Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 6748 2023-02-01 02:10:20 |
2 format correct -848 word(s) 5900 2023-02-01 02:29:47 | |
3 format correct Meta information modification 5900 2023-02-01 02:31:06 | |
4 format correct Meta information modification 5900 2023-02-01 02:32:23 | |
5 format correct -11 word(s) 5889 2023-02-01 02:38:44 | |
6 Figures 1-5 were created with BioRender.com. + 89 word(s) 5978 2023-02-01 09:52:54 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
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. Encyclopedia. Available online: https://encyclopedia.pub/entry/40694 (accessed on 01 May 2024).
García-Chico C,  López-Ortiz S,  Peñín-Grandes S,  Pinto-Fraga J,  Valenzuela PL,  Emanuele E, et al. Physical Exercise and the Hallmarks of Breast Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/40694. Accessed May 01, 2024.
García-Chico, Celia, Susana López-Ortiz, Saúl Peñín-Grandes, José Pinto-Fraga, Pedro L. Valenzuela, Enzo Emanuele, Claudia Ceci, Grazia Graziani, Carmen Fiuza-Luces, Simone Lista, et al. "Physical Exercise and the Hallmarks of Breast Cancer" Encyclopedia, https://encyclopedia.pub/entry/40694 (accessed May 01, 2024).
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.,  Lucia, A., & Santos-Lozano, A. (2023, February 01). Physical Exercise and the Hallmarks of Breast Cancer. In Encyclopedia. https://encyclopedia.pub/entry/40694
García-Chico, Celia, et al. "Physical Exercise and the Hallmarks of Breast Cancer." Encyclopedia. Web. 01 February, 2023.
Physical Exercise and the Hallmarks of Breast Cancer
Edit

Growing evidence suggests that, among the different molecular/cellular pathophysiological mechanisms associated with cancer, there are 14 hallmarks that play a major role, including: (i) sustaining proliferative signaling, (ii) evading growth suppressors, (iii) activating invasion and metastasis, (iv) enabling replicative immortality, (v) inducing angiogenesis, (vi) resisting cell death, (vii) reprogramming energy metabolism, (viii) evading immune destruction, (ix) genome instability and mutations, (x) tumor-promoting inflammation, (xi) unlocking phenotypic plasticity, (xii) nonmutational epigenetic reprogramming, (xiii) polymorphic microbiomes, and (xiv) senescent cells. These hallmarks are also associated with the development of breast cancer, which represents the most prevalent tumor type in the world. For the first time, the effects of physical activity/exercise on these hallmarks are described. In summary, an active lifestyle, and particularly regular physical exercise, provides beneficial effects on all major hallmarks associated with breast cancer, and might therefore help to counteract the progression of the disease or its associated burden.

breast cancer training physical activity mammary cancer biomarkers
The factors promoting tumor proliferation, designated as “cancer hallmarks”, have been previously described in detail [1][2][3] and include: (i) sustaining proliferative signaling, (ii) evading growth suppressors, (iii) activating invasion and metastasis, (iv) enabling replicative immortality, (v) inducing angiogenesis, (vi) resisting cell death, (vii) reprogramming energy metabolism, (viii) evading immune destruction, (ix) genome instability and mutation, (x) tumor-promoting inflammation, (xi) unlocking phenotypic plasticity, (xii) nonmutational epigenetic reprogramming, (xiii) polymorphic microbiomes, and (xiv) senescent cells [1][2][3].
Although the underlying causes of breast cancer remain unknown [4], there is growing evidence highlighting the potential impact of certain risk factors, both extrinsic and intrinsic, on its pathogenesis. For instance, physical activity is related to reduced breast cancer mortality and recurrence in breast cancer patients [5][6], as well as fewer/less severe adverse effects following its treatment [7]
The research group has previously summarized the potential beneficial effects of exercise training on cancer hallmarks [8]. In addition, a systematic review summarized the effects of physical exercise on certain breast cancer biomarkers [9]. However, no review has previously explored how exercise specifically affects breast cancer hallmarks. Therefore, the effects of physical exercise on cancer hallmarks are summarized, after their specific contextualization in the field of breast cancer according to both preclinical and clinical studies.

2. The Effects of Physical Exercise on the Hallmarks Associated with Breast Cancer

2.1. Sustaining Proliferative Signaling

As their primary incontrovertible feature, cancer cells are able to modify the signals that regulate cell growth and division [2]
Although a large amount of evidence comes from animal models [10][11][12][13][14][15], data are also available from clinical evaluation of the effects of physical exercise and physical activity on circulating serum markers related to cancer cell proliferation [16].
In detail, preclinical data from male mice show that anaerobic exercise training for eight weeks (four sessions per week) reduces both tumor weight and cell proliferation ex vivo when compared to a sedentary group [15]. Likewise, aerobic exercise seems to affect certain variables associated with cell proliferation in a transgenic mouse model of breast cancer and N-methyl-N-nitrosourea (MNU)-induced mammary carcinogenesis in female rats, including the reduction in tumor size [10], the number of developed tumors [11], or the expression of proliferation markers, such as the Ki-67 nuclear protein [12]. However, there are discrepancies regarding the effect of exercise in the expression of Ki-67, since in some cases its expression is not significantly different between exercised and not exercised animals or is not paralleled by changes in training intensity [10][11]. Aerobic exercise also reduces the expression of both estrogen and progesterone receptors in animal models, thus further acting as a protective factor against tumor proliferation and development [14].
Another mechanism promoting cell growth and proliferation lies in the binding of insulin-like growth factor-1 (IGF-1) to the insulin-like growth factor receptor type 1 (IGF-1R). Indeed, high levels of IGF-1R are associated with a worse disease prognosis in breast cancer patients, as assessed through a computational analysis of cancer genomics information available in public databases [17]. Various pathways activate the aforementioned process, thus stimulating tumor survival [17]. In the only available clinical studies evaluating tumor proliferative signaling markers, combined training, based on resistance plus aerobic exercise, reduces serum IGF-1, estradiol, and insulin levels [18] in breast cancer survivors who are overweight or obese [16]. Additionally, moderate-intensity aerobic exercise decreases both insulin and IGF-1 levels [19].
Another cell-cycle specific antigen and proliferation marker in breast cancer is proliferating cell nuclear antigen (PCNA), having an important role in DNA replication [20]. Exposure of human breast cancer cell lines to an exercise-conditioned medium obtained from contracted myotubules, with or without doxorubicin, reveals a reduction in PCNA expression and a synergistic relationship with the anticancer drug [21] (Figure 1).
Figure 1. (a) Schematic drawing of cancer hallmarks, highlighting markers related to sustaining proliferative signaling, evading growth suppressors, resisting cell death, genomic instability, and senescent cells. (b) The effects of physical exercise on sustaining proliferative signaling, evading growth suppressors, resisting cell death, genomic instability, and senescent cells. Abbreviations: Akt: protein kinase b; BRCA1: breast cancer type 1; BRCA2: breast cancer type 2; EGFR: epidermal growth factor receptor; ER: estrogen receptor; Erk: extracellular signal regulated kinase; HIIT, high-intensity interval training; IGF-1: insulin-like growth factor 1; IGF-1R: insulin-like growth factor receptor type 1; IGFBP-3: insulin-like growth factor binding protein-3; miRNA: microRNA; MEK: mitogen activated protein kinase; mTOR: mammalian target of rapamycin; OSM: oncostatin M; PCNA: proliferating cell nuclear antigen; PI3K: phosphatidylinositol-3-kinase; PR: progesterone receptor; PTEN: phosphatase and tensin homolog; Rb: retinoblastoma; SASP: senescence-associated secretory phenotype; TP53: tumor protein 53. Figure 1 was created with BioRender.com.

2.2. Evading Growth Suppressors

Besides preserving cell proliferation, tumor cells are able to evade the mechanisms that, under physiological conditions, regulate cell growth and proliferation [2].
Aerobic exercise increases the TP53 gene levels in female mice with triple-negative breast cancer (TNBC), despite no relative change in phosphatase and tensin homolog (PTEN) expression [22], and reduces the expression of hyperphosphorylated retinoblastoma (Rb) protein (inactive form), in rats with mammary adenocarcinoma [23]. A combination of aerobic and resistance exercise increases the serum levels of insulin-like growth factor binding protein-3 (IGBP-3) in overweight or obese breast cancer survivors [16]. Moreover, exercise influences the expression of some miRNAs that act as tumor suppressors. Preclinical and clinical evidence shows that acute and chronic exercise interventions raise the expression of miR-133, which alters cancer progression by acting on oncogenes IGF-1R and epidermal growth factor receptor (EGFR) [24]. Additionally, high-intensity interval training (HIIT) upregulates the expression of miR-206 and miR-143 in women with breast cancer [25]. Evidence on the role of myokines, i.e., cytokines, peptides, or growth factors produced and released by skeletal muscle cells under contraction, is still limited in breast cancer. Available research shows that among myokines, aerobic exercise releases oncostatin M (OSM), thus resulting in lower tumor volume in breast cancer mice [26]. Breast cancer patients show significantly decreased serum irisin concentrations [27]; hence, it is reasonable to assume that physical exercise, by stimulating the production of irisin, activates the suppression of cell growth, as in other types of cancer (i.e., prostate cancer and glioblastoma) [24]. Moreover, irisin significantly decreases cell number, migration, and viability of malignant MDA-MB-231 breast epithelial cells in vitro, without affecting nonmalignant MCF-10a cells, and enhances the cytotoxic effects of doxorubicin [28].
Finally, exercise produces epinephrine and norepinephrine that activate the Hippo pathway, thus inhibiting breast cancer cell growth and the mTOR pathway [29].
In general, existing studies largely support the claim that physical exercise activates tumor suppressors and inactivates tumor promoters by hampering the establishment of a favorable microenvironment for cancer progression (Figure 1).

