Role of CDKIs in Diet-Induced Obesity and Cancer: Comparison
Please note this is a comparison between Version 2 by Jason Zhu and Version 1 by Reyes Benot Dominguez.

Overweight and obesity constitute the most impactful lifestyle-dependent risk factors for cancer and have been tightly linked to a higher number of tumor-related deaths nowadays. The excessive accumulation of energy can lead to an imbalance in the level of essential cellular biomolecules that may result in inflammation and cell-cycle dysregulation. Nutritional strategies and phytochemicals are gaining interest in the management of obesity-related cancers, with several ongoing and completed clinical studies that support their effectiveness. At the same time, cyclin-dependent kinases (CDKs) are becoming an important target in breast and ovarian cancer treatment, with various FDA-approved CDK4/6 inhibitors that have recently received more attention for their potential role in diet-induced obesity (DIO).

  • CDK inhibitors (CDKIs)
  • diet-induced obesity (DIO)
  • ovarian cancer
  • Breast cancer
  • Cyclin-dependent kinases
  • cell cycle inhibitors
  • CDK 4/6
  • RB1

1. Obesity and Cell-Cycle Progression in Cancer

Obesity has become an emergent pandemic involving 1/3 of the population worldwide, a multifaceted disorder characterized by the overabundant accumulation of adipocytes—fat cells—which in turn may aggravate the course of different types of chronic diseases. In fact, more aggressive tumor profiles have been seen in overweight breast cancer patients, where adipocytes can secrete hormones, growth factors, and adipokines and release free fatty acids (FFA). The energy obtained from the β-oxidation of FFA can be used by adipocytes to accelerate tumor cell growth and cancer progression, stimulating the oncogenic signaling and leading to angiogenesis and malignant cell migration [28,29] [1][2. Similarly, ovarian cancer (OC) risk has also been correlated with elevated BMI and lipid levels. A significant 16–30% increased predisposition to OC has been identified in obese women, with a major risk assigned to specific histological subtypes—mostly endometrioid and mucinous carcinomas—and postmenopausal patients [30] [3. This pathological condition is frequently associated with the overexpression of pro-inflammatory factors, cytokines, and adipokines, a process promoted by macrophage infiltration within the adipose tissue and able to exert tumor-promoting effects [31] [4. Actually, the delivery of pro-inflammatory metabolites in the bloodstream may degenerate into hypothalamus deregulation, cause the loss of energy homeostasis, and bring to the disruption of crucial biological pathways, including those determining cell-cycle regulation. As a result, elevated levels of markers of inflammation such as tumor necrosis factor-alpha (TNF-α), interleukins (IL) 1—6-8, plasminogen activator inhibitor 1 (PAI1 or SERPINE1), and C-reactive protein (CRP) are also frequently found in obese patients.
The maintenance of energy and body weight balance are critical processes mainly led by the hypothalamic neurons, which constitute one of the most essential targets for adipokines such as leptin, a product of the obese gene (Ob). Altered levels of leptin and related hormones with a key role in food intake and appetite stimuli—such as ghrelin or insulin—are also generally present among overweight patients [32] [5.

1.1. Adipokines' Role in Cell-Cycle Progression

Adipokines are a group of cytokines secreted in the adipose tissue involved in the metabolic signaling in the brain, with an extensively demonstrated function in fostering cancer development. The most likely molecular mediators of inflammation from the adipose tissue itself are the adipokines leptin (discovered in 1994) and adiponectin (described for the first time in 1995)—critical for the maintenance of balanced bodyweight—and pro-inflammatory agents.

1.1.1. Leptin

A wide number of epidemiological studies are focusing on leptin hormone and its receptor (LepR) as good targets for the treatment approach of DIO, and have directly correlated the respective anorexigenic effects with the tumor cascade outgrowth. Leptin has been suggested to strongly take part in cancer onset and proliferation, by activating several growth signaling pathways such as PI3K/Akt, MAPKs (ERK1/2), and JAK/STAT3 [32 [5][6,33], which drive cell-cycle progression acting on different target genes. These properties joined with the ability of leptin to promote angiogenesis, confer to this adipokine the main role as a growth factor for cancer cells [34] [7. Indeed, some options presently under study are devoted to inhibiting the leptin cytokine cascade by using multiple approaches, including antibodies [35] [8, peptides [36] [9, and PPAR ligands [37,38] [10][11. Some of these leptin antagonists have been seen to efficiently arrest the leptin-induced cell-cycle progression at the S-phase in triple-negative breast cancer (TNBC) [39,40,41] [12][13][14.
Ptak et al. examined the relationship between OC development and leptin rates in obese women, identifying a key role of adipokine in stimulating cell cycle-related effectors. The research group connected the tumor proliferation role of leptin with a greater expression of cyclin D—indicative of a poorer prognosis—and cyclin A in an in vitro model; also identifying a downregulation of p21 [33] [6. These results indicate leptin as a cell-cycle promoter, driving G1/S-phase transition in an OC model. Furthermore, an anti-apoptotic role of leptin was also observed in ovarian carcinogenesis through the blockade of caspase expression, which further stimulates cancer cell proliferation.
The expression levels of leptin constitute an interesting diagnostic tool that can be used to determine cancer risk, grade and type, stage, lymph node involvement, hormone receptors, and prognosis in breast [42,43,44] [15][16][17 and ovarian tumors [45] [18. High leptin levels have been also correlated with lower chemosensitivity, and a common nexus between leptin and several mechanisms that come usually activated in breast tumors. Moreover, LEPR expression has been seen to be able to increase the cancer stem cell state in breast tumors, further promoting cell proliferation, stemness, and poorer survival [50] [19.

1.1.2. Adiponectin (APN)

In contrast to leptin’s effects on cancer, APN has been demonstrated to exert a protective role in the course of different malignancies, especially BC. APN is gaining interest in the management of obesity, where the extremely low levels of this hormone have been linked to insulin resistance, glucose metabolism, and thermogenesis processes. Interestingly, insulin promotes MCF-7 breast tumor cells proliferation and migration via PI3K activation [51] [20, and APN is able to downregulate the PI3K/Akt/mTOR cascade, resulting in an overall decrease in cancer cell viability, survival, and growth. Focusing on cancer signaling, this adipokine also activates AMPK, which induces cell-cycle arrest, apoptosis, and senescence via p21 activation and p53 phosphorylation. Moreover, APN upregulation also impedes STAT3 signal pathway activation, which in turn is unable to endorse angiogenesis and invasion and incapable of evading anticancer immunity, hence blocking tumor progression. The anti-inflammatory and pro-apoptotic effects of APN also involve the abolition of the NF-kB cascade by hampering NF-kB phosphorylation [52,53] [21][22.
In an in vitro model of TNBC (MDA-MB-231 cell line), APN enhanced the overexpression of master genes that control cell-cycle progression, such as p53, and apoptosis (BAX, BCL2) [54,55] [23][24. This study also showed how the repression of the proto-oncogene MYC prevented cyclin D1 activation, consequently arresting the cell cycle at the G1/S-phase and hampering TNBC expansion. Nonetheless, the antiproliferative effects of APN in mammary cancers seem reliant on ERα expression, as an opposite role was observed in ERα + BC, where APN seems to promote cancer cell growth. On the opposite, low APN levels stimulate MAPK activation, which consequently phosphorylates SP1 and ER and enhances cyclin D1 expression, stimulating BC growth.
Despite the potential ERα-dependent effect of APN, patients with low leptin/APN ratios have shown a statistically longer cancer-specific survival for OC, which may show APN as a good candidate against DIO and derived metabolic diseases, including cancer [56] [25.

