R.J.; Kenfield, S.A.; Jimenez, A. Exercise-induced biochemical changes and their potential influence on cancer: A scientific review. Br. J. Sports Med. 2017, 51, 640–644. [Google Scholar] [CrossRef]
  32. Torregrosa, C.; Chorin, F.; Beltran, E.E.M.; Neuzillet, C.; Cardot-Ruffino, V. Physical Activity as the Best Supportive Care in Cancer: The Clinician’s and the Researcher’s Perspectives. Cancers 2022, 14, 5402. [Google Scholar] [CrossRef]
  33. García-Chico, C.; López-Ortiz, S.; Peñín-Grandes, S.; Pinto-Fraga, J.; Valenzuela, P.L.; Emanuele, E.; Ceci, C.; Graziani, G.; Fiuza-Luces, C.; Lista, S.; et al. Physical Exercise and the Hallmarks of Breast Cancer: A Narrative Review. Cancers 2023, 15, 324. [Google Scholar] [CrossRef]
  34. Razzak, Z.A.; Khan, A.A.; Farooqui, S.I. Effect of aerobic and anaerobic exercise on estrogen level, fat mass, and muscle mass among postmenopausal osteoporotic females. Int. J. Health Sci. 2019, 13, 10–16. [Google Scholar]
  35. Elliott, C.G.; Vidal-Almela, S.; Harvey, P.; O’Donnell, E.; Scheid, J.L.; Visintini, S.; Reed, J.L. Examining the Role of Physical Activity Interventions in Modulating Androgens and Cardiovascular Health in Postmenopausal Women: A Narrative Review. CJC Open. 2022, 5, 54–71. [Google Scholar] [CrossRef]
  36. Ennour-Idrissi, K.; Maunsell, E.; Diorio, C. Effect of physical activity on sex hormones in women: A systematic review and meta-analysis of randomized controlled trials. Breast Cancer Res. 2015, 17, 139. [Google Scholar] [CrossRef] [Green Version]
  37. Golbidi, S.; Laher, I. Exercise induced adipokine changes and the metabolic syndrome. J. Diabetes Res. 2014, 2014, 726861. [Google Scholar] [CrossRef] [Green Version]
  38. Schmidt, S.; Monk, J.M.; Robinson, L.E.; Mourtzakis, M. The integrative role of leptin, estrogen and the insulin family in obesity-associated breast cancer: Potential effects of exercise. Obes. Rev. 2015, 16, 473–487. [Google Scholar] [CrossRef] [Green Version]
  39. Fortunati, N.; Catalano, M.G.; Boccuzzi, G.; Frairia, R. Sex Hormone-Binding Globulin (SHBG), estradiol and breast cancer. Mol. Cell. Endocrinol. 2010, 316, 86–92. [Google Scholar] [CrossRef] [Green Version]
  40. Del Mar Grasa, M.; Gulfo, J.; Camps, N.; Alcalá, R.; Monserrat, L.; Moreno-Navarrete, J.M.; Ortega, F.J.; Esteve, M.; Remesar, X.; Fernández-López, J.A.; et al. Modulation of SHBG binding to testosterone and estradiol by sex and morbid obesity. Eur. J. Endocrinol. 2017, 176, 393–404. [Google Scholar] [CrossRef] [Green Version]
  41. Knight, M.G.; Anekwe, C.; Washington, K.; Akam, E.Y.; Wang, E.; Stanford, F.C. Weight regulation in menopause. Menopause 2021, 28, 960–965. [Google Scholar] [CrossRef]
  42. Geczik, A.M.; Falk, R.T.; Xu, X.; Ansong, D.; Yarney, J.; Wiafe-Addai, B.; Edusei, L.; Dedey, F.; Vanderpuye, V.; Titiloye, N.; et al. Measured body size and serum estrogen metabolism in postmenopausal women: The Ghana Breast Health Study. Breast Cancer Res. 2022, 24, 9. [Google Scholar] [CrossRef]
  43. Abildgaard, J.; Ploug, T.; Al-Saoudi, E.; Wagner, T.; Thomsen, C.; Ewertsen, C.; Bzorek, M.; Pedersen, B.K.; Pedersen, A.T.; Lindegaard, B. Changes in abdominal subcutaneous adipose tissue phenotype following menopause is associated with increased visceral fat mass. Sci. Rep. 2021, 11, 14750. [Google Scholar] [CrossRef]
