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 + 2109 word(s) 2109 2021-08-11 06:23:42 |
2 The format is correct Meta information modification 2109 2021-08-12 05:04:29 |

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.
Barbarino, M. CDK4/CDK6/cyclin-D1 and Radiotherapy for Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/13045 (accessed on 25 April 2024).
Barbarino M. CDK4/CDK6/cyclin-D1 and Radiotherapy for Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/13045. Accessed April 25, 2024.
Barbarino, Marcella. "CDK4/CDK6/cyclin-D1 and Radiotherapy for Cancer" Encyclopedia, https://encyclopedia.pub/entry/13045 (accessed April 25, 2024).
Barbarino, M. (2021, August 11). CDK4/CDK6/cyclin-D1 and Radiotherapy for Cancer. In Encyclopedia. https://encyclopedia.pub/entry/13045
Barbarino, Marcella. "CDK4/CDK6/cyclin-D1 and Radiotherapy for Cancer." Encyclopedia. Web. 11 August, 2021.
CDK4/CDK6/cyclin-D1 and Radiotherapy for Cancer
Edit

The expanding clinical application of CDK4- and CDK6-inhibiting drugs in the managements of breast cancer has raised a great interest in testing these drugs in other neoplasms. The potential of combining these drugs with other therapeutic approaches seems to be an interesting work-ground to explore. Even though a potential integration of CDK4 and CDK6 inhibitors with radiotherapy (RT) has been hypothesized, this kind of approach has not been sufficiently pursued, neither in preclinical nor in clinical studies. Similarly, the most recent discoveries focusing on autophagy, as a possible target pathway able to enhance the antitumor efficacy of CDK4 and CDK6 inhibitors is promising but needs more investigations. 

cyclin inhibitors radiotherapy autophagy

1. Introduction

The expanding clinical application of CDK4- and CDK6-inhibiting drugs in the management of hormone-sensitive breast cancer [1][2][3][4] has raised a great interest in also testing these G1/S cell-cycle blocking agents in other neoplasms [5][6][7], for which the potential clinical benefit is still largely unknown.
The innovative use of CDK4 and CDK6 inhibitors in combination with hormone therapy (HT) as a treatment for estrogen receptor positive/human epidermal growth factor receptor-2 negative (ESR+/HER2−) metastatic breast cancer (BC) patientsis safe and successful in term of response rate (RR), progression-free survival (PFS), and overall survival (OS). Nevertheless, 10% of patients are refractory to this treatment whereas half of the patients show progression within 24 months of therapy [8]. Therefore, to fully unleash their impact, combining these drugs with other therapeutic approaches seems a rational work-ground to explore.
Even though a potential integration of CDK4 and CDK6 inhibitors with radiotherapy (RT) has been hypothesized, this kind of approach has not been sufficiently pursued, neither in preclinical nor in clinical studies. Similarly, the most recent discoveries focusing on autophagy as a possible target pathway able to enhance the antitumor efficacy of CDK4 and CDK6 inhibitors are promising but need more investigation.