2.3. Activating Invasion and Metastasis

Tumor complexity is related to its microenvironment (tumor microenvironment, TME), the composition of which differs among tumor types and breast cancer subtypes [30][31]. TME comprises the proliferating tumor cells along with a range of noncancerous cells (i.e., stromal cells) incorporated in the tumor mass [30]. The noncancerous component of the TME includes immune cells, blood vessels, the extracellular matrix, and stromal cells [31]. Macrophages are stromal cells in TME, wherein they differentiate to become tumor-associated macrophages (TAMs). TAMs can polarize into two phenotypes, namely M1 (showing antitumor effects) and M2 (supporting tumor growth, local invasion, metastasis, and exerting an immune-suppressive function) [32]. In this context, TME affects angiogenesis, proliferation, invasion, and metastasis through the release of growth factors and cytokines [31].
Some cytokines and adipokines, such as leptin, the previously mentioned irisin (also defined as an adipo-myokine), and OSM, as well as resistin, are involved in metastasis and invasion in breast cancer [33]
Chronic adaptations to moderate-intensity aerobic exercise seem to prevent the activation of a metastatic cascade and the resulting formation of cancer metastasis [34] by controlling angiogenesis, eliminating circulating tumor cells, and reducing endothelial cells’ permeability, while high-intensity physical exercise seems to favor cancer spreading, likely due to disproportionate stress.
Studies on animal models do not report significant results: in a rat model of breast cancer, moderate-intensity exercise training tends to protect from pulmonary metastases, but the small sample evaluated does not allow to reach statistical significance [35]. Conversely, a preclinical study conducted in mice show that four weeks of wheel running after breast cancer cell injection is associated with a higher number of pulmonary metastases compared to sedentary controls, as well as with a lower nitric oxide production [36]. By contrast, a pilot clinical trial in breast cancer patients shows that aerobic exercise training for 12 weeks combined with neoadjuvant chemotherapy promotes higher nitric oxide production and lower tumor invasiveness [37].
In any case, physical exercise helps TAMs polarize to an M1 phenotype, enhancing their antitumor effects. Additionally, moderate-to-high intensity exercise decreases the macrophages’ recruitment to the TME of breast cancer [32]. In contrast, sedentary behaviors result in M2 phenotype polarization, thus promoting tumor growth, invasion, and metastasis [32]. Physical exercise induces the release of myokines related to tumor invasion and metastasis, such as irisin and OSM. As mentioned above, irisin levels are decreased in breast cancer patients [27][38], but physical exercise can improve irisin serum levels to prevent spinal metastasis [39]. In this view, physical exercise induces the release of antimetastatic adipokines (e.g., adiponectin) [40] and, consequently, the downregulation of prometastatic adipokines, such as leptin [41] and tumor necrosis factor-α (TNF-α) [42]. Exercise also upregulates myomiR-133a, i.e., a member of a subset of miRNAs specific to striated muscle and expressed at higher levels in skeletal muscle [24], which influences myoblast differentiation and contributes to the suppression of several tumors, including breast cancer, by controlling tumor invasiveness and metastasis [24][43].
An acute bout of aerobic exercise stabilizes hypoxia-inducible factor 1-α (HIF-1α) after a period of regular endurance training through the expression of negative regulators, as a mechanism of adaptation of skeletal muscle; this long-term endurance exercise downregulation of HIF-1α response [44] could possibly decrease epithelial-to-mesenchymal transition (EMT) promoted by this protein.
Chemokine (C-X-C motif) receptor type 4 (CXCR4) and its ligand, chemokine (C-X-C motif) ligand type 12 (CXCL12), are relatively highly expressed in invasive breast cancer cells, homing them to distant lymph nodes, the lungs, and the liver [45]. In contrast to the previously discussed results, CXCR4 gene expression is increased in a transgenic mouse model of breast cancer with access to a wheel, compared with nonrunners, although pulmonary metastatic foci and the proliferative index, as indicated by morphometric analyses and measurement of Ki-67 levels, respectively, show no differences between runners and nonrunners, and no difference has been observed in the expression of CXCL12 gene [10] (Figure 2).
Figure 2. (a) Schematic drawing of cancer hallmarks, highlighting markers related to activating invasion and metastasis, inducing angiogenesis, tumor-promoting inflammation, and unlocking phenotypic plasticity. (b) The effects of physical exercise on activating invasion and metastasis, inducing angiogenesis, tumor-promoting inflammation, and unlocking phenotypic plasticity. Abbreviations: CEP: endothelial progenitor cells; EMT: epithelial to mesenchymal transition; HIF-1: hypoxia-inducible factor-1; HIF-1α: hypoxia-inducible factor-1α; HIIT: high-intensity interval training; IL-1β: interleukin-1β; IL-2: interleukin-2; Il-4: interleukin-4; IL-6: interleukin-6; IL-8: interleukin-8; IL-10: interleukin-10; MCP-1: monocyte chemoattractant protein-1; miR21: microRNA 21; OSM: oncostatin M; TGF-β: transforming growth factor-β; TNF-α: tumor necrosis factor-α; VEGF: vascular endothelial growth factor; VEGF-A: vascular endothelial growth factor-A. Figure 2 was created with BioRender.com. Templates used in Figure 2: adapted from "Tumor Cell Metastasis”, by BioRender.com (2022); adapted from "Blood Vessel (Straight, Light Background)”, by BioRender.com (2022). Retrieved from https://app.biorender.com/biorender-templates.

2.4. Enabling Replicative Immortality

Cancer cells acquire endless replicative immortality to develop macroscopic tumors [2] via the activation of telomerase or a DNA homologous recombination-based mechanism (alternative lengthening of telomeres, ALT) [46]. Telomere shortening following each cell division is related to genomic instability, oxidative stress, and inflammation [47]. Telomerase activation restores genomic stability and enhances tumor progression by preserving the telomeric length despite cell division [8].
Physical exercise affects cancer development and its dependence on genetic alterations of chromosomes, DNA methylation, miRNA expression, and changes in chromatin structure [48][49]. Furthermore, exercise attenuates telomere attrition and helps maintain a balance with oxidative stress and inflammatory status [47]—factors inversely correlated with telomere length.
Related to the role of miRNAs, both acute and chronic exercise interventions increase the release of the senescence-associated myomiR-133 into circulation [24], thus supporting the inhibition of telomerase expression [50][51].
The evidence that telomere lengthening decreases mortality in women with breast cancer [52] further supports the hypothesis that regular physical exercise represents a potential protective factor against this tumor hallmark (Figure 3).
Figure 3. (a) Schematic drawing of cancer hallmarks, highlighting markers related to enabling replicative immortality, reprogramming energy metabolism, and nonmutational epigenetic reprogramming. (b) The effects of physical exercise on enabling replicative immortality, reprogramming energy metabolism, and nonmutational epigenetic reprogramming. ALT: alternative lengthening of telomeres; APC: adenomatous polyposis coli; CO2: carbon dioxide; CCND2: cyclin D2; HIN1: high in normal-1; L3MBTL1: lethal(3) malignant brain tumor-like 1; LDH-A: lactate dehydrogenase-A; LDH-B: lactate dehydrogenase-B; O2: oxygen; ROS: reactive oxygen species; TET: ten eleven translocation proteins; TWIST1: twist family bHLH transcription factor 1. Figure 3 was created with BioRender.com. Templates used in Figure 3: adapted from "Telomere Biology in Cancer Cells”, by BioRender.com (2022). Retrieved from https://app.biorender.com/biorender-templates.

2.5. Inducing Angiogenesis

The formation of a hypoxic microenvironment is a well-known event in the development of mutagenesis and cancer [53]. Hypoxia maintains stem cells in an undifferentiated state, enabling only cancer cells to differentiate and accumulate genetic and epigenetic alterations uninterrupted [53]. As the tumor mass increases in size, the oxygen availability decreases. In response to hypoxia, the development of new blood vessels from the pre-existing vasculature, i.e., angiogenesis, occurs to obtain the nutrients and oxygen required for massive cell growth, as well as to promote malignant cells entering the systemic circulation, thus resulting in metastasis [54].
Preclinical research in an orthotopic murine model of breast cancer shows that exercise promotes tumor vessel maturity and microvessel density [55], supported by reduction in intratumoral hypoxia and improvement in tissue perfusion, oxygen supply [56], and nitric oxide bioavailability [57], thus leading to an unfavorable TME for cancer progression and to a better drug delivery [58]. Indeed, since neovessels constitute the major route for cancer cells’ proliferation and dissemination, a process of vasculature “normalization”, through which abnormal vessels are remodeled, can be beneficial in terms of a better response to anticancer treatment [58].
Another preclinical study conducted in mice suggests that performing treadmill endurance exercise decreases VEGF-A expression, as assessed through Enzyme-linked immunosorbent assay (ELISA) from tissue samples [59], while a rat model of a mammary tumor confirms an increased VEGF-A expression, detected immunohistochemically, and tumor vascularization after long-term training [60]. In this context, a clinical study reports that an increase in the total physical exercise time and weight loss leads to a decreased circulating VEGF-A expression in overweight women with breast cancer [61] (Figure 2). However, a combined training (i.e., aerobic and resistance training) intervention, following surgery, radiotherapy, or chemotherapy does not substantiate these results [62], since serum mice levels of VEGF-A are not significantly different between exercised and control groups.