  44. Bhardwaj, P.; Au, C.C.; Benito-Martin, A.; Ladumor, H.; Oshchepkova, S.; Moges, R.; Brown, K.A. Estrogens and breast cancer: Mechanisms involved in obesity-related development, growth and progression. J. Steroid Biochem. Mol. Biol. 2019, 189, 161–170. [Google Scholar] [CrossRef] [PubMed]
  45. Kim, D.S.; Scherer, P.E. Obesity, Diabetes, and Increased Cancer Progression. Diabetes Metab. J. 2021, 45, 799–812. [Google Scholar] [CrossRef] [PubMed]
  46. Su, B.; Wong, C.; Hong, Y.; Chen, S. Growth factor signaling enhances aromatase activity of breast cancer cells via post-transcriptional mechanisms. J. Steroid Biochem. Mol. Biol. 2011, 123, 101–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Bleach, R.; Sherlock, M.; O’Reilly, M.W.; McIlroy, M. Growth Hormone/Insulin Growth Factor Axis in Sex Steroid Associated Disorders and Related Cancers. Front. Cell Dev. Biol. 2021, 9, 630503. [Google Scholar] [CrossRef]
  48. Mustian, K.M.; Sprod, L.K.; Palesh, O.G.; Peppone, L.J.; Janelsins, M.C.; Mohile, S.G.; Carroll, J. Exercise for the management of side effects and quality of life among cancer survivors. Curr. Sport. Med. Rep. 2009, 8, 325–330. [Google Scholar] [CrossRef] [Green Version]
  49. Piraux, E.; Caty, G.; Aboubakar Nana, F.; Reychler, G. Effects of exercise therapy in cancer patients undergoing radiotherapy treatment: A narrative review. SAGE Open Med. 2020, 8, 2050312120922657. [Google Scholar] [CrossRef]
  50. Zimmerman, A.; Planek, M.I.C.; Chu, C.; Oyenusi, O.; Paner, A.; Reding, K.; Skeete, J.; Clark, B.; Okwuosa, T.M. Exercise, cancer and cardiovascular disease: What should clinicians advise? Cardiovasc. Endocrinol. Metab. 2020, 10, 62–71. [Google Scholar] [CrossRef]
  51. Clemente-Suárez, V.J.; Redondo-Flórez, L.; Rubio-Zarapuz, A.; Martínez-Guardado, I.; Navarro-Jiménez, E.; Tornero-Aguilera, J.F. Nutritional and Exercise Interventions in Cancer-Related Cachexia: An Extensive Narrative Review. Int. J. Environ. Res. Public Health. 2022, 19, 4604. [Google Scholar] [CrossRef]
  52. Campbell, K.L.; Winters-Stone, K.M.; Wiskemann, J.; May, A.M.; Schwartz, A.L.; Courneya, K.S.; Zucker, D.S.; Matthews, C.E.; Ligibel, J.A.; Gerber, L.H.; et al. Exercise Guidelines for Cancer Survivors: Consensus Statement from International Multidisciplinary Roundtable. Med. Sci. Sports Exerc. 2019, 51, 2375–2390. [Google Scholar] [CrossRef] [Green Version]
  53. Brown, J.C.; Winters-Stone, K.; Lee, A.; Schmitz, K.H. Cancer, physical activity, and exercise. Compr. Physiol. 2012, 2, 2775–2809. [Google Scholar]
  54. Galvão, D.A.; Taaffe, D.R.; Spry, N.; Newton, R.U. Exercise can prevent and even reverse adverse effects of androgen suppression treatment in men with prostate cancer. Prostate Cancer Prostatic Dis. 2007, 10, 340–346. [Google Scholar] [CrossRef]
  55. Baumann, F.T.; Reike, A.; Hallek, M.; Wiskemann, J.; Reimer, V. Does Exercise Have a Preventive Effect on Secondary Lymphedema in Breast Cancer Patients Following Local Treatment?—A Systematic Review. Breast Care 2018, 13, 380–385. [Google Scholar] [CrossRef]