2. The State of the Art in the Clinical Domain

2.1. Clinical Trials of Cyclin-D1/CDK4 and CDK6 Inhibition in Breast Cancer

In a phase II clinical trial (PALOMA-1), the addition of palbociclib to letrozole significantly increased the median progression-free survival (PFS) from 10.2 months to 20.2 months [9] vs. letrozole alone for post-menopausal women with previously untreated ESR1+/HER2− advanced breast cancer. Palbociclib received accelerated approval by the US Food and Drug Administration (FDA) in February 2015 [10]. The placebo-randomized, double-blind, controlled phase III trial (PALOMA-3) compared treatment with palbociclib and fulvestrant to fulvestrant plus placebo in women with ESR1+/HER2− metastatic breast cancer that had relapsed or progressed on prior hormone therapy, including a substantial portion of patients (33%) with prior chemotherapy for metastatic disease. The interim analysis of this study demonstrated a significantly improved median PFS (9.5 months versus 4.6 months, respectively) [11][12]. Palbociclib is presently under further investigation in over 50 trials in related clinical settings.
Ribociclib is a selective strong inhibitor of CDK4 and CDK6 [13], blocks Rb phosphorylation, and causes cell cycle arrest of Rb-positive tumor cells, similarly to palbociclib [14]. The MONALEESA-2 trial demonstrated that ribociclib plus letrozole significantly improved progression-free survival, compared with placebo plus letrozole as a first-line therapy in postmenopausal patients with HR-positive, HER2-negative advanced breast cancer [9].
Ongoing trials are demonstrating significant progression-free survival improvements with ribociclib in different settings [15][16]. However, both palpociclib and ribociclib were characterized by a not-negligible systemic toxicity [17].
Abemaciclib was tested in a phase I trial, showing efficacy against non-small cell lung cancer, melanoma, and mantle cell lymphoma, in addition to breast cancer [18]. In fact, other than its high affinity for CCND1/CDK4, it also inhibits cyclin-B/CDK1 (related to G2 arrest) and cyclin E-A/CDK2 (related to palpociclib resistance). Differently from the cytostatic effect alpociclib and ribociclib, abemaciclib achieved a durable cytotoxic effect in Rb-negative cells [19]. Moreover, in a first line randomized phase III double-blind study (MONARCH 3) for ESR1+/HER2− advanced breast cancer, abemaciclib presented a better hematological tolerability compared to palpociclib and ribociclib, and achieved impressive results in terms of PFS and objective response rate (ORR) [20].
A synthesis of these clinical trials is reported in Table 1.
Table 1. Synthesis of the registrative clinical trials of cyclin inhibitors in metastatic breast cancer.
Scheme. INTERVENTION PATIENT CHARACTERISTICS MEDIAN PFS(Months) Clinical Gain/Approval
PALOMA-1 Palbociclib + letrozole
vs.
letrozole alone
Post-menopausal women with untreated ER+/HER2−advanced breast cancer 20.2
vs.
10.2
Significant gain in terms of median PFS (10 Months).
FDA approval in 2015
PALOMA-2 Palbociclib + letrozole
vs.
letrozole alone
ESR1+/HER2−advanced breast cancer 27.6
vs.
14.5
Delayed ChT: 40.4
vs.
29.9 months
PALOMA-3 Palbociclib + fulvestrant
vs.
placebo + fulvestrant
ESR1+/HER2− metastatic breast cancer after hormone therapy (30% received prior ChT) 9.5 (11.2)
vs.
4.6
Significant gain in terms of median PFS
MONALEESA-2 Ribociclib + letrozole
vs.
letrozole alone
Postmenopausal women with ESR1+/HER2 advanced breast cancer Not reached
vs.
14.7
FDA approval in 2017
MONARCH-3 Abemaciclib + aromatase inhib.
vs.
placebo + aromatase inhib.
Postmenopausal women with ESR1+/HER2 locoregionally recurrent or metastatic breast cancer with no prior systemic therapy 28.1
vs.
14.7
Significantly prolonged PFSFDA approval in 2018

A metanalysis demonstrated that three CDK4 and CDK6 inhibitors have similar efficacy when associated with AIs, and are superior to either fulvestrant or AI monotherapy independently of any stratification criteria, thus supporting AI plus CDK4 and CDK6 inhibition as the best approach in ESR1+/HER2− metastatic breast cancer [10][12]

All the chemical structures of the molecules are presented in Figure 1.
Figure 1. Chemical structures of cyclin inhibitors that are currently approved.

2.2. Real-Life Clinical Reports on Cyclin-D1/CDK4 and CDK6 Inhibitors, Including Association with Radiotherapy

Delivering unplanned RT (mainly with palliative/symptomatic aims) in advanced breast cancer patients on Cyclin-D1/CDK4 and CDK6 inhibitors in prospective trials has produced empirically based treatment not supported by an appropriate rationale and consolidated approach, e g., in terms of RT intent, dose, and scheduling. However, several observational studies provided clearly suggest the lack of additive adverse events when palbociclib or ribociclib are combined with letrozole and concomitant palliative RT [21][22][23]. Additionally, a retrospective single-institution analysis suggested that 15 out a cohort of 42 pts with ESR1+ breast cancer with brain metastases showed promising results using CDK4 and CDK6 inhibitors palbociclib or abemaciclib for 6 months combined with stereotactic radiation [20] and only 5% of the patients developed radiation necrosis. However, only a slight improvement could be shown in survival. However, early radiation toxicities, including esophagitis and severe dermatitis have been reported in a breast cancer patient on palpociclib and palliative RT on cervical lymph nodes [24].