1.2. Additional Pro-Inflammatory Cytokines

The relationship between chronic inflammation, obesity, and several types of cancer has been extensively investigated, correlating the aggressiveness of tumor disease with higher levels of circulating inflammatory biomarkers such as cytokines [57] [26. These functional proteins released by immune, stromal, and tumor cells affect cell proliferation via cell-cycle-regulatory proteins. As an example, the transforming growth factor-beta (TGF-β), is a key cytokine that in normal conditions induces tolerance and suppresses inflammation and in the early phases of tumorigenesis acts as a cytostatic tumor suppressive agent acting through p21 and p27 CDKI expression and inducing cancer cell apoptosis[58,59]s[27][28. During tumor progression, however, TGF-β or its pathway is altered and decoupled from their tumor suppressor activity leading them to promote EMT and favoring a tumor immunosuppressive microenvironment that further enhances tumor invasiveness, as has been seen in HER2- BC [59,60] [28][29.
Inflammatory cytokines such as IL-6, IL-21, IL-1β, and TNF-α diminish the cytotoxic capacity of immune CD8+ T cells to produce IFN-γ, which plays a main role in angiogenesis and MHC expression—tumor recognition. Higher values of these cytokines directly increase IL-17 production, activating angiogenesis and tumor growth [61] [30. Indeed, risen expression of IL-1, IL-5, IL-6, IL-17, and NFκB were linked to aggressive phenotypes in BC patients and were correlated to a poorer prognosis and lower survival rates [62,63] [31][32.
Interestingly, IL-6 has been demonstrated to switch on the JAK/STAT3 pathway and enhance EMT, which in turn promotes cancer proliferation, metastasis, and chemoresistance. Moreover, IL-6 is the main pro-inflammatory factor responsible for inducing the overexpression of tumor-related RAC1B, known to sustain tumor cell survival and promote escape from oncogene-induced senescence. Finally, increased levels of serum IL-6 have been correlated with poor prognosis, tumor size, and disease status [64] [33. IL-17 can trigger the production of IL-6, which increases tumor cell migration and invasion, therefore contributing to tumor drug sensitivity and resistance to chemotherapy [65] [34.
IL-8 is also frequently found at high levels in BC, exerting inflammatory and angiogenic actions. In fact, a recently published study has shown that the pro-tumorigenic and metastatic effect of IL-8 passes by the activation of PI3K-Akt/MAPK and EMT signaling pathways leading to tumor cell migration[66]n[35. IL-4 instead upregulates adhesion molecules, inhibits cell proliferation and apoptosis, and mediates signal transduction in breast (MDA-MB-231) and ovarian tumors (SKOV-3), among others [67] [36. Controversially, it has also been claimed that IL-4 possesses potent antitumor activity against various cancer types, including breast tumors, a reason by which additional research is needed prior to reaching a unique conclusion for this pro-inflammatory factor.
Interleukin-9 (IL-9) is a cytokine with pleiotropic functions that plays an important role in regulating tumor cell growth. IL-9 is increasingly produced by tumor-infiltrating T cells (TILs), as well as tumor cells themselves and a subset of Foxp3 expressing regulatory T cells (Tregs). FoxP3+ Treg cells are known to suppress antitumor immunity, suggesting that IL-9 derived from these cells might control immune responses [68] [37.
IL-10 is an immunosuppressive cytokine that can inhibit the ability of dendritic cells and macrophages to activate CD4 + T cells. IL-10 is frequently present at sites of chronic inflammation, promoting immunosuppression of humoral responses through the induction of isotype switching to IgG4. In a recent study, autho researchrs found a significant expression of IL-10 in tumor-infiltrating B-cells of TNBC patients, driving isotype switch to the IgG4 isotype in an IL-10 dependent manner [69] [38.
These observations suggest that IL-10 may play a role in directing antitumor immune escape. Moreover, both IgG4 and tumor IL-10 are associated with shorter recurrence-free survival (RFS) and overall survival (OS). In BC, IL-10 expression positively correlates with locally advanced disease and higher tumor grade and has been proposed as a good prognostic indicator of disease-free survival (DFS) [70] [39. In melanoma, IL-10 expression by tumor cells is associated with melanoma progression [71] [40, while overexpression of serous IL-10 leads to an adverse survival in most cancer types [72] [41.
Pro-inflammatory stimuli have been seen to be able to raise pro-angiogenic factors in TNBC cells that physically interact with mesenchymal stem cells—MSCs—and stromal cells, accelerating the metastatic phenotype [73] [42. Additionally, the Notch pathway, probably via CXCL8 cytokine release, has been demonstrated to promote the cell-to-cell interaction, affecting proliferation, differentiation, and death of cells—fostering TNBC spreading and invasion [74] [43. In the same way, TNF-α has been seen to exert a tumor-promoting role in BC progression and induce metastasis, fostering tumor escape from immune system control [75] [44. Nonetheless, some studies have recently addressed a controversial role of TNF-α, showing pro-apoptotic and anticarcinogenic functions towards different tumor types, which could also be dependent on TME or specific conditions such as the TNFR that controls the pathway or the ER/PR molecular BC type [76,77] [45][46.

2. A Common Strategy in Cancer and DIO: Targeting Cell-Cycle Progression/CDKs

Several anti-obesity drugs are being tested for their potential interest as antitumoral agents, including lipid-lowering agents [88] [47. For instance, the antihyperlipidemic agent orlistat has extensively proven to induce S-phase cell-cycle arrest and apoptosis in BC [89] [48; whereas recently Harborg et al. carried out a cohort study that explored the link between the use of statins and the risk of developing BC, confirming an indirect relationship between them in postmenopausal early BC patients [90] [49. Similarly, a noticeable 19% decrease in the OC demises was also noticed in a parallel study comparing mortality among statin users versus patients who never took statins before, with a major benefit assigned to Simvastatin [91] [50. Furthermore, statin therapy not only did not entail a comparable toxic profile versus chemotherapy, but evidence also supports the ability of these antilipidemic drugs to promote apoptosis in malignant cells, reducing cancer progression and invasiveness [92] [51.
As one of the primordial purposes of oncosuppressors is to avoid cell-cycle progression by directly altering cyclin-dependent kinases expression, and CDK proteins are important players in cell-cycle modulation cascades, novel CDK inhibitors-based strategies have been proposed not only for the management of cancer but also for DIO and vice versa. The activity of these serine/threonine protein kinases is highly dependent on the activation of phase-specific cyclins, and the employment of CDKIs has emerged as an innovative strategy in tumor treatment [25,93,94] [52][53][54. In the same way, multiple plant-derived biomolecules and by-products have shown CDK inhibitory functions raising interest as antitumoral agents [95,96,97] [55][56][57.

2.1. Food-Based Approaches in Cancer Therapy

Many anti-DIO strategies based on food intake time restrictions are being tested in vivo to better understand how specific nutritional deprivations affect different types of malignancies. These include several types of periodic fasting and intermittent food supply diets such as time-restricted feeding, short-restricted fasting, short-term starvation, alternate-day fasting, or fasting-mimicking diet (FMD). These fasting and dietary limitations are showing encouraging results in the management of obesity, notably impairing chronic disease burden and cancer onset [98,99] [58][59.
Moreover, these strategies also represent a good and safe alternative that minimally affects non-tumoral cells, while selectively altering the survival chances of neoplastic cells, mainly by decreasing insulin and related factors, glucose, leptin, and cytokines [100] [60.
The ratio and specific type of macronutrients assumed can importantly change the course of the disease. Dietary patterns with a high content of animal-based proteins were correlated with a major risk of cancer demises compared with feeding habits mainly entailing vegetable-derived proteins [101] [61. Following this thought, plant-based nutraceuticals are bringing attention as antitumor strategies, and micronutrients and phytochemicals of particular interest are undergoing clinical and preclinical trials in the cancer field. 
To exemplify, more than 50 CTs have investigated the beneficial properties of broccoli-derived molecules (mainly sulforaphane and glucoraphanin) in cancer disease remission [102,103] [62][63. Nevertheless, only a few of them have brought into focus the molecular mechanisms involving the downregulation of cell cycle-related proteins such as cyclins and CDKs [104] [64, or the induction of CDKIs and correlated pathways involving signaling cascades such as the mammalian target of rapamycin (mTOR) [105] [65 or STAT 3[106]3[66.
Indole-3-carbinol (I3C) represents an additional natural anticancer agent belonging to the same broccoli vegetable family (Brassicaceae). It was found to block G1/S cell-cycle progression in breast and endometrial cancers, including MCF-7, BT20, and MDA-MB-231 cell lines [121] [67. The effective reduction of cyclins D1, E, CDK-2, -4, and -6 and the increase of p21, p27, and p15 expression were also validated in response to I3C treatment [97,122] [57][68.
Roscovitine constitutes another biological molecule employed in anti-DIO therapy with a key role in cell-cycle modulation. Specifically, its activity results in an accumulation of cells in the G2 phase on (ER-α)+ MCF-7 breast cancer cells, preventing them from entering the next cell cycle [123] [69.
Concomitantly to the inhibition of cell-cycle progression, roscovitine—later commercialized as Seliciclib®, a first-generation CDKI—showed a remarkable ability to induce apoptosis via a p53-dependent pathway [124] [70. Additionally, fangchinoline, an alkaloid isolated from the Menispermaceae plant family, has been seen to impede G1/S cell cycle transition in MDA-MB-231 and MCF-7 breast cancer cells. The cell cycle blocking effects of fangchinoline alkaloid were further confirmed by a drop in the levels of cyclins D1, D3, and E; CDK-2, -4, and -6, as well as an increased expression of CDKIs p21 and p27 tumor suppressor proteins [125] [71.
The alkaloid Berberine has proved to have cytotoxic and antiproliferative actions in BC [126] [72 and OC cells [127 [73][74,128], by targeting the Akt downstream pathway, whereas the flavonoid quercetin (Quercus sp.) was able to stop the cell cycle at G1/S and G2/M checkpoints. Downregulation of Quercetin-3-methyl ether significantly prompted cell-cycle arrest at the G2-M phase in MDA-MB-231 and MCF-7 human BC cells, decreasing cell proliferation, invasion, and migration and inducing apoptosis [129,130] [75][76. A fall in CDK-2, -6, -7, and cyclins A, D1, and E were also confirmed [131] [77.
Curcumin is widely known to promote cell-cycle arrest at G1/S and G2/M phases and to stimulate the expression of tumor suppressor proteins p53, and the p21 and p27 endogenous CDKIs [132,133] [78][79. Preclinical studies indicated a beneficial effect of this Curcuma longa-derived polyphenol in reducing severe skin side effects of radiotherapy in BC patients [134] [80. Furthermore, a synergistic apoptotic action via PARP and p53 activation was seen in combined therapies of curcumin and citral extract, as well as an activation of the oxidative stress signaling via ROS production [135] [81.