  56. Światowy, W.J.; Drzewiecka, H.; Kliber, M.; Sąsiadek, M.; Karpiński, P.; Pławski, A.; Jagodziński, P.P. Physical Activity and DNA Methylation in Humans. Int. J. Mol. Sci. 2021, 22, 12989. [Google Scholar] [CrossRef]
  57. Barrón-Cabrera, E.; Ramos-Lopez, O.; González-Becerra, K.; Riezu-Boj, J.I.; Milagro, F.I.; Martínez-López, E.; Martínez, J.A. Epigenetic Modifications as Outcomes of Exercise Interventions Related to Specific Metabolic Alterations: A Systematic Review. Lifestyle Genom. 2019, 12, 25–44. [Google Scholar] [CrossRef]
  58. Nakajima, K.; Takeoka, M.; Mori, M.; Hashimoto, S.; Sakurai, A.; Nose, H.; Higuchi, K.; Itano, N.; Shiohara, M.; Oh, T.; et al. Exercise effects on methylation of ASC gene. Int. J. Sports Med. 2010, 31, 671–675. [Google Scholar] [CrossRef] [Green Version]
  59. Drummond, M.J.; McCarthy, J.J.; Sinha, M.; Spratt, H.M.; Volpi, E.; Esser, K.A.; Rasmussen, B.B. Aging and microRNA expression in human skeletal muscle: A microarray and bioinformatics analysis. Physiol. Genom. 2011, 43, 595–603. [Google Scholar] [CrossRef] [Green Version]
  60. Feng, Z.; Zhang, C.; Wu, R.; Hu, W. Tumor suppressor p53 meets microRNAs. J. Mol. Cell Biol. 2011, 3, 44–50. [Google Scholar] [CrossRef]
  61. Zacharewicz, E.; Lamon, S.; Russell, A.P. MicroRNAs in skeletal muscle and their regulation with exercise, ageing, and disease. Front. Physiol. 2013, 4, 266. [Google Scholar] [CrossRef] [Green Version]
  62. Hong, J.; Park, J. Systematic Review: Recommendations of Levels of Physical Activity among Colorectal Cancer Patients (2010–2019). Int. J. Environ. Res. Public Health 2021, 18, 2896. [Google Scholar] [CrossRef]
  63. Oruç, Z.; Kaplan, M.A. Effect of exercise on colorectal cancer prevention and treatment. World J. Gastrointest. Oncol. 2019, 11, 348–366. [Google Scholar] [CrossRef]
  64. Catsburg, C.; Miller, A.B.; Rohan, T.E. Adherence to cancer prevention guidelines and risk of breast cancer. Int. J. Cancer 2014, 135, 2444–2452. [Google Scholar] [CrossRef] [PubMed]
  65. Ferioli, M.; Zauli, G.; Martelli, A.M.; Vitale, M.; McCubrey, J.A.; Ultimo, S.; Capitani, S.; Neri, L.M. Impact of physical exercise in cancer survivors during and after antineoplastic treatments. Oncotarget 2018, 9, 14005–14034. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Courneya, K.S.; Booth, C.M. Exercise as cancer treatment: A clinical oncology framework for exercise oncology research. Front. Oncol. 2022, 12, 957135. [Google Scholar] [CrossRef] [PubMed]
  67. Witlox, L.; Hiensch, A.E.; Velthuis, M.J.; Steins Bisscho, C.N.; Los, M.; Erdkamp, F.L.G.; Bloemendal, H.J.; Verhaar, M.; Ten Bokkel Huinink, D.; Van der Wall, E.; et al. Four-year effects of exercise on fatigue and physical activity in patients with cancer. BMC Med. 2018, 16, 86. [Google Scholar] [CrossRef] [Green Version]
  68. Burini, R.C.; Anderson, E.; Durstine, J.L.; Carson, J.A. Inflammation, physical activity, and chronic disease: An evolutionary perspective. Sports Med. Health Sci. 2020, 2, 1–6. [Google Scholar] [CrossRef]
  69. Imierska, M.; Kurianiuk, A.; Błachnio-Zabielska, A. The Influence of Physical Activity on the Bioactive Lipids Metabolism in Obesity-Induced Muscle Insulin Resistance. Biomolecules 2020, 10, 1665. [Google Scholar] [CrossRef]