2.6. Resisting Cell Death

Cancer cells acquire a characteristic resistance to apoptosis, which prevents them from dying and sustains uncontrolled proliferation [63]. The most relevant programmed cell death pathways described so far are: (i) the extrinsic (i.e., death receptor based), which receives and processes extracellular pro-death signals, and (ii) the intrinsic (i.e., mitochondrial), which senses and integrates intracellularly originated signals [2].
Aerobic physical exercise induces apoptosis, as demonstrated in breast cancer murine models [55][64], in terms of increased expression of Bax and caspase-3 [64]; anaerobic training increases the rate of tumor cells’ apoptosis in male rats (two folds higher compared to sedentary rats, paralleled by an increase of Bax and reduction in Bcl-2 expression) [15]. High-intensity aerobic exercise is also associated with a higher expression of Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cancer cells, a biomarker of apoptotic signaling, in a chemically induced rat model of breast cancer [11]. However, decreased levels of anti-apoptotic regulators (Bcl-2 and Bcl-Xl), increased expression of the pro-apoptotic factor Bax, and caspase-3 activation are preclinically evident even when low-intensity aerobic exercise is performed [13]. Thus, even though only preclinical investigations are available, exercise seems to induce manifest improvements in apoptosis and reduces breast cancer resistance to programmed cell death, without any correlation to difficulty and energy expenditure (Figure 1).

2.7. Reprogramming Energy Metabolism

Cancer cells are able to modify their metabolism by using glycolysis even under aerobic conditions (i.e., in the presence of oxygen and fully functioning mitochondria), a phenomenon known as the “Warburg effect” [65]. This process allows cancer cells to survive in situations of fluctuating oxygen supply and results in high lactate generation [66]. Moreover, the altered glucose metabolism, in terms of increased fermentation to lactate, is responsible for the typical acidic pH of the TME [65].
Energy consumption induced by exercise significantly affects whole body and intracellular metabolism [67], potentially contributing to alterations into glucose synthesis and utilization.
In an experimental TNBC murine model involving sedentary or trained for 12 weeks animals, physical exercise correlates with a smaller tumor mass, mitochondria with a lower respiratory rate in the state of maximum electron transport capacity, and modulated macronutrient oxidation, almost exclusively represented by carbohydrate oxidation, while the sedentary condition metabolizes both carbohydrates and lipids [22]. In another preclinical study, mice with breast cancer undergoing aerobic physical training show a decreased type 1 monocarboxylate transporter expression and a shift in lactate dehydrogenase (LDH) isoforms (higher LDH-B and lower LDH-A expression), compared to the control group (Figure 3). These changes are associated with a lower concentration of accumulated lactate, together with a reduced tumor mass [68]. Of note, data exist demonstrating that the hypoxic TME leads to increased LDH-A expression, the silencing of which suppresses the tumorigenicity of breast cancer [69], whereas increased lactate production correlates with LDH-B downregulation [70] (Figure 3).
Altogether, the observed metabolic changes due to the performance of physical exercise seem to maintain glucose homeostasis in face of the required energy expenditure. Therefore, exercise could help in the regulation of cellular metabolism; however, the included studies are insufficient to draw conclusions on this matter.

2.8. Evading Immune Destruction

Tumor cells have the ability to evade the immune system mediated destruction through several mechanisms [71]. Finally, malignant cells may actively suppress the immune response by expressing immune inhibitory ligands or receptors (“immune checkpoints”) or by inducing apoptosis of certain antitumor lymphocytes. Referring to these immune checkpoints, the binding of the programmed death-ligand 1 (PDL-1) with its programmed death-1 (PD-1) receptor provides a suppressive signal to T lymphocytes with a decrease in the immune response [72]. Moreover, tumor cells are able to induce a TME supporting their proliferation and growth, enriched in immunosuppressive cells and soluble factors. Specifically, myeloid-derived suppressor cells (MDSC) are able to disrupt the binding of major histocompatibility complex (MHC) to CD8+ T cells and induce nitration of MHC class I molecules on cancer cells, making tumors resistant to the immune system [71].
In light of the abovementioned issues, the benefits of physical activity in improving the efficacy of the anticancer immune response may be particularly relevant in breast cancer patients in terms of survival, recurrence, and mortality. In fact, moderate physical activity recovers the immune function by increasing the number and activity of certain cells belonging to the immune system, such as neutrophils, monocytes, eosinophils, and lymphocytes [73].
Particularly, aerobic exercise is acknowledged to significantly improve NK cell activation in treadmill-trained female rats, decrease the accumulation of myeloid-derived suppressor cells (MDSC) in the TME, as well as promote an inverse correlation between MDSC and CD8+ T cells [74]. Furthermore, aerobic exercise performed by female breast cancer survivors can enhance NK cell cytotoxic activity and thymidine uptake [75]. Concerning resistance training, it appears to help female breast cancer survivors to decrease TNF-α production by their NK and natural killer T (NKT) cells [76]. In addition, when aerobic exercise is compared to muscular resistance exercise, the latter generates greater benefits: women with primary breast cancer undergoing chemotherapy who engage in muscular resistance exercise show a lower decrease in γδ T cells and CD8+ T cells than those who engage in endurance exercise [77]. However, a recent review highlights that there is still not enough evidence to support the specific effects of exercise on NK cells [78]: in breast cancer patients undergoing surgery, chemotherapy, or radiation therapy and performing moderate aerobic performance, the cytotoxic activity of NK cells is not significantly affected [79].
Lastly, combined training (i.e., aerobic and resistance) shows favorable results for women with breast cancer, as it increases the percentages of CD4+ and CD69+ T lymphocytes [80] (Figure 4). Nevertheless, in another trial, the combined exercise, together with diet intervention, does not show favorable results supporting the experimental group [81]. Therefore, the outcomes related to this hallmark in breast cancer patients seem to be heterogeneous, and further research is required to elucidate whether regular exercise can improve immune function in cancer survivors.
Figure 4. (a) Schematic drawing of cancer hallmarks, highlighting markers related to evading immune destruction and polymorphic microbiomes. (b) The effects of physical exercise on evading immune destruction and polymorphic microbiomes. Abbreviations: AM: akkermansia muciniphilia; F.nucleatum: Fusobacterium nucleatum; MDSC: myeloid-derived suppressor cells; MHC: major histocompatibility complex; NK: natural killer; NKT: natural killer T; PD-1: programmed death-1; PDL-1: programmed death-ligand 1; PI-9: proteinase inhibitor-9; TNF-α: Tumor Necrosis Factor-α. Figure 4 was created with BioRender.com. Templates used in Figure 4:" Gut-Brain-Axis, by BioRender.com (2022). Retrieved from https://app.biorender.com/biorender-templates.

2.9. Genomic Instability and Mutation

Genomic instability designates the accumulation of mutations in a genome’s stabilizing genes [82], with consequent increase in the rate at which cells acquire genomic alterations. It is considered a necessary process for carcinogenesis to begin and progress [83].
As previously stated, physical activity is associated with a reduction of the risk of suffering from breast cancer and, in breast cancer patients, with a reduced tendency to progress toward the more aggressive and invasive form of the disease [84]; such benefits also seem to include women with high familial and genetic risk factors [85]. Indeed, many studies are exploring the benefits of physical exercise in individuals carrying the hereditary BRCA gene mutations. BRCA1 and BRCA2 proteins contribute to preserving the integrity of the genome, since they participate in the cellular response to DNA damage [86]. A pilot study reports how high-intensity interval exercise, combined with muscular resistance, increases the serum concentration of BRCA1 protein following a six-week intervention period in BRCA gene mutation carriers, and improves the systemic antioxidative status [87] (Figure 1). Therefore, the preservation of a healthy body weight and an active lifestyle in women carrying BRCA gene mutations could prevent disease development; however, further studies are needed to demonstrate this correlation [86].
Despite the outcomes related to BRCA gene mutations, no other experimental results are currently available that relate the practice of physical exercise to genomic instability in breast cancer patients. Therefore, at present, it is problematic to draw conclusions on this hallmark; future research is needed to analyze the potential benefits of exercise in this field and to standardize the best existing treatment options.

2.10. Tumor-Promoting Inflammation

Inflammation is a defense mechanism of the body that, physiologically, originates in response to trauma and/or infection and subsides once the problem is resolved. However, in the case of cancer, the process becomes chronic, due in part to an excessive production of cytokines and growth factors [88]. In breast cancer evolution, low-grade chronic inflammation is crucial [89].
Certain studies have investigated how muscle contraction can inhibit the activity of pro-inflammatory substances in distant or proximal muscular regions, with several authors aiming to address how physical activity modifies the inflammatory environment in breast cancer patients [90]. In particular, the effects of aerobic exercise are extensively explored.
In preclinical studies, female mice who trained on a treadmill show decreased plasma concentrations of the pro-inflammatory factors interleukin-6 (IL-6) and monocyte chemoattractant protein-1 (MCP-1) [91]. Furthermore, aerobic exercise in combination with tamoxifen and/or letrozole reduces the expression of TNF-α and increases the anti-inflammatory interleukin-10 (IL-10) serum concentrations [92].
Concerning aerobic exercise carried out in clinical studies, although some analyses report no significant benefits related to inflammation [75][93], other research stresses that this form of training, along with diet, helps decrease TNF-α and IL-6 serum concentrations in women with breast cancer [61]. In addition, the increase in total exercise time is associated with greater reduction in IL-6 serum levels [61]. Likewise, exercise is related to an increase in interleukin-8 (IL-8) and decrease in interleukin-2 (IL-2) and interleukin-1β (IL-1β) plasma concentrations [37]. Additionally, the implementation of HIIT seems to be effective in reducing the plasma and serum levels of various pro-inflammatory markers (TNF-α, IL-6) [94][95] and in increasing the expression of interleukin-4 (IL-4), IL-10, and adiponectin [94], all attenuating inflammatory responses. Regarding IL-6, the interpretation of clinical results is still debated: this cytokine might present an increasing tendency toward plasma accumulation [96] throughout the disease progression that, according to the results of two randomized controlled trials [97][98], would be exacerbated in the absence of exercise. At the same time, it seems that, as physical activity increases, systemic levels of IL-6 also grow.
In the case of resistance exercise, studies do not report secondary improvements or significant benefits in breast cancer survivors [76]. Nevertheless, there is clinical evidence showing that nontraining women present higher serum concentrations of MCP-1, compared to exercising patients [62].
Regarding combined exercise of aerobic and muscular resistance, it has been supposed that, over time, repeated bouts of exercise result in a higher steady-state level of serum IL-6 that contributes to a reduction in chronic inflammation, thanks to the possible anti-inflammatory properties of this complex cytokine [99]. Likewise, a decrease in leptin, considered a pro-inflammatory adipokine, has been observed in women with breast cancer following a combined training [99] (Figure 2). Several studies claim that this type of training is able to affect markers such as IL-6, TNF-α, leptin, IL-8, and C-reactive protein (CRP), decreasing their plasma levels as well as increasing adiponectin [100]. However, other investigations report that this type of exercise program does not generate significant benefits [80][81] and future research should be conducted to analyze its effects on inflammatory markers in breast cancer patients.