2.2. Cyclin-Dependent Kinase Inhibitors as Anticancer Drugs

Flavopiridol was the first and most extensively studied CDK inhibitor entering human clinical trials to treat various cancer types, including breast, lung, and bladder [138,139] [82][83. This non-selective CDK inhibitor alkaloid initially proved to induce cell-cycle arrest in G0/G1 and an S-phase delay, showing a high specificity against the CDK1/cyclin B complex in BC [140] [84. Even if the efficacy of flavopiridol in vivo has not been demonstrated to be sufficient to enable it to enter Phase III trials [139] [83 since the FDA approval of this CDKi as an orphan drug for acute myeloid leukemia in 2015, a larger set of molecules have entered clinical testing to evaluate their feasibility in the cancer treatment approach. Indeed, flavopiridol has opened a new window of opportunity for next-generation CDKIs, which means a higher drug specificity by the abolition of cyclin/CDK binding, which consequently impedes the protein complex-associated kinase activity and the subsequent cell-cycle progression. Among these new CDKIs, selective inhibitors of CDK4/6 are particularly gaining the major focus of interest [141] [85, whose activation is mainly dependent on cyclin D-type linkage.
Additional second-generation CDKIs include dinaciclib, a potent inhibitor that targets CDK1, CDK2, CDK5, and CDK9. Despite contrasting results arising, a Phase III randomized study revealed enthralling results in refractory leukemia patients in terms of efficacy, safety, and progression-free survival (PFS) [142] [86. Moreover, in vitro tests guaranteed the major ability of dinaciclib to suppress Rb phosphorylation versus flavopiridol, subsequently validating a notable cell-cycle arrest in a huge number of malignant cell-based assays [143] [87. CDK7 inhibitors are also emerging as anticancer therapeutic drugs by targeting diverse pathways, chiefly involving cell-cycle regulators such as CDK-activating kinase, that finally hinder the initiation of oncogenic transcription [93,144] [53][88. To date, four different CDK7i are under Phase II studies with encouraging outcomes in breast [145] [89 and ovarian cancers [146,147] [90][91.
CDK-4 has been identified as a potential blocking target in diet-related anti-obesity treatment, as it promotes adipogenicity [148] [92. The previously mentioned work of Iqbal and co-workers also described how lipid-enriched diets can induce pRb phosphorylation in the hypothalamus, which consequently inactivates the protein and promotes obesity in vivo. In fact, experiments carried out in mice treated with a first-generation CDKI—abemaciclib—have reported promising results in fat mass reduction and weight loss, and future assays are aimed to deduct which are the molecular mechanisms that may link the abrogation of CDK4/6 and the unphosphorylated form of pRb with the blockade of DIO in neurons [22] [93.
Interestingly, three different CDKIs 4/6 have recently been FDA approved for the treatment of lifelong aggressive and refractory HR+, Her2- BC therapy (palbociclib, PD0332991; ribociclib, LEE011; abemaciclib, LY835219) [149,150,151] [94][95][96. Several biomolecular pathways induced during the obese condition have been demonstrated to be in common with cancer mechanisms of tumor evasion, prompting the study of CDK inhibitors for the management of obesity disease [22] [93. With this scope, a common strategy linking FDA-authorized CDKIs, nutraceuticals, and dietary approaches could become a feasible tactic to handle overweight-related problems that may potentially favor cancer development.
Palbociclib (PD-0332991) was the first CDKI 4/6 demonstrating a substantial efficacy against breast cancer cells in combination with endocrine therapy in ER+ tumor models in vitro [152] [97. From 2009 onwards, palbociclib has undergone successful studies in concomitance with hormonal therapies—HT—that led to its FDA approval in 2015 [153] [98, including selective estrogen receptor degraders (SERDs) [154] [99, aromatase inhibitors—AIs—[155][100, and fulvestrant [156] [101. Some CTs also show the activity of this CDK4/6 blocker as a single agent both in ovarian [157] [102 and in metastatic breast cancers [158] [103, illustrating a good drug side-effect profile.
Cytotoxicity was dose-dependent but showed some variability from one cell line to another; moreover, a direct correlation between p16 hypoexpression, high RB1 levels, and a significant response to palbociclib treatment was verified, both in vitro and in a clinical cohort of 263 OC patients. Inhibition of RB1 phosphorylation and promotion of G1 cell-cycle arrest and apoptosis further supported the promising use of palbociclib in OC, also in later clinical studies [160] [104.
Another noteworthy observation was carried out in TNBC models, a highly aggressive BC subtype characterized by the lack of expression of targetable receptors and a rapid tendency to metastasize to lungs, brain, and bones.
Abemaciclib exhibited the highest potency and best delivery efficiency among the three next-generation CDKIs, also showing effects on other kinases such as CDK9 and PIM1 [162,163] [105][106. Patnaik et al. (2016) performed preclinical studies in OC human xenografts and patients undergoing abemaciclib therapy [164] [107. Promising results showed a good safety profile and clinical significance for this CDK 4/6 inhibitor, and a favorable and extended CA-125 response to treatment in advanced OC models. Novel studies concerning abemaciclib monotherapy have also been conducted in HR+/Her2- MBC patients who become refractory to endocrine therapy. Among these, the MONARCH-1 trial showed an overall response that accounted for 19.7% of total enrolled patients, whereas clinical benefit exceeded the 42% [165] [108, additionally confirming the antitumor activity and manageable toxicity profile of abemaciclib administered alone [166] [109.
In 2018, the MONALEESA-3 clinical trial outcomes prompted the FDA approval of ribociclib plus fulvestrant in postmenopausal HR+, Her2- advanced BC patients [182] [110. Following studies of Iyengar and co-workers evaluated the effectiveness of selective CDK4/6 inhibitor ribociclib in different models of high-grade serous ovarian cancer both in vitro and in vivo and identified a pivotal and very selective dose-response activity against cancer cells viability [186] [111. Cytotoxicity was even more evident upon ribociclib plus cisplatin association, showing a pronounced synergism in co-treatment therapies. The wide ability of CDK4/6 inhibitors to impede cell-cycle progression through the G0/G1 phase was confirmed, whereas the accumulation of cells at the G2/M phase suggested a potential role of ribociclib at this checkpoint as well. Interestingly, the addition of ribociclib also prevented cisplatin chemotherapy-surviving cells to progress over the G2/M cell-cycle phase. Combinations of ribociclib and letrozole also revealed promising results in early HR+ mammary tumors, identifying CDKIs as valuable alternatives to reduce relapses and side effects derived from ET [184,187] [112][113.
So as the main mechanism underlying G1-targeted CDK4/6 inhibitors go through avoiding RB1 tumor suppressor phosphorylation and its subsequent inactivation, the effects of some agents such as palbociclib require the presence of a functional pRb protein to work properly [188] [114. Cancers presenting deletions at the pRb protein level represent a treatment challenge, as the lack of a functional target makes these tumors resistant to CDKIs 4/6, making single-agent therapy ineffective [189] [115.
For this reason, CDK 4/6 inhibitors are presently experiencing multiple large, randomized clinical trials to test the prospective combined approaches with anti-estrogen and hormonal therapies [153,171,190,191] [98][116][117][118. Actually, therapies targeting ER such as tamoxifen, aromatase inhibitors, or fulvestrant that also affect cyclin D1 expression and promote G1 phase cell accumulation may potentiate the blocking function of a CDK4/6 inhibitor in cell-cycle progression [153,155,173] [98][100][119. As a matter of fact, cyclin D1 has strongly exhibited a main role in the development of Her2-driven breast tumors [192] [120.
Intriguing early stage trials are also investigating possible CDKIs and PI3K inhibitors combinations in TNBC. Both palbociclib and ribociclib were administered together with taselisib/alpelisib—respectively—showing a greater synergistic response in terms of cell-cycle arrest and apoptosis compared to single-agent use [193,194] [121][122.
There are several ongoing and completed Phase II/III studies testing dual CDK4/6 inhibitors against breast and ovarian carcinoma, further verifying the antitumor effectiveness of this group of drugs, some of which already manifesting superiority over ET in monotherapy. Additional trials combining palbociclib with other agents (e.g., capecitabine) did not exert comparable results in terms of clinical benefit, QoL, and safety profile [195] [123.