  70. Pedersen, B.K.; Saltin, B. Exercise as medicine—Evidence for prescribing exercise as therapy in 26 different chronic diseases. Scand. J. Med. Sci. Sports. 2015, 25 (Suppl. 3), 1–72. [Google Scholar] [CrossRef] [Green Version]
  71. Calder, P.C.; Ahluwalia, N.; Brouns, F.; Buetler, T.; Clement, K.; Cunningham, K.; Esposito, K.; Jönsson, L.S.; Kolb, H.; Lansink, M.; et al. Dietary factors and low-grade inflammation in relation to overweight and obesity. Br. J. Nutr. 2011, 106 (Suppl. 3), S5–S78. [Google Scholar] [CrossRef]
  72. Minihane, A.M.; Vinoy, S.; Russell, W.R.; Baka, A.; Roche, H.M.; Tuohy, K.M.; Teeling, J.L.; Blaak, E.E.; Fenech, M.; Vauzour, D.; et al. Low-grade inflammation, diet composition and health: Current research evidence and its translation. Br. J. Nutr. 2015, 114, 999–1012. [Google Scholar] [CrossRef] [Green Version]
  73. Divella, R.; Gadaleta Caldarola, G.; Mazzocca, A. Chronic Inflammation in Obesity and Cancer Cachexia. J. Clin. Med. 2022, 11, 2191. [Google Scholar] [CrossRef]
  74. Ramos-Nino, M.E. The role of chronic inflammation in obesity-associated cancers. ISRN Oncol. 2013, 2013, 697521. [Google Scholar] [CrossRef] [Green Version]
  75. Zimta, A.A.; Tigu, A.B.; Muntean, M.; Cenariu, D.; Slaby, O.; Berindan-Neagoe, I. Molecular Links between Central Obesity and Breast Cancer. Int. J. Mol. Sci. 2019, 20, 5364. [Google Scholar] [CrossRef] [Green Version]
  76. Budek, M.; Nuszkiewicz, J.; Piórkowska, A.; Czuczejko, J.; Szewczyk-Golec, K. Inflammation Related to Obesity in the Etiopathogenesis of Gastroenteropancreatic Neuroendocrine Neoplasms. Biomedicines 2022, 10, 2660. [Google Scholar] [CrossRef]
  77. Prescott, S.L.; Logan, A.C. Transforming Life: A Broad View of the Developmental Origins of Health and Disease Concept from an Ecological Justice Perspective. Int. J. Environ. Res. Public Health 2016, 13, 1075. [Google Scholar] [CrossRef] [Green Version]
  78. Mansour, S.R.; Moustafa, M.A.A.; Saad, B.M.; Hamed, R.; Moustafa, A.A. Impact of diet on human gut microbiome and disease risk. New Microbes New Infect. 2021, 41, 100845. [Google Scholar] [CrossRef]
  79. Key, T.J.; Bradbury, K.E.; Perez-Cornago, A.; Sinha, R.; Tsilidis, K.K.; Tsugane, S. Diet, nutrition, and cancer risk: What do we know and what is the way forward? BMJ 2020, 368, m511. [Google Scholar] [CrossRef] [Green Version]
  80. Zhang, F.F.; Cudhea, F.; Shan, Z.; Michaud, D.S.; Imamura, F.; Eom, H.; Ruan, M.; Rehm, C.D.; Liu, J.; Du, M.; et al. Preventable Cancer Burden Associated With Poor Diet in the United States. JNCI Cancer Spectr. 2019, 3, pkz034. [Google Scholar] [CrossRef] [Green Version]
  81. Turati, F.; Dalmartello, M.; Bravi, F.; Serraino, D.; Augustin, L.; Giacosa, A.; Negri, E.; Levi, F.; La Vecchia, C. Adherence to the World Cancer Research Fund/American Institute for Cancer Research Recommendations and the Risk of Breast Cancer. Nutrients 2020, 12, 607. [Google Scholar] [CrossRef] [Green Version]
  82. Ribeiro, G.; Ferri, A.; Clarke, G.; Cryan, J.F. Diet and the microbiota–gut–brain-axis: A primer for clinical nutrition. Curr. Opin. Clin. Nutr. Metab. Care. 2022, 25, 443–450. [Google Scholar] [CrossRef]