3. Molecular Grounds for Future Developments

3.1. CCND1 as a Target of Ionizing Radiation in the CCND1-CDK4, and CDK6 Complex

Only the CCND1-CDK4 and CDK6 complex has raised interest as a possible actionable target, whereas, in contrast, despite its prognostic relevance in cancer [25], the regulatory member CCND1 per se has received less attention. As previously quoted, palpociclib induces autophagy in cancer, targeting CDK4 and CDK6 [26][27], via the LKB1 and Ser325 phosphorylation resulting in a CCND1-mediated, reduced activation of AMPK [28]. The CCND1 gene encoding CCND1 is activated in B-cell lymphomas and overexpressed in many other human cancers (esophageal, lung, breast, bladder carcinomas, etc.) where it acts as a proto-oncogene. Therefore, the CCND1 is a potential pivotal actionable target for cell cycle arrest. In normal cells, CCND1 has a very short life due to UPS degradation whereas in cancer cells, inhibition of turnover and increased levels of CCND1 were shown. Conversely, the knockdown of the activity of a specific deubiquitinase for CCND1 was shown, inducing growth suppression in cancer cells addicted to its expression [29]. Ubiquitination of CCND1 is achievable in response to IR [30] and, in cancer cells, IR or other DNA-damaging agents may activate selective autophagy [26]. Autophagy shares common molecular mechanisms with UPS, with which it is intertwined in a close and complex relationship [31]. As described by these authors, ubiquitin-specific recognition can have a pivotal role for CCND1 autophagy degradation. Selective autophagy requires some receptor proteins necessary for the process, such as p62 (also known as sequestosome 1 or SQSTM1), NBR1 (neighbor of the BRCA 1 gene), CALCOCO2 (calcium binding and coiled-coil domain 2), and optineurin. p62 (the most relevant receptor protein for the subject dealt with here) is a component of several transduction signal cascades, such as Keap-Nrf2, and essential for UPS degradation.
Possible future strategies aimed at enhancing the IR-autophagic degradation of CCND1 in combination with drugs targeting the CCND1-CDK4 and CDK6 complex for G1/S arrest could overcome the IR-resistance of G1 and S phases of cell cycle