References

  1. Blücher, C.; Stadler, S.C. Obesity and Breast Cancer: Current Insights on the Role of Fatty Acids and Lipid Metabolism in Promoting Breast Cancer Growth and Progression. Front. Endocrinol. 2017, 8, 293.
  2. Wu, Q.; Li, B.; Li, Z.; Li, J.; Sun, S.; Sun, S. Cancer-associated adipocytes: Key players in breast cancer progression. J. Hematol. Oncol. 2019, 12, 1–15.
  3. Olsen, C.M.; Nagle, C.M.; Whiteman, D.C.; Ness, R.; Pearce, C.L.; Pike, M.C.; Rossing, M.A.; Terry, K.L.; Wu, A.H.; A Risch, H.; et al. Obesity and risk of ovarian cancer subtypes: Evidence from the Ovarian Cancer Association Consortium. Endocr. Relat. Cancer 2013, 20, 251–262.
  4. Howe, L.R.; Subbaramaiah, K.; Hudis, C.A.; Dannenberg, A.J. Molecular Pathways: Adipose Inflammation as a Mediator of Obesity-Associated Cancer. Clin. Cancer Res. 2013, 19, 6074–6083.
  5. Kwon, O.; Kim, K.W.; Kim, M.S. Leptin signalling pathways in hypothalamic neurons. Cell. Mol. Life Sci. 2016, 73, 1457–1477.
  6. Ptak, A.; Kolaczkowska, E.; Gregoraszczuk, E.L. Leptin stimulation of cell cycle and inhibition of apoptosis gene and protein expression in OVCAR-3 ovarian cancer cells. Endocrine 2013, 43, 394–403.
  7. Newman, G.; Gonzalez-Perez, R.R. Leptin–cytokine crosstalk in breast cancer. Mol. Cell. Endocrinol. 2014, 382, 570–582.
  8. Carpenter, B.; Hemsworth, G.R.; Wu, Z.; Maamra, M.; Strasburger, C.J.; Ross, R.J.; Artymiuk, P.J. Structure of the Human Obesity Receptor Leptin-Binding Domain Reveals the Mechanism of Leptin Antagonism by a Monoclonal Antibody. Structure 2012, 20, 487–497.
  9. Catalano, S.; Leggio, A.; Barone, I.; De Marco, R.; Gelsomino, L.; Campana, A.; Malivindi, R.; Panza, S.; Giordano, C.; Liguori, A.; et al. A novel leptin antagonist peptide inhibits breast cancer growth in vitro and in vivo. J. Cell. Mol. Med. 2015, 19, 1122–1132.
  10. Catalano, S.; Mauro, L.; Bonofiglio, D.; Pellegrino, M.; Qi, H.; Rizza, P.; Vizza, D.; Bossi, G.; Andò, S. In Vivo and in Vitro Evidence That PPARγ Ligands Are Antagonists of Leptin Signaling in Breast Cancer. Am. J. Pathol. 2011, 179, 1030–1040.
  11. Atoum, M.F.; Alzoughool, F.; Al-Hourani, H. Linkage Between Obesity Leptin and Breast Cancer. Breast Cancer Basic Clin. Res. 2020, 14, 1–8.
  12. Otvos, L.; Kovalszky, I.; Riolfi, M.; Ferla, R.; Olah, J.; Sztodola, A.; Nama, K.; Molino, A.; Piubello, Q.; Wade, J.D.; et al. Efficacy of a leptin receptor antagonist peptide in a mouse model of triple-negative breast cancer. Eur. J. Cancer 2011, 47, 1578–1584.
  13. Lipsey, C.C.; Harbuzariu, A.; Daley-Brown, D.; Gonzalez-Perez, R.R. Oncogenic role of leptin and Notch interleukin-1 leptin crosstalk outcome in cancer. World J. Methodol. 2016, 6, 43–55.
  14. Harmon, T.; Harbuzariu, A.; Yang, L.; Gonzalez-Perez, R.R. 267 Novel adjuvant therapy with leptin peptide receptor antagonist-2 conjugated to nanoparticles (IONP-LPrA2) to minimize chemoresistance in triple negative breast cancer. Eur. J. Cancer 2014, 50, 89.
  15. Giordano, C.; Chemi, F.; Panza, S.; Barone, I.; Bonofiglio, D.; Lanzino, M.; Cordella, A.; Campana, A.; Hashim, A.; Rizza, P.; et al. Leptin as a mediator of tumor-stromal interactions promotes breast cancer stem cell activity. Oncotarget 2016, 7, 1262–1275.
  16. Barone, I.; Giordano, C.; Bonofiglio, D.; Andò, S.; Catalano, S. Leptin, obesity and breast cancer: Progress to understanding the molecular connections. Curr. Opin. Pharmacol. 2016, 31, 83–89.
  17. Bowers, L.W.; Rossi, E.L.; McDonell, S.B.; Doerstling, S.S.; Khatib, S.A.; Lineberger, C.G.; Albright, J.E.; Tang, X.; Degraffenried, L.A.; Hursting, S.D. Leptin Signaling Mediates Obesity-Associated CSC Enrichment and EMT in Preclinical TNBC Models. Mol. Cancer Res. 2018, 16, 869–879.
  18. Lane, D.; Matte, I.; Garde-Granger, P.; Laplante, C.; Carignan, A.; Rancourt, C.; Piché, A. Inflammation-regulating factors in ascites as predictive biomarkers of drug resistance and progression-free survival in serous epithelial ovarian cancers. BMC Cancer 2015, 15, 1–11.
  19. Wang, T.; Fahrmann, J.F.; Lee, H.; Li, Y.-J.; Tripathi, S.C.; Yue, C.; Zhang, C.; Lifshitz, V.; Song, J.; Yuan, Y.; et al. JAK/STAT3-Regulated Fatty Acid β-Oxidation Is Critical for Breast Cancer Stem Cell Self-Renewal and Chemoresistance. Cell Metab. 2018, 27, 136–150.e5.
  20. Di Zazzo, E.; Feola, A.; Zuchegna, C.; Romano, A.; Donini, C.F.; Bartollino, S.; Costagliola, C.; Frunzio, R.; Laccetti, P.; Di Domenico, M.; et al. The p85 Regulatory Subunit of PI3K Mediates cAMP-PKA and Insulin Biological Effects on MCF-7 Cell Growth and Motility. Sci. World J. 2014, 2014, 565839.
  21. Di Zazzo, E.; Polito, R.; Bartollino, S.; Nigro, E.; Porcile, C.; Bianco, A.; Daniele, A.; Moncharmont, B. Adiponectin as Link Factor between Adipose Tissue and Cancer. Int. J. Mol. Sci. 2019, 20, 839.
  22. Katira, A.; Tan, P.H. Evolving role of adiponectin in cancer-controversies and update. Cancer Biol. Med. 2016, 13, 101–119.
  23. Mauro, L.; Pellegrino, M.; De Amicis, F.; Ricchio, E.; Giordano, F.; Rizza, P.; Catalano, S.; Bonofiglio, D.; Sisci, D.; Panno, M.L.; et al. Evidences that estrogen receptor α interferes with adiponectin effects on breast cancer cell growth. Cell Cycle 2014, 13, 553–564.
  24. Dieudonne, M.N.; Bussiere, M.; dos Santos, E.; Leneveu, M.C.; Giudicelli, Y.; Pecquery, R. Adiponectin mediates antiproliferative and apoptotic responses in human MCF7 breast cancer cells. Biochem. Biophys. Res. Commun. 2006, 345, 271–279.
  25. Diaz, E.S.; Karlan, B.Y.; Li, A.J. Obesity-associated adipokines correlate with survival in epithelial ovarian cancer. Gynecol. Oncol. 2013, 129, 353–357.
  26. Lawrence, T. Inflammation and cancer: A failure of resolution? Trends Pharmacol. Sci. 2007, 28, 162–165.
  27. Lan, T.; Chen, L.; Wei, X. Inflammatory Cytokines in Cancer: Comprehensive Understanding and Clinical Progress in Gene Therapy. Cells 2021, 10, 100.
  28. Donovan, J.; Slingerland, J. Transforming growth factor-β and breast cancer: Cell cycle arrest by transforming growth factor-β and its disruption in cancer. Breast Cancer Res. 2000, 2, 116–124.