  83. Merra, G.; Noce, A.; Marrone, G.; Cintoni, M.; Tarsitano, M.G.; Capacci, A.; De Lorenzo, A. Influence of Mediterranean Diet on Human Gut Microbiota. Nutrients 2020, 13, 7. [Google Scholar] [CrossRef]
  84. Itsiopoulos, C.; Mayr, H.L.; Thomas, C.J. The anti-inflammatory effects of a Mediterranean diet: A review. Curr. Opin. Clin. Nutr. Metab. Care. 2022, 25, 415–422. [Google Scholar] [CrossRef] [PubMed]
  85. Romagnolo, D.F.; Selmin, O.I. Mediterranean Diet and Prevention of Chronic Diseases. Nutr. Today 2017, 52, 208–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Dominguez, L.J.; Di Bella, G.; Veronese, N.; Barbagallo, M. Impact of Mediterranean Diet on Chronic Non-Communicable Diseases and Longevity. Nutrients 2021, 13, 2028. [Google Scholar] [CrossRef] [PubMed]
  87. Muscogiuri, G.; Verde, L.; Sulu, C.; Katsiki, N.; Hassapidou, M.; Frias-Toral, E.; Cucalón, G.; Pazderska, A.; Yumuk, V.D.; Colao, A.; et al. Mediterranean Diet and Obesity-related Disorders: What is the Evidence? Curr. Obes. Rep. 2022, 11, 287–304. [Google Scholar] [CrossRef]
  88. Janssen, J.A. The Impact of Westernization on the Insulin/IGF-I Signaling Pathway and the Metabolic Syndrome: It Is Time for Change. Int. J. Mol. Sci. 2023, 24, 4551. [Google Scholar] [CrossRef]
  89. Lécuyer, L.; Laouali, N.; Dossus, L.; Shivappa, N.; Hébert, J.R.; Agudo, A.; Tjonneland, A.; Halkjaer, J.; Overvad, K.; Katzke, V.A.; et al. Inflammatory potential of the diet and association with risk of differentiated thyroid cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort. Eur. J. Nutr. 2022, 61, 3625–3635. [Google Scholar] [CrossRef]
  90. Hayati, Z.; Montazeri, V.; Shivappa, N.; Hebert, J.R.; Pirouzpanah, S. The association between the inflammatory potential of diet and the risk of histopathological and molecular subtypes of breast cancer in northwestern Iran: Results from the Breast Cancer Risk and Lifestyle study. Cancer 2022, 128, 2298–2312. [Google Scholar] [CrossRef]
  91. Bifulco, M.; Pisanti, S. The mystery of longevity in Cilento: A mix of a good dose of genetic predisposition and a balanced diet based on the Mediterranean model. Eur. J. Clin. Nutr. 2017, 71, 1020–1021. [Google Scholar] [CrossRef]
  92. Naureen, Z.; Dhuli, K.; Donato, K.; Aquilanti, B.; Velluti, V.; Matera, G.; Iaconelli, A.; Bertelli, M. Foods of the Mediterranean diet: Tomato, olives, chili pepper, wheat flour and wheat germ. J. Prev. Med. Hyg. 2022, 63 (Suppl. 3), E4–E11. [Google Scholar]
  93. Naureen, Z.; Bonetti, G.; Medori, M.C.; Aquilanti, B.; Velluti, V.; Matera, G.; Iaconelli, A.; Bertelli, M. Foods of the Mediterranean diet: Garlic and Mediterranean legumes. J. Prev. Med. Hyg. 2022, 63 (Suppl. 3), E12–E20. [Google Scholar]
  94. Koelman, L.; Egea Rodrigues, C.; Aleksandrova, K. Effects of Dietary Patterns on Biomarkers of Inflammation and Immune Responses: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Adv. Nutr. 2022, 13, 101–115. [Google Scholar] [CrossRef]