2.11. Unlocking Phenotypic Plasticity

Cell genetic and epigenetic traits are not fixed, but dependent on the activation of microenvironmental stimuli and alterations in gene expression. Such activation allows breast cancer cells to switch between different phenotypic states, a feature referred to as cell plasticity [101]. Tumor-induced hypoxia reduces the levels of ten-eleven translocation (TET) demethylases, leading to hypermethylation [3]. Publications addressing the phenotype variability of several tumor types and the effect of exercise on these different phenotypes are presently limited. However, systemic acidosis induced by physical exercise could affect signals from the TME and prevent tumor transition from a noninvasive to an invasive phenotype [102].
An explanation for understanding the mechanism by which exercise could modulate the phenotypic variability of breast cancer and surrounding cells, such as myocytes of the pectoralis major or minor, is currently missing [103]. However, it is known that each muscle fiber contains a subset of satellite, or progenitor, cells that remain quiescent unless activated. Resistance exercise may trigger a release of inflammatory substances and growth factors, thereby stimulating satellite cells and promoting their entry into the cell cycle [103]. This stimulation, a priori beneficial in healthy individuals, requires further examination in breast cancer patients.
Notably, an analysis in women with operable breast cancer highlights that supervised aerobic exercise can increase the number of circulating endothelial progenitor cells (CEP) [37], thus potentially enhancing angiogenesis and decreasing tumor hypoxia. Moreover, in relation to phenotypic plasticity, CEP may facilitate the acquisition of an invasive tumor phenotype and metastasis. However, further analyses examining the effects of aerobic exercise are needed to determine whether the increase in CEP significantly impacts breast cancer invasiveness.
As previously reported, physical exercise promotes the polarization of TAMs to the M1 phenotype at the expense of the M2 phenotype, the first exerting antitumor effects, the latter promoting tumor aggressiveness (Figure 2). Consistently, sedentary lifestyles result in M2 phenotype polarization, which promotes tumor growth, invasion, and metastasis [32].
Therefore, a close connection seems to bind two different cancer hallmarks—such as phenotypic plasticity and invasion/metastasis—also evidenced by the role played by EMT in cancer metastasis. However, at present, poor data are available and the effects of physical exercise on the plasticity of breast cancer cells and CEP are still unclear.

2.12. Nonmutational Epigenetic Reprogramming

Over the last 10 years, possible mutation-less cancer evolution, based on the epigenetic reprogramming of the most characteristic cancer phenotypes, has been increasingly the subject of study. Hypoxia limits blood nutrient bioavailability and enhances the malignant transformation of mammary cells [3]; accordingly, previous results demonstrate that strength exercise could increase systemic acidosis [102] and decrease tumor hypoxia [37], thus decreasing the survival conditions of TME. However, it is known that epigenetic alterations (e.g., promoter methylation and/or histone modification) could also lead to disease initiation [104] in breast cancer; data have been accumulated on the effects of exercise on this cancer hallmark.
One clinical trial has demonstrated that aerobic exercise upregulates the expression of crucial tumor suppressor genes by reducing their methylation (usually responsible for transcriptional silencing), which would correlate with increased survival in breast cancer patients [105]. In particular, lethal(3) malignant brain tumor-like 1 (L3MBTL1) is an example of a tumor suppressor gene whose methylation increases in the control group and decreases in the exercise group. High L3MBTL1 expression leads to a 60% reduction of the risk of breast cancer death, compared to low expression levels. Furthermore, low-grade tumors show higher L3MBTL1 expression than high-grade tumors. Therefore, exercise apparently induces favorable effects on DNA methylation by reducing the methylation of the tumor suppressor gene L3MBTL1 and increasing its expression (Figure 3) [105].
Another clinical investigation reports the significant impact of promoter methylation of certain genes, namely cyclin D2 (CCND2), twist family bHLH transcription factor 1 (TWIST1), adenomatous polyposis coli (APC), and high in normal-1 (HIN1), on survival after breast cancer diagnosis, and the inverse relationship between physical activity before diagnosis and breast cancer mortality. Indeed, mortality is lower only among physically active women with promoter methylation of APC, CCND2, HIN1, and TWIST1 in tumors, but not in those with unmethylated tumor markers. At the same time, no significant association between physical activity and overall methylation has been observed [106].
Oxidative stress is another molecular mechanism assumed to be responsible for epigenetic alterations preceding cancer development, which could increase the methylation of tumor suppressor genes and, as shown in the aforementioned study [105], worsen the clinical situation of breast cancer patients. Exercise can act as an antioxidant, unless it is performed extensively in unprepared tissues, where it may increase the production of reactive oxygen species (ROS) and lead to epigenetic alterations [107].
Although studies are available on the effects of exercise on the methylation of certain genes, the scientific literature on this topic is limited and exercise-dependent epigenetic alterations require further investigation. The potential effects of exercise on oxidative stress should also be explored to determine whether it effectively modifies the methylation status of certain genes.

2.13. Polymorphic Microbiomes

Current scientific evidence is trying to describe the ability of the gut microbiome to confer susceptibility to various types of cancer and its impact on therapeutic response [108]. Specifically, for breast cancer, the presence in greater abundance of Mobiluncus, Brevundimonas, Actinomyces [109], and Fusobacterium nucleatum [110] has been reported. The latter has been correlated with an increase in tumor progression, metastasis, and the inhibition of antitumor immunity through the decrease in CD4+ and CD8+ T cells [110].
Lifestyle, including dietary patterns and physical activity, is known to substantially influence the composition of the microbiota [111]. Therefore, the effects that exercise performance provides on this hallmark of cancer are increasingly gaining importance. Indeed, physical exercise is able to alter the composition of the gut microbiota and the resulting metabolic products, by reducing inflammatory signaling pathways usually induced by being overweight or obese [108].
It is known that diet affects the gut microbiota, and that physical activity provides a positive effect on intestinal motility, as it reduces transient deposition time and contact time between pathogens and the gastrointestinal mucosal layer [73]. Interestingly, however, exercise can beneficially modify the gut microbiota independently from diet. Observational studies indicate that physical activity correlates with a higher ratio of Firmicutes to Bacteroidetes, in turn associated with higher oxygen consumption; women who exercise a minimum of three hours per week have a higher abundance of butyrate-producing bacteria and Akkermansia muciniphilia (AM), a condition that correlates with a lean body mass index [112][113]. Consistently, among women with early-stage breast cancer, body composition is associated with AM growth and microbiota diversity: women exhibiting a higher amount of body fat present a lower enrichment of AM [114], while higher levels of AM may be associated with a decrease in body fat, which could translate into an improvement in the prognosis and progression of the disease.
Another hypothesis relating the carcinogenesis of breast cancer to the intestinal microbiota is microbiota role as a principal regulator of estrogen metabolism. [115] Evidence exists of a correlation between high levels of physical activity and a decrease in the concentration of the sex hormones that play a major role in promoting the proliferation of neoplastic breast epithelium [108].
Likewise, a pilot study exploring the correlation between microbiota composition and alterations in cardiorespiratory fitness and psychosocial outcomes in breast cancer survivors, reports an association between fatigue interference and microbial diversity [116]. Moreover, an association has been observed between gut microbial composition and cardiorespiratory fitness. Therefore, exercise could affect the microbiota-gut-brain axis, promoting favorable changes in certain measures, such as fatigue and cardiorespiratory fitness [116] (Figure 4).
Apart from these analyses, currently there are no available clinical trials demonstrating the influence of exercise on the gut microbiota of women with breast cancer. Nevertheless, given the benefits of exercise on the gut microbiota in healthy individuals, further research is needed to address how exercise may affect the gut microbiome of breast cancer patients.