  29. Batlle, E.; Massagué, J. Transforming Growth Factor-β Signaling in Immunity and Cancer. Immunity 2019, 50, 924–940.
  30. Murugaiyan, G.; Saha, B. Protumor vs Antitumor Functions of IL-17. J. Immunol. 2009, 183, 4169–4175.
  31. Eiró, N.; González, L.; González, L.O.; Fernandez-Garcia, B.; Lamelas, M.L.; Marín, L.; González-Reyes, S.; del Casar, J.M.; Vizoso, F.J. Relationship between the Inflammatory Molecular Profile of Breast Carcinomas and Distant Metastasis Development. PLoS ONE 2012, 7, e49047.
  32. Macciò, A.; Madeddu, C.; Gramignano, G.; Mulas, C.; Floris, C.; Massa, D.; Astara, G.; Chessa, P.; Mantovani, G. Correlation of body mass index and leptin with tumor size and stage of disease in hormone-dependent postmenopausal breast cancer: Preliminary results and therapeutic implications. J. Mol. Med. 2010, 88, 677–686.
  33. Pereira, J.F.S.; Bessa, C.; Matos, P.; Jordan, P. Pro-Inflammatory Cytokines Trigger the Overexpression of Tumour-Related Splice Variant RAC1B in Polarized Colorectal Cells. Cancers 2022, 14, 1393.
  34. Browning, L.; Patel, M.R.; Horvath, E.B.; Tawara, K.; Jorcyk, C.L. IL-6 and ovarian cancer: Inflammatory cytokines in promotion of metastasis. Cancer Manag. Res. 2018, 10, 6685–6693.
  35. Deng, F.; Weng, Y.; Li, X.; Wang, T.; Fan, M.; Shi, Q. Overexpression of IL-8 promotes cell migration via PI3K-Akt signaling pathway and EMT in triple-negative breast cancer. Pathol. Res. Pract. 2020, 216, 152902.
  36. Li, Z.; Jiang, J.; Wang, Z.; Zhang, J.; Xiao, M.; Wang, C.; Lu, Y.; Qin, Z. Endogenous Interleukin-4 Promotes Tumor Development by Increasing Tumor Cell Resistance to Apoptosis. Cancer Res. 2008, 68, 8687–8694.
  37. Heim, L.; Yang, Z.; Tausche, P.; Hohenberger, K.; Chiriac, M.T.; Koelle, J.; Geppert, C.-I.; Kachler, K.; Miksch, S.; Graser, A.; et al. IL-9 Producing Tumor-Infiltrating Lymphocytes and Treg Subsets Drive Immune Escape of Tumor Cells in Non-Small Cell Lung Cancer. Front. Immunol. 2022, 13, 859783.
  38. Toney, N.J.; Opdenaker, L.M.; Cicek, K.; Frerichs, L.; Kennington, C.R.; Oberly, S.; Archinal, H.; Somasundaram, R.; Sims-Mourtada, J. Tumor-B-cell interactions promote isotype switching to an immunosuppressive IgG4 antibody response through upregulation of IL-10 in triple negative breast cancers. J. Transl. Med. 2022, 20, 1–15.
  39. Ahmad, N.; Ammar, A.; Storr, S.J.; Green, A.R.; Rakha, E.; Ellis, I.O.; Martin, S.G. IL-6 and IL-10 are associated with good prognosis in early stage invasive breast cancer patients. Cancer Immunol. Immunother. 2018, 67, 537–549.
  40. Itakura, E.; Huang, R.R.; Wen, D.R.; Paul, E.; Wünsch, P.H.; Cochran, A.J. IL-10 expression by primary tumor cells correlates with melanoma progression from radial to vertical growth phase and development of metastatic competence. Mod. Pathol. 2011, 24, 801–809.
  41. Zhao, S.; Wu, D.; Wu, P.; Wang, Z.; Huang, J.; Gao, J.X. Serum IL-10 Predicts Worse Outcome in Cancer Patients: A Meta-Analysis. PLoS ONE 2015, 10, e0139598.
  42. Liubomirski, Y.; Lerrer, S.; Meshel, T.; Morein, D.; Rubinstein-Achiasaf, L.; Sprinzak, D.; Wiemann, S.; Körner, C.; Ehrlich, M.; Ben-Baruch, A. Notch-Mediated Tumor-Stroma-Inflammation Networks Promote Invasive Properties and CXCL8 Expression in Triple-Negative Breast Cancer. Front. Immunol. 2019, 10, 804.
  43. Liubomirski, Y.; Ben-Baruch, A. Notch-Inflammation Networks in Regulation of Breast Cancer Progression. Cells 2020, 9, 1576.
  44. Al-Hatamleh, M.A.I.; Ahmad, S.; Boer, J.; Lim, J.; Chen, X.; Plebanski, M.; Mohamud, R. A Perspective Review on the Role of Nanomedicine in the Modulation of TNF-TNFR2 Axis in Breast Cancer Immunotherapy. J. Oncol. 2019, 2019, 6313242.
  45. Gubernatorova, E.; Polinova, A.; Petropavlovskiy, M.; Namakanova, O.; Medvedovskaya, A.; Zvartsev, R.; Telegin, G.; Drutskaya, M.; Nedospasov, S. Dual Role of TNF and LTα in Carcinogenesis as Implicated by Studies in Mice. Cancers 2021, 13, 1775.
  46. Cruceriu, D.; Baldasici, O.; Balacescu, O.; Berindan-Neagoe, I. The dual role of tumor necrosis factor-alpha (TNF-α) in breast cancer: Molecular insights and therapeutic approaches. Cell. Oncol. 2020, 43, 1–18.
  47. Di Bello, E.; Zwergel, C.; Mai, A.; Valente, S. The Innovative Potential of Statins in Cancer: New Targets for New Therapies. Front. Chem. 2020, 8, 516.
  48. Menendez, J.A.; Vellon, L.; Lupu, R. Antitumoral actions of the anti-obesity drug orlistat (Xenical™) in breast cancer cells: Blockade of cell cycle progression, promotion of apoptotic cell death and PEA3-mediated transcriptional repression of Her2/neu (erbB-2) oncogene. Ann. Oncol. 2005, 16, 1253–1267.
  49. Harborg, S.; Heide-Jørgensen, U.; Ahern, T.P.; Ewertz, M.; Cronin-Fenton, D.; Borgquist, S. Statin use and breast cancer recurrence in postmenopausal women treated with adjuvant aromatase inhibitors: A Danish population-based cohort study. Breast Cancer Res. Treat. 2020, 183, 153–160.
  50. Couttenier, A.; Lacroix, O.; Vaes, E.; Cardwell, C.R.; de Schutter, H.; Robert, A. Statin use is associated with improved survival in ovarian cancer: A retrospective population-based study. PLoS ONE 2017, 12, e0189233.
  51. Beckwitt, C.H.; Brufsky, A.; Oltvai, Z.N.; Wells, A. Statin drugs to reduce breast cancer recurrence and mortality. Breast Cancer Res. 2018, 20, 1–11.
  52. Law, M.E.; Corsino, P.E.; Narayan, S.; Law, B.K. Cyclin-Dependent Kinase Inhibitors as Anticancer Therapeutics. Mol. Pharmacol. 2015, 88, 846–852.
  53. Sava, G.P.; Fan, H.; Coombes, R.C.; Buluwela, L.; Ali, S. CDK7 inhibitors as anticancer drugs. Cancer Metastasis Rev. 2020, 39, 805–823.
  54. Mayer, E.L. Targeting Breast Cancer with CDK Inhibitors. Curr. Oncol. Rep. 2015, 17, 1–5.
  55. Tu, Y.; Kim, E.; Gao, Y.; Rankin, G.O.; Li, B.; Chen, Y.C. Theaflavin-3, 3′-digallate induces apoptosis and G2 cell cycle arrest through the Akt/MDM2/p53 pathway in cisplatin-resistant ovarian cancer A2780/CP70 cells. Int. J. Oncol. 2016, 48, 2657–2665.