2.2. Ionizing Radiation, Autophagy Enhancement, and CCND1

Ionizing radiation can directly induce autophagy; although the mechanism underlying this effect has not been fully elucidated so far, two main mechanisms have been evoked: (1) similarly to rapamycin, IR reverts auto-phosphorylation of mTOR (e.g., in MCF-7 breast cancer cell culture) leading to the formation of autophagic vacuoles, segregation of cytoplasmic materials, and appearance of LC3 positivity and eventually mitochondrial hyperpolarization and decreased cell survival [32].
(2) Radiation causes a protective pathway via endoplasmic reticulum (ER) stress, accumulation of ER misfolded proteins (unfolded protein response—UPR), and autophagy. Therefore, UPR pathway and autophagy are upregulated in some cancer types as an escape strategy leading to IR-activated autophagy responsible for resistance to RT [33][34][35]. Nonetheless, autophagy is always depictable as a “double-edged sword”. As previously reviewed [31], most of the literature dedicated to this subject is focused on drug manipulation of autophagy (i.e., either induction or inhibition) for improving IR-sensitivity in cancer cells, more than to IR in conditioning, per se, autophagy towards cell death.
Emphasis should be placed in autophagy during the cell cycle, as different studies have investigated the autophagy activation in the different phases of the cell cycle [32][33]. In contrast, other evidence suggests a protective suppression of autophagy during mitosis aimed at preventing the inappropriate inactivation of molecular species and organelles, functional for cellular replication, or even aggregated chromosomes [36]. IR-based autophagy induced during the G1 and S counteracts the accelerated cell cycle in cancer and lays the groundwork for combined therapies combined with IR. It has been shown that CCND1 suppresses autophagy in mammary epithelium [37] whereas CCND1 inhibits autophagy by activating AMPK in breast cancer cells [28]. Further, in 2016, our group published a study in recurrent GB patients that prompted us to hypothesize a role for the IR-temozolomide treatment in inducing CCND1 autophagic degradation in Beclin-1-positive GBs [38]. Consistently, another group demonstrated the inverse correlation between low autophagic activity and high CCND1 expression that were also related to poorer prognosis [4]. In human and murine models of hepatocellular carcinomas (HCC), induction of autophagy led to CCND1 ubiquitination and its recruitment into autophagosomes via SQSTM1, formation of autophago-lysosomes, and degradation with suppression of tumor growth. Further, Chen et al. in 2019 [39] showed that autophagy itself causes CCND1 autophagic degradation in breast cancer cell lines with subsequent G1 cell cycle arrest underpinning the hypothesis of cell cycle modulation by CCND1 via mTORC1 signaling inhibition. Finally, we proposed in 2020 an overview of the role of autophagy, and namely its pivotal Beclin-1 protein, in CCND1 degradation, as a possibly actionable mechanism for G1/S cell cycle arrest in cancer (Figure 2) [40].
Figure 2. A simplified overview of the role of autophagy and Beclin-1 in CCND1 degradation, as a possible mechanism actionable for G 1/S cell cycle arrest in cancer. Radiation and chemotherapy are also considered in this domain. Some steps of the pathway are indicated, as shown in different tumors. A: Ubiquitation in CCND1 levels is achievable by gamma irradiation [3]. B: Ubiquitin–proteasome system and autophagy functionally cooperate with one another for protein homeostasis [1]. C: Autophagy is induced in response to environmental stress resulting in abnormal or degraded proteins [5]. Beclin-1 (encoded by the BECN1 gene) is an autophagy protein, which has a critical role in the autophagy machinery. LC3 is a protein of the late autophagy pathway, necessary for autophagosome, biogenesis, and is used as an autophagy marker. D: Mammalian target of rapamycin (mTOR) inhibitors (everolimus [2], rapamycin [6][41], amiodarone [41]) activate autophagy, also concurrently with radiation or temozolomide chemotherapy [6]. E: The autophagosome engulfs degraded proteins [5], subsequently digested via its fusion with lysosomes. CCND1 colocalizes with Beclin-1 in the cytoplasm [7] after cancer therapy and this suggests its autophagy degradation. F and G: CCND1 degradation deactivates the formation of the CCND1/CDK4 and CDK6 complex, thus activating the Rb1-dependent onco-suppressive pathway [42] and hampering cell cycle progression [2][6]. In gray: an alternative pathway of CCND1 degradation is through the ubiquitin–proteasome mechanism (UPD). Active mTOR complex 1 (mTORC1) inhibits both UPD and autophagy (dashed lines). PI3K, phosphatidylinositol 3-kinase. Image copyright © 2021, The American Physiological Society [40].
It is still unclear whether autophagy could represent a true per se pro-death process, or can simply enhance the IR and chemotherapy sensitivity of cancer cells. The impact of autophagy in a cell death progress is cautiously named “autophagy-related cell death”. This definition is underpinned by the clinical observation that a high tissue expression of cytoplasmic Beclin-1 was related to better prognosis in patients with malignant glioma on temozolomide chemotherapy and radiotherapy [43]. Notwithstanding, one should also take account of the tumor suppression of the autophagy-independent functions of Beclin-1 [44].