2.14. Senescent Cells

Senescent cells do not divide but remain viable and metabolically active. Cellular senescence can be classified as (i) non-DNA damage-related (embryonic developmentally programmed senescence and physiological senescence) and (ii) DNA damage-related. Senescence signaling pathways are activated by the phosphorylation of selected proteins, such as p53/p21, p16, and Rb [117]. Indeed, p16 and p21 proteins are cell cycle inhibitors that are upregulated in senescent cells [118]. The senescence-associated secretory phenotype (SASP) contributes to immune cell recruitment and senescent cancer cell procurement [117]. Although most research highlights the role of cellular senescence as a protective mechanism against cancer, recent research shows that, in certain contexts, senescent cells can stimulate tumor development through SASP [3] by transmitting, in a paracrine way, signaling molecules to cancer cells that could activate other cancer hallmarks [3].
One study, encompassing two different experiments, did not find an increase in p53 protein in response to aerobic exercise in a murine breast cancer model [119]. Another study shows that exercise (HIIT protocol) can increase the levels of p53 protein after four weeks of training in female BALB/c mice receiving 4T1 breast cancer cell transplants [120] and reduce tumor burden through the upregulation of p53.
Cells entering senescence due to solely the exogenous overexpression of p21 and p16 proteins do not express SASP, despite requiring p53 to stop proliferation, and maintain senescent cell characteristics. On the other hand, cells lacking functional p53 protein are able to secrete elevated concentrations of many SASP components. A large number of SASP components could promote chronic inflammation, which in turn could stimulate proliferation, differentiation, angiogenesis, and metastasis [121].
The results of the aforementioned studies [119][120] are contradictory and make it difficult to understand whether exercise increases p53 protein in animal cancer models, and how it affects p53’s relationship with SASP or cellular senescence process.
Notably, it is not yet known whether exercise in breast cancer patients could act on cellular senescence mechanisms in an anti- or pro-proliferative manner (Figure 1). The recent belief that by activating SASP it is possible to promote tumor development paves the way for future research linking senescence to exercise in women with breast cancer.