  56. Gorzynik-Debicka, M.; Przychodzen, P.; Cappello, F.; Kuban-Jankowska, A.; Marino Gammazza, A.; Knap, N.; Wozniak, M.; Gorska-Ponikowska, M. Potential Health Benefits of Olive Oil and Plant Polyphenols. Int. J. Mol. Sci. 2018, 19, 686.
  57. Bailon-Moscoso, N.; Cevallos-Solorzano, G.; Romero-Benavides, J.; Ramirez Orellana, M. Natural Compounds as Modulators of Cell Cycle Arrest: Application for Anticancer Chemotherapies. Curr. Genom. 2017, 18, 106–131.
  58. Woodie, L.N.; Johnson, R.M.; Ahmed, B.; Fowler, S.; Haynes, W.; Carmona, B.; Reed, M.; Suppiramaniam, V.; Greene, M.W. Western diet-induced obesity disrupts the diurnal rhythmicity of hippocampal core clock gene expression in a mouse model. Brain Behav. Immun. 2020, 88, 815–825.
  59. Longo, V.D.; Panda, S. Fasting, circadian rhythms, and time-restricted feeding in healthy lifespan. Cell Metab. 2016, 23, 1048–1059.
  60. Buono, R.; Longo, V.D. Starvation, Stress Resistance, and Cancer. Trends Endocrinol. Metab. 2018, 29, 271–280.
  61. Levine, M.E.; Suarez, J.A.; Brandhorst, S.; Balasubramanian, P.; Cheng, C.-W.; Madia, F.; Fontana, L.; Mirisola, M.G.; Guevara-Aguirre, J.; Wan, J.; et al. Low Protein Intake Is Associated with a Major Reduction in IGF-1, Cancer, and Overall Mortality in the 65 and Younger but Not Older Population. Cell Metab. 2014, 19, 407–417.
  62. Alumkal, J.J.; Slottke, R.; Schwartzman, J.; Cherala, G.; Munar, M.; Graff, J.N.; Beer, T.M.; Ryan, C.W.; Koop, D.R.; Gibbs, A.; et al. A phase II study of sulforaphane-rich broccoli sprout extracts in men with recurrent prostate cancer. Investig. New Drugs 2015, 33, 480–489.
  63. Amjad, A.I.; Parikh, R.A.; Appleman, L.J.; Hahm, E.R.; Singh, K.; Singh, S.V. Broccoli-Derived Sulforaphane and Chemoprevention of Prostate Cancer: From Bench to Bedside. Curr. Pharmacol. Rep. 2015, 1, 382–390.
  64. Kim, J.-H.; Kwon, K.H.; Jung, J.-Y.; Han, H.-S.; Shim, J.H.; Oh, S.; Choi, K.-H.; Choi, E.-S.; Shin, J.-A.; Leem, D.-H.; et al. Sulforaphane Increases Cyclin-Dependent Kinase Inhibitor, p21 Protein in Human Oral Carcinoma Cells and Nude Mouse Animal Model to Induce G2/M Cell Cycle Arrest. J. Clin. Biochem. Nutr. 2010, 46, 60–67.
  65. Justin, S.; Rutz, J.; Maxeiner, S.; Chun, F.K.H.; Juengel, E.; Blaheta, R.A. Chronic Sulforaphane Administration Inhibits Resistance to the mTOR-Inhibitor Everolimus in Bladder Cancer Cells. Int. J. Mol. Sci. 2020, 21, 4026.
  66. Simões, B.M.; Santiago-Gómez, A.; Chiodo, C.; Moreira, T.; Conole, D.; Lovell, S.; Alferez, D.; Eyre, R.; Spence, K.; Sarmiento-Castro, A.; et al. Targeting STAT3 signaling using stabilised sulforaphane (SFX-01) inhibits endocrine resistant stem-like cells in ER-positive breast cancer. Oncogene 2020, 39, 4896–4908.
  67. Cover, C.M.; Hsieh, S.J.; Tran, S.H.; Hallden, G.; Kim, G.S.; Bjeldanes, L.F.; Firestone, G.L. Indole-3-carbinol Inhibits the Expression of Cyclin-dependent Kinase-6 and Induces a G1 Cell Cycle Arrest of Human Breast Cancer Cells Independent of Estrogen Receptor Signaling. J. Biol. Chem. 1998, 273, 3838–3847.
  68. Aggarwal, B.B.; Ichikawa, H. Molecular Targets and Anticancer Potential of Indole-3-Carbinol and Its Derivatives. Cell Cycle 2005, 4, 1201–1215.
  69. Wang, H.; Liu, L.; Lin, J.Z.; Aprahamian, T.R.; Farmer, S.R. Browning of White Adipose Tissue with Roscovitine Induces a Distinct Population of UCP1 + Adipocytes. Cell Metab. 2016, 24, 835–847.
  70. Wȩsierska-Ga̧dek, J.; Gritsch, D.; Zulehner, N.; Komina, O.; Maurer, M. Interference with ER-α enhances the therapeutic efficacy of the selective CDK inhibitor roscovitine towards ER-positive breast cancer cells. J. Cell. Biochem. 2011, 112, 1103–1117.
  71. Xing, Z.; Zhang, Y.; Zhang, X.; Yang, Y.; Ma, Y.; Pang, D. Fangchinoline Induces G1 Arrest in Breast Cancer Cells Through Cell-Cycle Regulation. Phytotherapy Res. 2013, 27, 1790–1794.
  72. Kim, J.; Yu, J.-H.; Ko, E.; Lee, K.-W.; Song, A.; Park, S.; Shin, I.; Han, W.; Noh, D. The alkaloid Berberine inhibits the growth of Anoikis-resistant MCF-7 and MDA-MB-231 breast cancer cell lines by inducing cell cycle arrest. Phytomedicine 2010, 17, 436–440.
  73. Chen, Q.; Qin, R.; Fang, Y.; Li, H. Berberine Sensitizes Human Ovarian Cancer Cells to Cisplatin Through miR-93/PTEN/Akt Signaling Pathway. Cell. Physiol. Biochem. 2015, 36, 956–965.
  74. Jin, P.; Zhang, C.; Li, N. Berberine Exhibits Antitumor Effects in Human Ovarian Cancer Cells. Anti-Cancer Agents Med. Chem. 2015, 15, 511–516.
  75. Cao, L.; Yang, Y.; Ye, Z.; Lin, B.; Zeng, J.; Li, C.; Liang, T.; Zhou, K.; Li, J. Quercetin-3-methyl ether suppresses human breast cancer stem cell formation by inhibiting the Notch1 and PI3K/Akt signaling pathways. Int. J. Mol. Med. 2018, 42, 1625–1636.
  76. Li, J.; Zhu, F.; Lubet, R.A.; De Luca, A.; Grubbs, C.; Ericson, M.E.; D'Alessio, A.; Normanno, N.; Dong, Z.; Bode, A.M. Quercetin-3-methyl ether inhibits lapatinib-sensitive and -resistant breast cancer cell growth by inducing G2 /M arrest and apoptosis. Mol. Carcinog. 2013, 52, 134–143.
  77. Meeran, S.M.; Katiyar, S.K. Cell cycle control as a basis for cancer chemoprevention through dietary agents. Front. Biosci. 2008, 13, 2191–2202.
  78. Giordano, A.; Tommonaro, G. Curcumin and Cancer. Nutrients 2019, 11, 2376.
  79. Hu, S.; Xu, Y.; Meng, L.; Huang, L.; Sun, H. Curcumin inhibits proliferation and promotes apoptosis of breast cancer cells. Exp. Ther. Med. 2018, 16, 1266–1272.
  80. Ryan, J.L.; Heckler, C.E.; Ling, M.; Katz, A.; Williams, J.P.; Pentland, A.P.; Morrow, G.R. Curcumin for Radiation Dermatitis: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial of Thirty Breast Cancer Patients. Radiat. Res. 2013, 180, 34–43.
  81. Patel, P.B.; Thakkar, V.R.; Patel, J.S. Cellular Effect of Curcumin and Citral Combination on Breast Cancer Cells: Induction of Apoptosis and Cell Cycle Arrest. J. Breast Cancer 2015, 18, 225–234.
  82. Senderowicz, A.M. Flavopiridol: The First Cyclin-Dependent Kinase Inhibitor in Human Clinical Trials. Investig. New Drugs 1999, 17, 313–320.