  95. Millar, S.R.; Navarro, P.; Harrington, J.M.; Shivappa, N.; Hébert, J.R.; Perry, I.J.; Phillips, C.M. Dietary score associations with markers of chronic low-grade inflammation: A cross-sectional comparative analysis of a middle- to older-aged population. Eur. J. Nutr. 2022, 61, 3377–3390. [Google Scholar] [CrossRef]
  96. Ryan, M.C.; Itsiopoulos, C.; Thodis, T.; Ward, G.; Trost, N.; Hofferberth, S.; O’Dea, K.; Desmond, P.V.; Johnson, N.A.; Wilson, A.M. The Mediterranean diet improves hepatic steatosis and insulin sensitivity in individuals with non-alcoholic fatty liver disease. J. Hepatol. 2013, 59, 138–143. [Google Scholar] [CrossRef]
  97. Mirabelli, M.; Chiefari, E.; Arcidiacono, B.; Corigliano, D.M.; Brunetti, F.S.; Maggisano, V.; Russo, D.; Foti, D.P.; Brunetti, A. Mediterranean Diet Nutrients to Turn the Tide against Insulin Resistance and Related Diseases. Nutrients 2020, 12, 1066. [Google Scholar] [CrossRef] [Green Version]
  98. Augimeri, G.; Galluccio, A.; Caparello, G.; Avolio, E.; La Russa, D.; De Rose, D.; Morelli, C.; Barone, I.; Catalano, S.; Andò, S.; et al. Potential Antioxidant and An-ti-Inflammatory Properties of Serum from Healthy Adolescents with Optimal Mediterra-nean Diet Adherence: Findings from DIMENU Cross-Sectional Study. Antioxidants 2021, 10, 1172. [Google Scholar] [CrossRef]
  99. Mentella, M.C.; Scaldaferri, F.; Ricci, C.; Gasbarrini, A.; Miggiano, G.A.D. Cancer and Mediterranean Diet: A Review. Nutrients 2019, 11, 2059. [Google Scholar] [CrossRef] [Green Version]
  100. La Russa, D.; Marrone, A.; Mandalà, M.; Macirella, R.; Pellegrino, D. Antioxidant/Anti-Inflammatory Effects of Caloric Restriction in an Aged and Obese Rat Model: The Role of Adiponectin. Biomedicines 2020, 8, 532. [Google Scholar] [CrossRef]
  101. Martin, B.; Mattson, M.P.; Maudsley, S. Caloric restriction and intermittent fasting: Two potential diets for successful brain aging. Ageing Res. Rev. 2006, 5, 332–353. [Google Scholar] [CrossRef] [Green Version]
  102. Colleluori, G.; Villareal, D.T. Weight strategy in older adults with obesity: Calorie restriction or not? Curr. Opin. Clin. Nutr. Metab. Care 2023, 26, 17–22. [Google Scholar] [CrossRef]
  103. Brandhorst, S.; Longo, V.D. Fasting and Caloric Restriction in Cancer Prevention and Treatment. Recent Results Cancer Res. 2016, 207, 241–266. [Google Scholar]
  104. Kopelovich, L.; Fay, J.R.; Sigman, C.C.; Crowell, J.A. The mammalian target of rapamycin pathway as a potential target for cancer chemoprevention. Cancer Epidemiol. Biomark. Prev. 2007, 16, 1330–1340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Salvadori, G.; Mirisola, M.G.; Longo, V.D. Intermittent and Periodic Fasting, Hormones, and Cancer Prevention. Cancers 2021, 13, 4587. [Google Scholar] [CrossRef] [PubMed]
  106. O’Flanagan, C.H.; Smith, L.A.; McDonell, S.B.; Hursting, S.D. When less may be more: Calorie restriction and response to cancer therapy. BMC Med. 2017, 15, 106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Peisch, S.F.; Van Blarigan, E.L.; Chan, J.M.; Stampfer, M.J.; Kenfield, S.A. Prostate cancer progression and mortality: A review of diet and lifestyle factors. World J. Urol. 2017, 35, 867–874. [Google Scholar] [CrossRef] [Green Version]