References

  1. Hanahan, D.; Weinberg, R.A. The Hallmarks of Cancer. Cell 2000, 100, 57–70.
  2. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674.
  3. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46.
  4. Ataollahi, M.R.; Sharifi, J.; Paknahad, M.R.; Paknahad, A. Breast Cancer and Associated Factors: A Review. J. Med. Life 2015, 8, 6–11.
  5. Cannioto, R.A.; Hutson, A.; Dighe, S.; McCann, W.; McCann, S.E.; Zirpoli, G.R.; Barlow, W.; Kelly, K.M.; DeNysschen, C.A.; Hershman, D.L.; et al. Physical Activity before, during, and after Chemotherapy for High-Risk Breast Cancer: Relationships with Survival. J. Natl. Cancer Inst. 2021, 113, 54–63.
  6. Palesh, O.; Kamen, C.; Sharp, S.; Golden, A.; Neri, E.; Spiegel, D.; Koopman, C. Physical Activity and Survival in Women with Advanced Breast Cancer. Cancer Nurs. 2018, 41, E31–E38.
  7. Cormie, P.; Zopf, E.M.; Zhang, X.; Schmitz, K.H. The Impact of Exercise on Cancer Mortality, Recurrence, and Treatment-Related Adverse Effects. Epidemiol. Rev. 2017, 39, 71–92.
  8. Ruiz-Casado, A.; Martín-Ruiz, A.; Pérez, L.M.; Provencio, M.; Fiuza-Luces, C.; Lucia, A. Exercise and the Hallmarks of Cancer. Trends Cancer 2017, 3, 423–441.
  9. Invernizzi, M.; Lippi, L.; Folli, A.; Turco, A.; Zattoni, L.; Maconi, A.; de Sire, A.; Fusco, N. Integrating molecular biomarkers in breast cancer rehabilitation. What is the current evidence? A systematic review of randomized controlled trials. Front. Mol. Biosci. 2022, 9, 930361.
  10. Goh, J.; Tsai, J.; Bammler, T.K.; Farin, F.M.; Endicott, E.; Ladiges, W.C. Exercise Training in Transgenic Mice Is Associated with Attenuation of Early Breast Cancer Growth in a Dose-Dependent Manner. PLoS ONE 2013, 8, e80123.
  11. Malicka, I.; Siewierska, K.; Pula, B.; Kobierzycki, C.; Haus, D.; Paslawska, U.; Cegielski, M.; Dziegiel, P.; Podhorska-Okolow, M.; Wozniewski, M. The Effect of Physical Training on the N-Methyl-N-Nitrosourea-Induced Mammary Carcinogenesis of Sprague–Dawley Rats. Exp. Biol. Med. 2015, 240, 1408–1415.
  12. Siewierska, K.; Malicka, I.; Kobierzycki, C.; Paslawska, U.; Cegielski, M.; Grzegrzolka, J.; Piotrowska, A.; Podhorska-Okolow, M.; Dziegiel, P.; Wozniewski, M. The Impact of Exercise Training on Breast Cancer. Vivo 2018, 32, 249–254.
  13. Kim, M.K.; Kim, Y.; Park, S.H.; Kim, E.; Kim, Y.; Kim, Y.; Kim, J.H. Effects of Steady Low-Intensity Exercise on High-Fat Diet Stimulated Breast Cancer Progression Via the Alteration of Macrophage Polarization. Integr. Cancer 2020, 19, 1534735420949678.
  14. Siewierska, K.; Malicka, I.; Kobierzycki, C.; Grzegrzolka, J.; Piotrowska, A.; Paslawska, U.; Cegielski, M.; Podhorska-Okolow, M.; Dziegiel, P.; Wozniewski, M. Effect of Physical Training on the Levels of Sex Hormones and the Expression of Their Receptors in Rats with Induced Mammary Cancer in Secondary Prevention Model–Preliminary Study. Vivo 2020, 34, 495–501.
  15. de Lima, C.; Alves, L.; Iagher, F.; Machado, A.F.; Kryczyk, M.; Yamazaki, R.K.; Brito, G.A.P.; Nunes, E.A.; Naliwaiko, K.; Fernandes, L.C. Tumor Growth Reduction in Walker 256 Tumorbearing Rats Performing Anaerobic Exercise: Participation of Bcl-2, Bax, Apoptosis, and Peroxidation. Appl. Physiol. Nutr. Metab. 2011, 36, 533–538.
  16. Dieli-Conwright, C.M.; Courneya, K.S.; Demark-Wahnefried, W.; Sami, N.; Lee, K.; Buchanan, T.A.; Spicer, D.; Tripathy, D.; Bernstein, L.; Mortimer, J.E. Effects of Aerobic and Resistance Exercise on Metabolic Syndrome, Sarcopenic Obesity, and Circulating Biomarkers in Overweight or Obese Survivors of Breast Cancer: A Randomized Controlled Trial. J. Clin. Oncol. 2018, 36, 875–883.
  17. Rigiracciolo, D.C.; Nohata, N.; Lappano, R.; Cirillo, F.; Talia, M.; Scordamaglia, D.; Gutkind, J.S.; Maggiolini, M. IGF-1/IGF-1R/FAK/YAP Transduction Signaling Prompts Growth E Ff Ects in Triple-Negative Breast Cancer (TNBC) Cells. Cells 2020, 9, 1010.
  18. Ligibel, J.A.; Campbell, N.; Partridge, A.; Chen, W.Y.; Salinardi, T.; Chen, H.; Adloff, K.; Keshaviah, A.; Winer, E.P. Impact of a Mixed Strength and Endurance Exercise Intervention on Insulin Levels in Breast Cancer Survivors. J. Clin. Oncol. 2008, 26, 907–912.
  19. Irwin, M.L.; Varma, K.; Alvarez-Reeves, M.; Cadmus, L.; Wiley, A.; Chung, G.G.; DiPietro, L.; Mayne, S.T.; Yu, H. Randomized Controlled Trial of Aerobic Exercise on Insulin and Insulin-like Growth Factors in Breast Cancer Survivors: The Yale Exercise and Survivorship Study. Cancer Epidemiol. Biomark. Prev. 2009, 18, 306–313.
  20. Juríková, M.; Danihel, Ľ.; Polák, Š.; Varga, I. Ki67, PCNA, and MCM Proteins: Markers of Proliferation in the Diagnosis of Breast Cancer. Acta Histochem. 2016, 118, 544–552.
  21. Dela Cruz, M.A.; Roy, P.; Chowdhury, S.; Chan, S.; Roy, H.K. Abstract P3-07-18: Exercise and Triple Negative Breast Cancer: Unravelling the Anti-Neoplastic Molecular Factors through Novel Culture Method. Cancer Res. 2017, 77, P3-07-18.
  22. Vulczak, A.; Souza, A.D.O.; Ferrari, G.D.; Azzolini, A.E.C.S.; Pereira-Da-Silva, G.; Alberici, L.C. Moderate Exercise Modulates Tumor Metabolism of Triple-Negative Breast Cancer. Cells 2020, 9, 628.
  23. Zhu, Z.; Jiang, W.; Sells, J.L.; Neil, E.S.; Mcginley, J.N.; Thompson, J. Effect of Nonmotorized Wheel Running on Mammary Carcinogenesis: Circulating Biomarkers, Cellular Processes, and Molecular Mechanisms in Rats. Cancer Epidemiol. Biomark. Prev. 2009, 17, 1920–1929.
  24. Papadopetraki, A.; Maridaki, M.; Zagouri, F.; Dimopoulos, M.A.; Koutsilieris, M.; Philippou, A. Physical Exercise Restrains Cancer Progression through Muscle-Derived Factors. Cancers 2022, 14, 1892.
  25. Alizadeh, S.; Isanejad, A.; Sadighi, S.; Khalighfard, S.; Alizadeh, A.M. Effect of a High-Intensity Interval Training on Serum MicroRNA Levels in Women with Breast Cancer Undergoing Hormone Therapy. A Single-Blind Randomized Trial. Ann. Phys. Rehabil. Med. 2019, 62, 329–335.
  26. Hojman, P.; Dethlefsen, C.; Brandt, C.; Hansen, J.; Pedersen, L.; Pedersen, B.K. Exercise-Induced Muscle-Derived Cytokines Inhibit Mammary Cancer Cell Growth. Am. J. Physiol. Endocrinol. Metab. 2011, 301, 504–510.
  27. Zhang, D.; Tan, X.; Tang, N.; Huang, F.; Chen, Z.; Shi, G. Review of Research on the Role of Irisin in Tumors. OncoTargets Ther. 2020, 13, 4423–4430.
  28. Gannon, N.P.; Vaughan, R.A.; Garcia-Smith, R.; Bisoffi, M.; Trujillo, K.A. Effects of the Exercise-Inducible Myokine Irisin on Malignant and Non-Malignant Breast Epithelial Cell Behavior in Vitro. Int. J. Cancer 2015, 136, E197–E202.
  29. Dethlefsen, C.; Hansen, L.S.; Lillelund, C.; Andersen, C.; Gehl, J.; Christensen, J.F.; Pedersen, B.K.; Hojman, P. Exercise-Induced Catecholamines Activate the Hippo Tumor Suppressor Pathway to Reduce Risks of Breast Cancer Development. Cancer Res. 2017, 77, 4894–4904.
  30. Mittal, S.; Brown, N.J.; Holen, I. The Breast Tumor Microenvironment: Role in Cancer Development, Progression and Response to Therapy. Expert Rev. Mol. Diagn. 2018, 18, 227–243.
  31. Anderson, N.M.; Simon, M.C. The Tumor Microenvironment. Curr. Biol. 2020, 30, R921–R925.
  32. Goh, J.; Kirk, E.A.; Lee, S.X.; Ladiges, W.C. Exercise, Physical Activity and Breast Cancer: The Role of Tumor-Associated Macrophages. Eexrc. Immunol. Rev. 2012, 18, 158–176.
  33. Christodoulatos, G.S.; Spyrou, N.; Kadillari, J.; Psallida, S.; Dalamaga, M. The Role of Adipokines in Breast Cancer: Current Evidence and Perspectives. Curr. Obes. Rep. 2019, 8, 413–433.
  34. van Doorslaer de Ten Ryen, S.; Deldicque, L. The Regulation of the Metastatic Cascade by Physical Activity: A Narrative Review. Cancers 2020, 12, 153.
  35. Alvarado, A.; Gil da Costa, R.M.; Faustino-Rocha, A.I.; Ferreira, R.; Lopes, C.; Oliveira, P.A.; Colaço, B. Effects of Exercise Training on Breast Cancer Metastasis in a Rat Model. Int. J. Exp. Pathol. 2017, 98, 40–46.
  36. Smeda, M.; Przyborowski, K.; Proniewski, B.; Zakrzewska, A.; Kaczor, D.; Stojak, M.; Buczek, E.; Nieckarz, Z.; Zoladz, J.A.; Wietrzyk, J.; et al. Breast Cancer Pulmonary Metastasis Is Increased in Mice Undertaking Spontaneous Physical Training in the Running Wheel; a Call for Revising Beneficial Effects of Exercise on Cancer Progression. Am. J. Cancer Res. 2017, 7, 1926–1936.
  37. Jones, L.W.; Fels, D.R.; West, M.; Allen, J.D.; Broadwater, G.; Barry, W.T.; Wilke, L.G.; Masko, E.; Douglas, P.S.; Dash, R.C.; et al. Modulation of Circulating Angiogenic Factors and Tumor Biology by Aerobic Training in Breast Cancer Patients Receiving Neoadjuvant Chemotherapy. Cancer Prev. Res. 2013, 6, 925–937.
  38. Provatopoulou, X.; Georgiou, G.P.; Kalogera, E.; Kalles, V.; Matiatou, M.A.; Papapanagiotou, I.; Sagkriotis, A.; Zografos, G.C.; Gounaris, A. Serum Irisin Levels Are Lower in Patients with Breast Cancer: Association with Disease Diagnosis and Tumor Characteristics. BMC Cancer 2015, 15, 898.
  39. Zhang, Z.P.; Zhang, X.F.; Li, H.; Liu, T.J.; Zhao, Q.P.; Huang, L.H.; Cao, Z.J.; He, L.M.; Hao, D.J. Serum Irisin Associates with Breast Cancer to Spinal Metastasis. Medicine 2018, 97, e0524.
  40. Saxena, N.K.; Sharma, D. Metastasis Suppression by Adiponectin: LKB1 Rises up to the Challenge. Cell Adhes. Migr. 2010, 4, 358–362.