  83. Asghar, U.; Witkiewicz, A.K.; Turner, N.C.; Knudsen, E.S. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat. Rev. Drug Discov. 2015, 14, 130–146.
  84. Tan, A.R.; Swain, S.M. Review of flavopiridol, a cyclin-dependent kinase inhibitor, as breast cancer therapy. Semin. Oncol. 2002, 29, 77–85.
  85. Murphy, C.G.; Dickler, M.N. The Role of CDK4/6 Inhibition in Breast Cancer. Oncologist 2015, 20, 483–490.
  86. Ghia, P.; Scarfò, L.; Perez, S.; Pathiraja, K.; DeRosier, M.; Small, K.; McCrary Sisk, C.; Patton, N. Efficacy and safety of dinaciclib vs ofatumumab in patients with relapsed/refractory chronic lymphocytic leukemia. Blood 2017, 129, 1876–1878.
  87. Parry, D.; Guzi, T.; Shanahan, F.; Davis, N.; Prabhavalkar, D.; Wiswell, D.; Seghezzi, W.; Paruch, K.; Dwyer, M.P.; Doll, R.; et al. Dinaciclib (SCH 727965), a Novel and Potent Cyclin-Dependent Kinase Inhibitor. Mol. Cancer Ther. 2010, 9, 2344–2353.
  88. Hu, S.; Marineau, J.J.; Rajagopal, N.; Hamman, K.B.; Choi, Y.J.; Schmidt, D.R.; Ke, N.; Johannessen, L.; Bradley, M.J.; Orlando, D.A.; et al. Discovery and Characterization of SY-1365, a Selective, Covalent Inhibitor of CDK7. Cancer Res. 2019, 79, 3479–3491.
  89. Patel, H.; Periyasamy, M.; Sava, G.P.; Bondke, A.; Slafer, B.W.; Kroll, S.H.B.; Barbazanges, M.; Starkey, R.; Ottaviani, S.; Harrod, A.; et al. ICEC0942, an Orally Bioavailable Selective Inhibitor of CDK7 for Cancer Treatment. Mol. Cancer Ther. 2018, 17, 1156–1166.
  90. Johannessen, L.H.; Hu, S.; Ke, N.; D'Ippolito, A.; Rajagopal, N.; Marineau, J.; Savinainen, A.; Zamboni, W.; Hodgson, G. Abstract C091: Preclinical evaluation of PK, PD, and antitumor activity of the oral, non-covalent, potent and highly selective CDK7 inhibitor, SY-5609, provides rationale for clinical development in multiple solid tumor indications. Mol. Cancer Ther. 2019, 18, C091.
  91. Zhang, Z.; Peng, H.; Wang, X.; Yin, X.; Ma, P.; Jing, Y.; Cai, M.-C.; Liu, J.; Zhang, M.; Zhang, S.; et al. Preclinical Efficacy and Molecular Mechanism of Targeting CDK7-Dependent Transcriptional Addiction in Ovarian Cancer. Mol. Cancer Ther. 2017, 16, 1739–1750.
  92. Abella, A.; Dubus, P.; Malumbres, M.; Rane, S.G.; Kiyokawa, H.; Sicard, A.; Vignon, F.; Langin, D.; Barbacid, M.; Fajas, L. Cdk4 promotes adipogenesis through PPARγ activation. Cell Metab. 2005, 2, 239–249.
  93. Iqbal, N.J.; Lu, Z.; Liu, S.M.; Schwartz, G.J.; Chua, S.; Zhu, L. Cyclin-dependent kinase 4 is a preclinical target for diet-induced obesity. JCI Insight 2018, 3, e123000.
  94. Walker, A.J.; Wedam, S.; Amiri-Kordestani, L.; Bloomquist, E.; Tang, S.; Sridhara, R.; Chen, W.; Palmby, T.R.; Zirkelbach, J.F.; Fu, W.; et al. FDA Approval of Palbociclib in Combination with Fulvestrant for the Treatment of Hormone Receptor–Positive, HER2-Negative Metastatic Breast Cancer. Clin. Cancer Res. 2016, 22, 4968–4972.
  95. Hamilton, E.; Cortes, J.; Dieras, V.; Ozyilkan, O.; Chen, S.-C.; Petrakova, K.; Manikhas, A.; Jerusalem, G.; Hegg, R.; Lu, Y.; et al. Abstract PD1-11: NextMONARCH 1: Phase 2 study of abemaciclib plus tamoxifen or abemaciclib alone in HR+, HER2- advanced breast cancer. Cancer Res. 2019, 79, PD1-11.
  96. Shah, A.; Bloomquist, E.; Tang, S.; Fu, W.; Bi, Y.; Liu, Q.; Yu, J.; Zhao, P.; Palmby, T.R.; Goldberg, K.B.; et al. FDA Approval: Ribociclib for the Treatment of Postmenopausal Women with Hormone Receptor–Positive, HER2-Negative Advanced or Metastatic Breast Cancer. Clin. Cancer Res. 2018, 24, 2999–3004.
  97. Finn, R.S.; Dering, J.; Conklin, D.; Kalous, O.; Cohen, D.J.; Desai, A.J.; Ginther, C.; Atefi, M.; Chen, I.; Fowst, C.; et al. PD 0332991, a selective cyclin D kinase 4/6 inhibitor, preferentially inhibits proliferation of luminal estrogen receptor-positive human breast cancer cell lines in vitro. Breast Cancer Res. 2009, 11, R77.
  98. Finn, R.S.; Cristofanilli, M.; Ettl, J.; Gelmon, K.A.; Colleoni, M.; Giorgetti, C.; Gauthier, E.; Liu, Y.; Lu, D.R.; Zhang, Z.; et al. Treatment effect of palbociclib plus endocrine therapy by prognostic and intrinsic subtype and biomarker analysis in patients with bone-only disease: A joint analysis of PALOMA-2 and PALOMA-3 clinical trials. Breast Cancer Res. Treat. 2020, 184, 23–35.
  99. Wardell, S.E.; Ellis, M.J.; Alley, H.M.; Eisele, K.; VanArsdale, T.; Dann, S.G.; Arndt, K.T.; Primeau, T.; Griffin, E.; Shao, J.; et al. Efficacy of SERD/SERM Hybrid-CDK4/6 Inhibitor Combinations in Models of Endocrine Therapy–Resistant Breast Cancer. Clin. Cancer Res. 2015, 21, 5121–5130.
  100. Malorni, L.; Curigliano, G.; Minisini, A.M.; Cinieri, S.; Tondini, C.; Arpino, G.; Pavesi, L.; Martignetti, A.; Criscitiello, C.; Puglisi, F.; et al. A phase II trial of the CDK4/6 inhibitor palbociclib (P) as single agent or in combination with the same endocrine therapy (ET) received prior to disease progression, in patients (pts) with hormone receptor positive (HR+) HER2 negative (HER2−) metastatic breast cancer (mBC) (TREnd trial). J. Clin. Oncol. 2017, 35, 1002.
  101. Masuda, N.; Inoue, K.; Nakamura, R.; Rai, Y.; Mukai, H.; Ohno, S.; Hara, F.; Mori, Y.; Hashigaki, S.; Muramatsu, Y.; et al. Palbociclib in combination with fulvestrant in patients with hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer: PALOMA-3 subgroup analysis of Japanese patients. Int. J. Clin. Oncol. 2019, 24, 262–273.
  102. Konecny, G.E.; Hendrickson, A.E.W.; Jatoi, A.; Burton, J.K.; Paroly, J.; Glaspy, J.A.; Dowdy, S.C.; Slamon, D.J. A multicenter open-label phase II study of the efficacy and safety of palbociclib a cyclin-dependent kinases 4 and 6 inhibitor in patients with recurrent ovarian cancer. J. Clin. Oncol. 2016, 34, 5557.
  103. DeMichele, A.; Clark, A.S.; Tan, K.S.; Heitjan, D.F.; Gramlich, K.; Gallagher, M.; Lal, P.; Feldman, M.; Zhang, P.; Colameco, C.; et al. CDK 4/6 Inhibitor Palbociclib (PD0332991) in Rb+Advanced Breast Cancer: Phase II Activity, Safety, and Predictive Biomarker Assessment. Clin. Cancer Res. 2015, 21, 995–1001.