References

  1. Pajonk, F.; Vlashi, E.; McBride, W.H. Radiation resistance of cancer stem cells: The 4 R’s of radiobiology revisited. Stem Cells 2010, 28, 639–648.
  2. Whittaker, S.; Madani, D.; Joshi, S.; Chung, S.A.; Johns, T.; Day, B.; Khasraw, M.; McDonald, K.L. Combination of palbociclib and radiotherapy for glioblastoma. Cell Death Discov. 2017, 3, 17033.
  3. Nardone, V.; Falivene, S.; Giugliano, F.M.; Gaetano, M.; Giordano, P.; Muto, M.; Daniele, B.; Guida, C. The role of radiation therapy and systemic therapies in elderly with breast cancer. Transl. Cancer Res. 2020, 9, S97–S109.
  4. Renzulli, M.; Zanotti, S.; Clemente, A.; Mineo, G.; Tovoli, F.; Reginelli, A.; Barile, A.; Cappabianca, S.; Taffurelli, M.; Golfieri, R. Hereditary breast cancer: Screening and risk reducing surgery. Gland. Surg. 2019, 8, S142–S149.
  5. Sicinska, E.; Aifantis, I.; Le Cam, L.; Swat, W.; Borowski, C.; Yu, Q.; Ferrando, A.A.; Levin, S.D.; Geng, Y.; Von Boehmer, H.; et al. Requirement for cyclin D3 in lymphocyte development and T cell leukemias. Cancer Cell 2003, 4, 451–461.
  6. Wu, S.Y.; Lan, S.H.; Wu, S.R.; Chiu, Y.C.; Lin, X.Z.; Su, I.J.; Tsai, T.F.; Yen, C.J.; Lu, T.H.; Liang, F.W.; et al. Hepatocellular carcinoma-related cyclin D1 is selectively regulated by autophagy degradation system. Haepatology 2018, 68, 141–154.
  7. Hamilton, E.; Infante, J.R. Targeting CDK4/6 in patients with cancer. Cancer Treat. Rev. 2016, 45, 129–138.
  8. McCartney, A.; Migliaccio, I.; Bonechi, M.; Biagioni, C.; Romagnoli, D.; De Luca, F.; Galardi, F.; Risi, E.; De Santo, I.; Benelli, M.; et al. Mechanisms of Resistance to CDK4/6 Inhibitors: Potential Implications and Biomarkers for Clinical Practice. Front. Oncol. 2019, 9, 666.
  9. Hortobagyi, G.; Stemmer, S.; Burris, H.; Yap, Y.-S.; Sonke, G.; Paluch-Shimon, S.; Campone, M.; Petrakova, K.; Blackwell, K.; Winer, E.; et al. Updated results from MONALEESA-2, a phase III trial of first-line ribociclib plus letrozole versus placebo plus letrozole in hormone receptor-positive, HER2-negative advanced breast cancer. Ann. Oncol. 2019, 30, 1842.
  10. Beaver, J.A.; Amiri-Kordestani, L.; Charlab, R.; Chen, W.; Palmby, T.; Tilley, A.; Zirkelbach, J.F.; Yu, J.; Liu, Q.; Zhao, L.; et al. FDA Approval: Palbociclib for the Treatment of Postmenopausal Patients with Estrogen Receptor-Positive, HER2-Negative Metastatic Breast Cancer. Clin. Cancer Res. 2015, 21, 4760–4766.
  11. Cristofanilli, M.; Turner, N.C.; Bondarenko, I.; Ro, J.; Im, S.-A.; Masuda, N.; Colleoni, M.; DeMichele, A.; Loi, S.; Verma, S.; et al. Fulvestrant plus palbociclib versus fulvestrant plus placebo for treatment of hormone-receptor-positive, HER2-negative metastatic breast cancer that progressed on previous endocrine therapy (PALOMA-3): Final analysis of the multicentre, double-blind, phase 3 randomised controlled trial. Lancet Oncol. 2016, 17, 425–439.
  12. Turner, N.C.; Ro, J.; André, F.; Loi, S.; Verma, S.; Iwata, H.; Harbeck, N.; Loibl, S.; Huang Bartlett, C.; Zhang, K. Palbociclib in Hormone-Receptor-Positive Advanced Breast Cancer. N. Engl. J. Med. 2015, 373, 209–219.
  13. 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.
  14. Rader, J.; Russell, M.R.; Hart, L.S.; Nakazawa, M.S.; Belcastro, L.T.; Martinez, D.; Li, Y.; Carpenter, E.L.; Attiyeh, E.F.; Diskin, S.J.; et al. Dual CDK4/CDK6 inhibition induces cell-cycle arrest and senescence in neuroblastoma. Clin. Cancer Res. 2013, 19, 6173–6182.
  15. 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. Overall Survival with Ribociclib plus Fulvestrant in Advanced Breast Cancer. N. Engl. J. Med. 2020, 382, 514–524.
  16. Tripathy, D.; Im, S.A.; Colleoni, M.; Franke, F.; Bardia, A.; Harbeck, N.; Hurvitz, S.A.; Chow, L.; Sohn, J.; Lee, K.S.; et al. Ribociclib plus endocrine therapy for premenopausal women with hormone-receptor-positive, advanced breast cancer (MONALEESA-7): A randomised phase 3 trial. Lancet Oncol. 2018, 19, 904–915.