  41. Ray, A.; Cleary, M.P. The Potential Role of Leptin in Tumor Invasion and Metastasis. Cytokine Growth Factor Rev. 2017, 38, 80–97.
  42. Ham, B.; Fernandez, M.C.; D’costa, Z.; Brodt, P. The Diverse Roles of the TNF Axis in Cancer Progression and Metastasis. Trends Cancer Res. 2016, 11, 1–27.
  43. Loh, H.Y.; Norman, B.P.; Lai, K.S.; Rahman, N.M.A.N.A.; Alitheen, N.B.M.; Osman, M.A. The Regulatory Role of MicroRNAs in Breast Cancer. Int. J. Mol. Sci. 2019, 20, 4940.
  44. Lindholm, M.E.; Rundqvist, H. Skeletal Muscle Hypoxia-Inducible Factor-1 and Exercise. Exp. Physiol. 2016, 101, 28–32.
  45. Boimel, P.J.; Smirnova, T.; Ni Zhou, Z.; Wyckoff, J.; Park, H.; Coniglio, S.J.; Qian, B.-Z.; Richard Stanley, E.; Cox, D.; Pollard, J.W.; et al. Contribution of CXCL12 Secretion to Invasion of Breast Cancer Cells. Breast Cancer Res. 2012, 14, R23.
  46. Okamoto, K.; Seimiya, H. Revisiting Telomere Shortening in Cancer. Cells 2019, 8, 107.
  47. Arsenis, N.C.; You, T.; Ogawa, E.F.; Tinsley, G.M.; Zuo, L. Physical Activity and Telomere Length: Impact of Aging and Potential Mechanisms of Action. Oncotarget 2017, 8, 45008–45019.
  48. Gillman, A.S.; Helmuth, T.; Koljack, C.E.; Hutchison, K.E.; Kohrt, W.M.; Bryan, A.D. The Effects of Exercise Duration and Intensity on Breast Cancer-Related Dna Methylation: A Randomized Controlled Trial. Cancers 2021, 13, 4128.
  49. Grazioli, E.; Dimauro, I.; Mercatelli, N.; Wang, G.; Pitsiladis, Y.; di Luigi, L.; Caporossi, D. Physical Activity in the Prevention of Human Diseases: Role of Epigenetic Modifications. BMC Genom. 2017, 18, 802.
  50. Mandujano-Tinoco, E.A.; García-Venzor, A.; Melendez-Zajgla, J.; Maldonado, V. New Emerging Roles of MicroRNAs in Breast Cancer. Breast Cancer Res. Treat. 2018, 171, 247–259.
  51. Hrdličková, R.; Nehyba, J.; Bargmann, W.; Bose, H.R. Multiple Tumor Suppressor MicroRNAs Regulate Telomerase and TCF7, an Important Transcriptional Regulator of the Wnt Pathway. PLoS ONE 2014, 9, e86990.
  52. Sanft, T.; Usiskin, I.; Harrigan, M.; Cartmel, B.; Lu, L.; Li, F.Y.; Zhou, Y.; Chagpar, A.; Ferrucci, L.M.; Pusztai, L.; et al. Randomized Controlled Trial of Weight Loss versus Usual Care on Telomere Length in Women with Breast Cancer: The Lifestyle, Exercise, and Nutrition (LEAN) Study. Breast Cancer Res. Treat. 2018, 172, 105–112.
  53. Zhang, Y.; Zhang, H.; Wang, M.; Schmid, T.; Xin, Z.; Kozhuharova, L.; Yu, W.K.; Huang, Y.; Cai, F.; Biskup, E. Hypoxia in Breast Cancer—Scientific Translation to Therapeutic and Diagnostic Clinical Applications. Front. Oncol. 2021, 11, 652266.
  54. Madu, C.O.; Wang, S.; Madu, C.O.; Lu, Y. Angiogenesis in Breast Cancer Progression, Diagnosis, and Treatment. J. Cancer 2020, 11, 4474–4494.
  55. Betof, A.S.; Lascola, C.D.; Weitzel, D.; Landon, C.; Scarbrough, P.M.; Devi, G.R.; Palmer, G.; Jones, L.W.; Dewhirst, M.W. Modulation of Murine Breast Tumor Vascularity, Hypoxia, and Chemotherapeutic Response by Exercise. J. Natl. Cancer Inst. 2015, 107, djv40.
  56. Jones, L.W.; Viglianti, B.L.; Tashjian, J.A.; Kothadia, S.M.; Keir, S.T.; Freedland, S.J.; Potter, M.Q.; Moon, J.; Schroeder, T.; Herndon, J.E.; et al. Effect of Aerobic Exercise on Tumor Physiology in an Animal Model of Human Breast Cancer. J. Appl. Physiol. 2010, 108, 343–348.
  57. Scott, J.M.; Jones, L.W.; Hornsby, W.E.; Koelwyn, G.J.; Khouri, M.G.; Joy, A.A.; Douglas, P.S.; Lakoski, S.G. Cancer Therapy-Induced Autonomic Dysfunction in Early Breast Cancer: Implications for Aerobic Exercise Training. Int. J. Cardiol. 2014, 171, e50-1.
  58. Magnussen, A.L.; Mills, I.G. Vascular Normalisation as the Stepping Stone into Tumour Microenvironment Transformation. Br. J. Cancer 2021, 125, 324–336.
  59. Amani Shalamzari, S.; Agha-Alinejad, H.; Alizadeh, S.; Shahbazi, S.; Kashani Khatib, Z.; Kazemi, A.; Saei, M.A.; Minayi, N. The Effect of Exercise Training on the Level of Tissue IL-6 and Vascular Endothelial Growth Factor in Breast Cancer Bearing Mice. Iran J. Basic Med. Sci. 2014, 17, 231–236.
  60. Faustino-Rocha, A.I.; Silva, A.; Gabriel, J.; Gil da Costa, R.M.; Moutinho, M.; Oliveira, P.A.; Gama, A.; Ferreira, R.; Ginja, M. Long-Term Exercise Training as a Modulator of Mammary Cancer Vascularization. Biomed. Pharmacother. 2016, 81, 273–280.
  61. Pakiz, B.; Flatt, S.W.; Bardwell, W.A.; Rock, C.L.; Mills, P.J. Effects of a Weight Loss Intervention on Body Mass, Fitness, and Inflammatory Biomarkers in Overweight or Obese Breast Cancer Survivors. Int. J. Behav. Med. 2011, 18, 333–341.
  62. Ergun, M.; Eyigor, S.; Karaca, B.; Kisim, A.; Uslu, R. Effects of Exercise on Angiogenesis and Apoptosis-Related Molecules, Quality of Life, Fatigue and Depression in Breast Cancer Patients. Eur. J. Cancer Care 2013, 22, 626–637.
  63. Jan, R.; Chaudhry, G.E. Understanding Apoptosis and Apoptotic Pathways Targeted Cancer Therapeutics. J. Cardiovasc. Thorac. Res. 2019, 9, 205–218.
  64. Alizadeh, A.M.; Heydari, Z.; Rahimi, M.; Bazgir, B.; Shirvani, H.; Alipour, S.; Heidarian, Y.; Khalighfard, S.; Isanejad, A. Oxytocin Mediates the Beneficial Effects of the Exercise Training on Breast Cancer. Exp. Physiol. 2018, 103, 222–235.
  65. Liberti, M.V.; Locasale, J.W. The Warburg Effect: How Does It Benefit Cancer Cells? Trends Biochem. Sci. 2016, 41, 211–218.
  66. Kroemer, G.; Pouyssegur, J. Tumor Cell Metabolism: Cancer’s Achilles’ Heel. Cancer Cell 2008, 13, 472–482.
  67. Hojman, P.; Gehl, J.; Christensen, J.F.; Pedersen, B.K. Molecular Mechanisms Linking Exercise to Cancer Prevention and Treatment. Cell Metab. 2018, 27, 10–21.
  68. Aveseh, M.; Nikooie, R.; Aminaie, M. Exercise-Induced Changes in Tumour LDH-B and MCT1 Expression Are Modulated by Oestrogen-Related Receptor Alpha in Breast Cancer-Bearing BALB/c Mice. J. Physiol. 2015, 593, 2635–2648.
  69. Wang, Z.Y.; Loo, T.Y.; Shen, J.G.; Wang, N.; Wang, D.M.; Yang, D.P.; Mo, S.L.; Guan, X.Y.; Chen, J.P. LDH-A Silencing Suppresses Breast Cancer Tumorigenicity through Induction of Oxidative Stress Mediated Mitochondrial Pathway Apoptosis. Breast Cancer Res. Treat. 2012, 131, 791–800.
  70. Kim, J.H.; Kim, E.L.; Lee, Y.K.; Park, C.B.; Kim, B.W.; Wang, H.J.; Yoon, C.H.; Lee, S.J.; Yoon, G. Decreased Lactate Dehydrogenase B Expression Enhances Claudin 1-Mediated Hepatoma Cell Invasiveness via Mitochondrial Defects. Exp. Cell Res. 2011, 317, 1108–1118.
  71. Jiang, X.; Shapiro, D.J. The Immune System and Inflammation in Breast Cancer. Mol. Cell Endocrinol. 2014, 382, 673–682.
  72. Nicolini, A.; Ferrari, P.; Carpi, A. Immune Checkpoint Inhibitors and Other Immune Therapies in Breast Cancer: A New Paradigm for Prolonged Adjuvant Immunotherapy. Biomedicines 2022, 10, 2511.
  73. Jurdana, M. Physical Activity and Cancer Risk. Actual Knowledge and Possible Biological Mechanisms. Radiol. Oncol. 2021, 55, 7–17.
  74. Wennerberg, E.; Lhuillier, C.; Rybstein, M.D.; Dannenberg, K.; Rudqvist, N.P.; Koelwyn, G.J.; Jones, L.W.; Demaria, S. Exercise Reduces Immune Suppression and Breast Cancer Progression in a Preclinical Model. Oncotarget 2020, 11, 452–461.
  75. Fairey, A.S.; Courneya, K.S.; Field, C.J.; Bell, G.J.; Jones, L.W.; Mackey, J.R. Randomized Controlled Trial of Exercise and Blood Immune Function in Postmenopausal Breast Cancer Survivors. J. Appl. Physiol. 2005, 98, 1534–1540.
  76. Hagstrom, A.D.; Marshall, P.W.M.; Lonsdale, C.; Papalia, S.; Cheema, B.S.; Toben, C.; Baune, B.T.; Fiatarone Singh, M.A.; Green, S. The Effect of Resistance Training on Markers of Immune Function and Inflammation in Previously Sedentary Women Recovering from Breast Cancer: A Randomized Controlled Trial. Breast Cancer Res. Treat. 2016, 155, 471–482.
  77. Schmidt, T.; Jonat, W.; Wesch, D.; Oberg, H.H.; Adam-Klages, S.; Keller, L.; Röcken, C.; Mundhenke, C. Influence of Physical Activity on the Immune System in Breast Cancer Patients during Chemotherapy. J. Cancer Res. Clin. Oncol. 2018, 144, 579–586.
  78. Valenzuela, P.L.; Saco-Ledo, G.; Santos-Lozano, A.; Morales, J.S.; Castillo-García, A.; Simpson, R.J.; Lucia, A.; Fiuza-Luces, C. Exercise Training and Natural Killer Cells in Cancer Survivors: Current Evidence and Research Gaps Based on a Systematic Review and Meta-Analysis. Sports Med. Open 2022, 8, 36.
  79. Nieman, D.C.; Cook, V.D.; Henson, D.A.; Suttles, J.; Rejeski, W.J.; Ribisl, P.M.; Fagoaga, O.R.; Nehlsen-Cannarella, S.L. Moderate Exercise Training and Natural Killer Cell Cytotoxic Activity in Breast Cancer Patients. Int. J. Sports Med. 1995, 16, 334–337.
  80. Hutnick, N.A.; Williams, N.I.; Kraemer, W.J.; Orsega-Smith, E.; Dixon, R.H.; Bleznak, A.D.; Mastro, A.M. Excercise and Lymphocyte Activation Following Chemotherapy for Breast Cancer. Med. Sci. Sports Exerc. 2005, 37, 1827–1835.
  81. Saxton, J.M.; Scott, E.J.; Daley, A.J.; Woodroofe, M.N.; Mutrie, N.; Crank, H.; Powers, H.J.; Coleman, R.E. Effects of an Exercise and Hypocaloric Healthy Eating Intervention on Indices of Psychological Health Status, Hypothalamic-Pituitary-Adrenal Axis Regulation and Immune Function after Early-Stage Breast Cancer: A Randomised Controlled Trial. Breast Cancer Res. 2014, 16, R39.