  104. Vaughn, D.J.; Hwang, W.T.; Lal, P.; Rosen, M.A.; Gallagher, M.; O’Dwyer, P.J. Phase 2 trial of the cyclin-dependent kinase 4/6 inhibitor palbociclib in patients with retinoblastoma protein-expressing germ cell tumors. Cancer 2015, 121, 1463–1468.
  105. Gelbert, L.M.; Cai, S.; Lin, X.; Sanchez-Martinez, C.; Del Prado, M.; Lallena, M.J.; Torres, R.; Ajamie, R.T.; Wishart, G.N.; Flack, R.S.; et al. Preclinical characterization of the CDK4/6 inhibitor LY2835219: In-Vivo cell cycle-dependent/independent anti-tumor activities alone/in combination with gemcitabine. Investig. New Drugs 2014, 32, 825–837.
  106. Tate, S.C.; Sykes, A.K.; Kulanthaivel, P.; Chan, E.M.; Turner, P.K.; Cronier, D.M. A Population Pharmacokinetic and Pharmacodynamic Analysis of Abemaciclib in a Phase I Clinical Trial in Cancer Patients. Clin. Pharmacokinet. 2018, 57, 335–344.
  107. Patnaik, A.; Rosen, L.S.; Tolaney, S.M.; Tolcher, A.W.; Goldman, J.W.; Gandhi, L.; Papadopoulos, K.P.; Beeram, M.; Rasco, D.W.; Hilton, J.F.; et al. Efficacy and Safety of Abemaciclib, an Inhibitor of CDK4 and CDK6, for Patients with Breast Cancer, Non-Small Cell Lung Cancer, and Other Solid Tumors. Cancer Discov. 2016, 6, 740–753.
  108. Dickler, M.N.; Tolaney, S.M.; Rugo, H.S.; Cortés, J.; Diéras, V.; Patt, D.; Wildiers, H.; Hudis, C.A.; O'Shaughnessy, J.; Zamora, E.; et al. MONARCH 1, A Phase II Study of Abemaciclib, a CDK4 and CDK6 Inhibitor, as a Single Agent, in Patients with Refractory HR+/HER2− Metastatic Breast Cancer. Clin. Cancer Res. 2017, 23, 5218–5224.
  109. Kim, E.S. Abemaciclib: First Global Approval. Drugs 2017, 77, 2063–2070.
  110. Slamon, D.J.; Neven, P.; Chia, S.; Fasching, P.A.; De Laurentiis, M.; Im, S.-A.; Petrakova, K.; Bianchi, G.V.; Esteva, F.J.; Martín, M.; et al. Phase III randomized study of ribociclib and fulvestrant in hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer: MONALEESA-3. J. Clin. Oncol. 2018, 36, 2465–2472.
  111. Iyengar, M.; O’Hayer, P.; Cole, A.; Sebastian, T.; Yang, K.; Coffman, L.; Buckanovich, R.J. CDK4/6 inhibition as maintenance and combination therapy for high grade serous ovarian cancer. Oncotarget 2018, 9, 15658–15672.
  112. Slamon, D.J.; Fasching, P.A.; Patel, R.; Verma, S.; Hurvitz, S.A.; Chia, S.K.L.; Crown, J.; Martin, M.; Barrios, C.H.; Spera, G.; et al. NATALEE: Phase III study of ribociclib (RIBO) + endocrine therapy (ET) as adjuvant treatment in hormone receptor–positive (HR+), human epidermal growth factor receptor 2–negative (HER2–) early breast cancer (EBC). J. Clin. Oncol. 2019, 37, TPS597.
  113. Curigliano, G.; Pardo, P.G.; Meric-Bernstam, F.; Conte, P.; Lolkema, M.; Beck, J.; Bardia, A.; Garcia, M.M.; Penault-Llorca, F.; Dhuria, S.; et al. Ribociclib plus letrozole in early breast cancer: A presurgical, window-of-opportunity study. Breast 2016, 28, 191–198.
  114. Malorni, L.; Piazza, S.; Ciani, Y.; Guarducci, C.; Bonechi, M.; Biagioni, C.; Hart, C.D.; Verardo, R.; Di Leo, A.; Migliaccio, I. A gene expression signature of retinoblastoma loss-of-function is a predictive biomarker of resistance to palbociclib in breast cancer cell lines and is prognostic in patients with ER positive early breast cancer. Oncotarget 2016, 7, 68012–68022.
  115. Gong, X.; Litchfield, L.M.; Webster, Y.; Chio, L.-C.; Wong, S.S.; Stewart, T.R.; Dowless, M.; Dempsey, J.; Zeng, Y.; Torres, R.; et al. Genomic Aberrations that Activate D-type Cyclins Are Associated with Enhanced Sensitivity to the CDK4 and CDK6 Inhibitor Abemaciclib. Cancer Cell 2017, 32, 761–776.e6.
  116. Hurvitz, S.A.; Martin, M.; Press, M.F.; Chan, D.; Fernandez-Abad, M.; Petru, E.; Rostorfer, R.; Guarneri, V.; Huang, C.-S.; Barriga, S.; et al. Potent Cell-Cycle Inhibition and Upregulation of Immune Response with Abemaciclib and Anastrozole in neoMONARCH, Phase II Neoadjuvant Study in HR+/HER2− Breast Cancer. Clin. Cancer Res. 2020, 26, 566–580.
  117. Finn, R.S.; Crown, J.P.; Lang, I.; Boer, K.; Bondarenko, I.M.; Kulyk, S.O.; Ettl, J.; Patel, R.; Pinter, T.; Schmidt, M.; et al. The cyclin-dependent kinase 4/6 inhibitor palbociclib in combination with letrozole versus letrozole alone as first-line treatment of oestrogen receptor-positive, HER2-negative, advanced breast cancer (PALOMA-1/TRIO-18): A randomised phase 2 study. Lancet Oncol. 2015, 16, 25–35.
  118. Bartlett, C.H.; Mardekian, J.; Cotter, M.J.; Huang, X.; Zhang, Z.; Parrinello, C.M.; Bourla, A.B. Concordance of real-world versus conventional progression-free survival from a phase 3 trial of endocrine therapy as first-line treatment for metastatic breast cancer. PLoS ONE 2020, 15, e0227256.
  119. Sledge, G.W., Jr.; Toi, M.; Neven, P.; Sohn, J.; Inoue, K.; Pivot, X.; Burdaeva, O.; Okera, M.; Masuda, N.; Kaufman, P.A.; et al. MONARCH 2: Abemaciclib in Combination With Fulvestrant in Women With HR+/HER2− Advanced Breast Cancer Who Had Progressed While Receiving Endocrine Therapy. J. Clin. Oncol. 2017, 35, 2875–2884.
  120. Yu, Q.; Geng, Y.; Sicinski, P. Specific protection against breast cancers by cyclin D1 ablation. Nature 2001, 411, 1017–1021.
  121. Teo, Z.L.; Versaci, S.; Dushyanthen, S.; Caramia, F.; Savas, P.; Mintoff, C.P.; Zethoven, M.; Virassamy, B.; Luen, S.J.; McArthur, G.; et al. Combined CDK4/6 and PI3Kα Inhibition Is Synergistic and Immunogenic in Triple-Negative Breast Cancer. Cancer Res. 2017, 77, 6340–6352.
  122. Asghar, U.; Barr, A.; Cutts, R.; Beaney, M.; Babina, I.; Sampath, D.; Giltnane, J.; Lacap, J.A.; Crocker, L.; Young, A.; et al. Single-Cell Dynamics Determines Response to CDK4/6 Inhibition in Triple-Negative Breast Cancer. Clin. Cancer Res. 2017, 23, 5561–5572.
  123. Martin, M.; Zielinski, C.; Ruiz-Borrego, M.; Carrasco, E.; Turner, N.; Ciruelos, E.M.; Muñoz, M.; Bermejo, B.; Margeli, M.; Anton, A.; et al. Palbociclib in combination with endocrine therapy versus capecitabine in hormonal receptor-positive, human epidermal growth factor 2-negative, aromatase inhibitor-resistant metastatic breast cancer: A phase III randomised controlled trial—PEARL. Ann. Oncol. 2020, 32, 488–499.
More
Video Production Service