  108. Nyrop, K.A.; Deal, A.M.; Williams, G.R.; Guerard, E.J.; Pergolotti, M.; Muss, H.B. Physical activity communication between oncology providers and patients with early-stage breast, colon, or prostate cancer. Cancer 2016, 122, 470–476. [Google Scholar] [CrossRef] [Green Version]
  109. Lynch, B.M.; Neilson, H.K.; Friedenreich, C.M. Physical activity and breast cancer prevention. Recent Results Cancer Res. 2011, 186, 13–42. [Google Scholar]
  110. Mugele, H.; Freitag, N.; Wilhelmi, J.; Yang, Y.; Cheng, S.; Bloch, W.; Schumann, M. High-intensity interval training in the therapy and aftercare of cancer patients: A systematic review with meta-analysis. J. Cancer Surviv. 2019, 13, 205–223. [Google Scholar] [CrossRef]
  111. Rock, C.L.; Thomson, C.; Gansler, T.; Gapstur, S.M.; McCullough, M.L.; Patel, A.V.; Andrews, K.S.; Bandera, E.V.; Spees, C.K.; Robien, K.; et al. American Cancer Society guideline for diet and physical activity for cancer prevention. CA Cancer J. Clin. 2020, 70, 245–271. [Google Scholar] [CrossRef]
  112. Djuricic, I.; Calder, P.C. Beneficial Outcomes of Omega-6 and Omega-3 Polyunsaturated Fatty Acids on Human Health: An Update for 2021. Nutrients 2021, 13, 2421. [Google Scholar] [CrossRef]
  113. Fekete, M.; Szarvas, Z.; Fazekas-Pongor, V.; Feher, A.; Csipo, T.; Forrai, J.; Dosa, N.; Peterfi, A.; Lehoczki, A.; Tarantini, S.; et al. Nutrition Strategies Promoting Healthy Aging: From Improvement of Cardiovascular and Brain Health to Prevention of Age-Associated Diseases. Nutrients 2022, 15, 47. [Google Scholar] [CrossRef]
  114. Lorenzo, P.M.; Izquierdo, A.G.; Rodriguez-Carnero, G.; Fernández-Pombo, A.; Iglesias, A.; Carreira, M.C.; Tejera, C.; Bellido, D.; Martinez-Olmos, M.A.; Leis, R.; et al. Epigenetic Effects of Healthy Foods and Lifestyle Habits from the Southern European Atlantic Diet Pattern: A Narrative Review. Adv. Nutr. 2022, 13, 1725–1747. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the consequences due to a sedentary lifestyle.
Figure 1. Schematic representation of the consequences due to a sedentary lifestyle.
Nutrients 15 01667 g001
Figure 2. The typical Mediterranean diet and lifestyle raccomandations to follow: Mediterranean Lifestyle.
Figure 2. The typical Mediterranean diet and lifestyle raccomandations to follow: Mediterranean Lifestyle.
Nutrients 15 01667 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Divella, R.; Marino, G.; Infusino, S.; Lanotte, L.; Gadaleta-Caldarola, G.; Gadaleta-Caldarola, G. The Mediterranean Lifestyle to Contrast Low-Grade Inflammation Behavior in Cancer. Nutrients 2023, 15, 1667. https://doi.org/10.3390/nu15071667

AMA Style

Divella R, Marino G, Infusino S, Lanotte L, Gadaleta-Caldarola G, Gadaleta-Caldarola G. The Mediterranean Lifestyle to Contrast Low-Grade Inflammation Behavior in Cancer. Nutrients. 2023; 15(7):1667. https://doi.org/10.3390/nu15071667

Chicago/Turabian Style

Divella, Rosa, Graziella Marino, Stefania Infusino, Laura Lanotte, Gaia Gadaleta-Caldarola, and Gennaro Gadaleta-Caldarola. 2023. "The Mediterranean Lifestyle to Contrast Low-Grade Inflammation Behavior in Cancer" Nutrients 15, no. 7: 1667. https://doi.org/10.3390/nu15071667

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

Article Metrics

Back to TopTop