  82. Kwei, K.A.; Kung, Y.; Salari, K.; Holcomb, I.N.; Pollack, J.R. Genomic Instability in Breast Cancer: Pathogenesis and Clinical Implications. Mol. Oncol. 2010, 4, 255–266.
  83. Duijf, P.H.G.; Nanayakkara, D.; Nones, K.; Srihari, S.; Kalimutho, M.; Khanna, K.K. Mechanisms of Genomic Instability in Breast Cancer. Trends Mol. Med. 2019, 25, 595–611.
  84. Costa, M.; Saldanha, P. Risk Reduction Strategies in Breast Cancer Prevention. Eur. J. Breast Health 2016, 13, 103–112.
  85. Kehm, R.D.; Genkinger, J.M.; MacInnis, R.J.; John, E.M.; Phillips, K.A.; Dite, G.S.; Milne, R.L.; Zeinomar, N.; Liao, Y.; Knight, J.A.; et al. Recreational Physical Activity Is Associated with Reduced Breast Cancer Risk in Adult Women at High Risk for Breast Cancer: A Cohort Study of Women Selected for Familial and Genetic Risk. Cancer Res. 2020, 80, 116–125.
  86. Pettapiece-Phillips, R.; Narod, S.A.; Kotsopoulos, J. The Role of Body Size and Physical Activity on the Risk of Breast Cancer in BRCA Mutation Carriers. Cancer Causes Control 2015, 26, 333–344.
  87. Bizjak, D.A.; Schulz, S.V.W.; Schumann, U.; Otto, S.; Kirsten, J.; Ebner, F.; Leinert, E.; Huober, J.; Janni, W.; Steinacker, J.M. Beneficial Molecular Adaptations in BRCA-Mutation Carriers by Combined HIT/HIRT Intervention: Results from a Pilot Study. Cancers 2020, 12, 1526.
  88. Munn, L. Cancer and Inflammation. Wiley Interdiscip. Rev. Syst. Biol. Med. 2017, 9, e1370.
  89. Danforth, D.N. The Role of Chronic Inflammation in the Development of Breast Cancer. Cancers 2021, 13, 3918.
  90. Mills, R.C. Breast Cancer Survivors, Common Markers of Inflammation, and Exercise: A Narrative Review. Breast Cancer 2017, 11, 1178223417743976.
  91. Murphy, E.A.; Davis, J.M.; Barrilleaux, T.L.; McClellan, J.L.; Steiner, J.L.; Carmichael, M.D.; Pena, M.M.; Hebert, J.R.; Green, J.E. Benefits of Exercise Training on Breast Cancer Progression and Inflammation in C3(1)SV40Tag Mice. Cytokine 2012, 55, 274–279.
  92. Isanejad, A.; Alizadeh, A.M.; Amani Shalamzari, S.; Khodayari, H.; Khodayari, S.; Khori, V.; Khojastehnjad, N. MicroRNA-206, Let-7a and MicroRNA-21 Pathways Involved in the Anti-Angiogenesis Effects of the Interval Exercise Training and Hormone Therapy in Breast Cancer. Life Sci. 2016, 151, 30–40.
  93. Jones, S.B.; Thomas, G.A.; Hesselsweet, S.D.; Alvarez-reeves, M.; Yu, H.; Irwin, M.L. Effect of Exercise on Markers of Inflammation in Breast Cancer Survivors: The Yale Exercise and Survivorship Study. Cancer Prev. Res. 2013, 6, 109–118.
  94. Hooshmand Moghadam, B.; Golestani, F.; Bagheri, R.; Cheraghloo, N.; Eskandari, M.; Wong, A.; Nordvall, M.; Suzuki, K.; Pournemati, P. The Effects of High-Intensity Interval Training vs. Moderate-Intensity Continuous Training on Inflammatory Markers, Body Composition, and Physical Fitness in Overweight/Obese Survivors of Breast Cancer: A Randomized Controlled Clinical Trial. Cancers 2021, 13, 4386.
  95. Alizadeh, A.M.; Isanejad, A.; Sadighi, S.; Mardani, M.; Kalaghchi, B.; Hassan, Z.M. High-Intensity Interval Training Can Modulate the Systemic Inflammation and HSP70 in the Breast Cancer: A Randomized Control Trial. J. Cancer Res. Clin. Oncol. 2019, 145, 2583–2593.
  96. Kim, S.H.; Song, Y.K.; Han, J.; Ko, Y.H.; Lee, H.; Kang, M.J.; Park, H.; Lee, H.; Kim, S. Pro-Inflammatory Cytokine Levels and Cancer-Related Fatigue in Breast Cancer Survivors: Effects of an Exercise Adherence Program. J. Breast Cancer 2020, 23, 205–217.
  97. Hiensch, A.E.; Mijwel, S.; Bargiela, D.; Wengström, Y.; May, A.M.; Rundqvist, H. Inflammation Mediates Exercise Effects on Fatigue in Patients with Breast Cancer. Med. Sci. Sports Exerc. 2021, 53, 496–504.
  98. Schmidt, M.E.; Meynköhn, A.; Habermann, N.; Wiskemann, J.; Oelmann, J.; Hof, H.; Wessels, S.; Klassen, O.; Debus, J.; Potthoff, K.; et al. Resistance Exercise and Inflammation in Breast Cancer Patients Undergoing Adjuvant Radiation Therapy: Mediation Analysis from a Randomized, Controlled Intervention Trial. Int. J. Radiat. Oncol. Biol. Phys. 2016, 94, 329–337.
  99. Rogers, L.Q.; Fogleman, A.; Trammell, R.; Hopkins-Price, P.; Vicari, S.; Rao, K.; Edson, B.; Verhulst, S.; Courneya, K.S.; Hoelzer, K. Effects of a Physical Activity Behavior Change Intervention on Inflammation and Related Health Outcomes in Breast Cancer Survivors: Pilot Randomized Trial. Integr. Cancer 2013, 12, 323–335.
  100. Dieli-Conwright, C.M.; Parmentier, J.H.; Sami, N.; Lee, K.; Spicer, D.; Mack, W.J.; Sattler, F.; Mittelman, S.D. Adipose Tissue Inflammation in Breast Cancer Survivors: Effects of a 16-Week Combined Aerobic and Resistance Exercise Training Intervention. Breast Cancer Res. Treat. 2018, 168, 147–157.
  101. Lüönd, F.; Tiede, S.; Christofori, G. Breast Cancer as an Example of Tumour Heterogeneity and Tumour Cell Plasticity during Malignant Progression. Br. J. Cancer 2021, 125, 164–175.
  102. Smallbone, K.; Maini, P.K.; Gatenby, R.A. Episodic, Transient Systemic Acidosis Delays Evolution of the Malignant Phenotype: Possible Mechanism for Cancer Prevention by Increased Physical Activity. Biol. Direct 2010, 5, 22.
  103. Clarkson, P.M.; Kaufman, S.A. Should Resistance Exercise Be Recommended during Breast Cancer Treatment? Med. Hypotheses 2010, 75, 192–195.
  104. Wu, Y.; Sarkissyan, M.; Vadgama, J.v. Epigenetics in Breast and Prostate Cancer. Methods Mol. Biol. 2015, 1238, 425–466.
  105. Zeng, H.; Irwin, M.L.; Lu, L.; Risch, H.; Mayne, S.; Mu, L.; Deng, Q.; Scarampi, L.; Mitidieri, M.; Katsaros, D.; et al. Physical Activity and Breast Cancer Survival: An Epigenetic Link through Reduced Methylation of a Tumor Suppressor Gene L3MBTL1. Breast Cancer Res. Treat. 2012, 133, 127–135.
  106. McCullough, L.E.; Chen, J.; Cho, Y.H.; Khankari, N.K.; Bradshaw, P.T.; White, A.J.; Teitelbaum, S.L.; Terry, M.B.; Neugut, A.I.; Hibshoosh, H.; et al. Modification of the Association between Recreational Physical Activity and Survival after Breast Cancer by Promoter Methylation in Breast Cancer-Related Genes. Breast Cancer Res. 2017, 19, 19.
  107. Adraskela, K.; Veisaki, E.; Koutsilieris, M.; Philippou, A. Physical Exercise Positively Influences Breast Cancer Evolution. Clin. Breast Cancer 2017, 17, 408–417.
  108. Hong, B.S.; Lee, K.P. A Systematic Review of the Biological Linking Physical activity and Breast Cancer. Phys. Act. Nutr. 2020, 24, 25–31.
  109. Wu, H.; Ganguly, S.; Tollefsbol, T.O. Modulating Microbiota as a New Strategy for Breast Cancer Prevention and Treatment. Microorganisms 2022, 10, 1727.
  110. Vitorino, M.; Alpuim Costa, D.; Vicente, R.; Caleça, T.; Santos, C. Local Breast Microbiota: A “New” Player on the Block. Cancers 2022, 14, 3811.
  111. Bodai, B.I.; Nakata, T.E. Breast Cancer: Lifestyle, the Human Gut Microbiota/Microbiome, and Survivorship. Perm. J. 2020, 24, 19.129.
  112. Sampsell, K.; Hao, D.; Reimer, R.A. The Gut Microbiota: A Potential Gateway to Improved Health Outcomes in Breast Cancer Treatment and Survivorship. Int. J. Mol. Sci. 2020, 21, 9239.
  113. Mailing, L.J.; Allen, J.M.; Buford, T.W.; Fields, C.J.; Woods, J.A. Exercise and the Gut Microbiome: A Review of the Evidence, Potential Mechanisms, and Implications for Human Health. Exerc. Sports Sci. Rev. 2019, 47, 75–85.
  114. Frugé, A.D.; Rogers, L.Q.; Morrow, C.D.; Tsuruta, Y.; Demark-Wahnefried, W. Fecal Akkermansia Muciniphila Is Associated with Body Composition and Microbiota Diversity in Overweight and Obese Women with Breast Cancer Participating in a Presurgical Weight Loss Trial. J. Acad. Nutr. Diet. 2020, 120, 650–659.
  115. Helmink, B.A.; Khan, M.A.W.; Hermann, A.; Gopalakrishnan, V.; Wargo, J.A. The Microbiome, Cancer, and Cancer Therapy. Nat. Med. 2019, 25, 377–388.
  116. Paulsen, J.A.; Ptacek, T.S.; Carter, S.J.; Liu, N.; Kumar, R.; Hyndman, L.K.; Lefkowitz, E.J.; Morrow, C.D.; Rogers, L.Q. Gut Microbiota Composition Associated with Alterations in Cardiorespiratory Fitness and Psychosocial Outcomes among Breast Cancer Survivors. Support. Care Cancer 2017, 25, 1563–1570.
  117. Milczarek, M. The Premature Senescence in Breast Cancer Treatment Strategy. Cancers 2020, 12, 1815.
  118. Garatachea, N.; Pareja-Galeano, H.; Sanchis-Gomar, F.; Santos-Lozano, A.; Fiuza-Luces, C.; Morán, M.; Emanuele, E.; Joyner, M.J.; Lucia, A. Exercise Attenuates the Major Hallmarks of Aging. Rejuvenation Res. 2015, 18, 57–89.
  119. Colbert, L.H.; Westerlind, K.C.; Perkins, S.N.; Haines, D.C.; Berrigan, D.; Donehower, L.A.; Fuchs-Young, R.; Hursting, S.D. Exercise Effects on Tumorigenesis in a P53-Deficient Mouse Model of Breast Cancer. Med. Sci. Sports Exerc. 2009, 41, 1597–1605.
  120. Nezamdoost, Z.; Saghebjoo, M.; Hoshyar, R.; Hedayati, M.; Keska, A. High-Intensity Training and Saffron: Effects on Breast Cancer-Related Gene Expression. Med. Sci. Sports Exerc. 2020, 52, 1470–1476.
  121. González-Puertos, V.Y.; Maciel-Barón, L.A.; Barajas-Gómez, B.A.; López-Diazguerrero, N.E.; Königsberg, M. Senescence-Associated Secretory Phenotype (SASP) Involvement in the Development of Cancer, Aging and Age Related Diseases. Gac. Médica México 2015, 151, 460–468.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , , , , ,
View Times: 598
Revisions: 6 times (View History)
Update Date: 01 Feb 2023
1000/1000