  17. Yang, Y.; Luo, J.; Chen, X.; Yang, Z.; Mei, X.; Ma, J.; Zhang, Z.; Guo, X.; Yu, X. CDK4/6 inhibitors: A novel strategy for tumor radiosensitization. J. Exp. Clin. Cancer Res. 2020, 39, 188.
  18. 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.
  19. Rossi, V.; Berchialla, P. Should All Patients with HR-Positive HER2-Negative Metastatic Breast Cancer Receive CDK 4/6 Inhibitor as First-Line Based Therapy? A Network Meta-Analysis of Data from the PALOMA 2, MONALEESA 2, MONALEESA 7, MONARCH 3, FALCON, SWOG and FACT trials. Cancers 2019, 11, 1661.
  20. Goetz, M.P.; Toi, M.; Campone, M.; Sohn, J.; Paluch-Shimon, S.; Huober, J.; Park, I.H.; Trédan, O.; Chen, S.C.; Manso, L.; et al. MONARCH 3: Abemaciclib As Initial Therapy for Advanced Breast Cancer. J. Clin. Oncol. 2017, 35, 3638–3646.
  21. Meattini, I.; Desideri, I.; Scotti, V.; Simontacchi, G.; Livi, L. Ribociclib plus letrozole and concomitant palliative radiotherapy for metastatic breast cancer. Breast 2018, 42, 1–2.
  22. Ratosa, I.; Orazem, M.; Scoccimarro, E.; Steinacher, M.; Dominici, L.; Aquilano, M.; Cerbai, C.; Desideri, I.; Ribnikar, D.; Marinko, T.; et al. Cyclin-Dependent Kinase 4/6 Inhibitors Combined with Radiotherapy for Patients with Metastatic Breast Cancer. Clin. Breast Cancer 2020, 20, 495–502.
  23. Ippolito, E.; Greco, C.; Silipigni, S.; Dell’Aquila, E.; Petrianni, G.M.; Tonini, G.; Fiore, M.; D’Angelillo, R.M.; Ramella, S. Concurrent radiotherapy with palbociclib or ribociclib for metastatic breast cancer patients: Preliminary assessment of toxicity. Breast 2019, 46, 70–74.
  24. Li, J.; Huo, X.; Zhao, F.; Ren, D.; Ahmad, R.; Yuan, X.; Du, F.; Zhao, J. Association of Cyclin-Dependent Kinases 4 and 6 Inhibitors with Survival in Patients with Hormone Receptor-Positive Metastatic Breast Cancer: A Systematic Review and Meta-analysis. JAMA Netw. Open 2020, 3, e2020312.
  25. VanArsdale, T.; Boshoff, C.; Arndt, K.T.; Abraham, R.T. Molecular Pathways: Targeting the Cyclin D-CDK4/6 Axis for Cancer Treatment. Clin. Cancer Res. 2015, 21, 2905–2910.
  26. Zheng, K.; He, Z.; Kitazato, K.; Wang, Y. Selective Autophagy Regulates Cell Cycle in Cancer Therapy. Theranostics 2019, 9, 104–125.
  27. Knudsen, E.S.; Witkiewicz, A.K. The Strange Case of CDK4/6 Inhibitors: Mechanisms, Resistance, and Combination Strategies. Trends Cancer 2017, 3, 39–55.
  28. Casimiro, M.C.; Di Sante, G.; Di Rocco, A.; Loro, E.; Pupo, C.; Pestell, T.G.; Bisetto, S.; Velasco-Velázquez, M.A.; Jiao, X.; Li, Z.; et al. Cyclin D1 Restrains Oncogene-Induced Autophagy by Regulating the AMPK-LKB1 Signaling Axis. Cancer Res. 2017, 77, 3391–3405.
  29. Shan, J.; Zhao, W.; Gu, W. Suppression of cancer cell growth by promoting cyclin D1 degradation. Mol. Cell 2009, 36, 469–476.
  30. Choo, D.W.; Baek, H.J.; Motoyama, N.; Cho, K.H.; Kim, H.S.; Kim, S.S. ATM is required for rapid degradation of cyclin D1 in response to gamma-irradiation. Biochem. Biophys. Res. Commun. 2009, 378, 847–850.
  31. Liu, W.J.; Ye, L.; Huang, W.F.; Guo, L.J.; Xu, Z.G.; Wu, H.L.; Yang, C.; Liu, H.F. p62 links the autophagy pathway and the ubiqutin-proteasome system upon ubiquitinated protein degradation. Cell Mol. Biol. Lett. 2016, 21, 29.
  32. Paglin, S.; Lee, N.Y.; Nakar, C.; Fitzgerald, M.; Plotkin, J.; Deuel, B.; Hackett, N.; McMahill, M.; Sphicas, E.; Lampen, N.; et al. Rapamycin-sensitive pathway regulates mitochondrial membrane potential, autophagy, and survival in irradiated MCF-7 cells. Cancer Res. 2005, 65, 11061–11070.
  33. Kim, K.W.; Moretti, L.; Mitchell, L.R.; Jung, D.K.; Lu, B. Endoplasmic reticulum stress mediates radiation-induced autophagy by perk-eIF2alpha in caspase-3/7-deficient cells. Oncogene 2010, 29, 3241–3251.
  34. De Cataldo, C.; Bruno, F.; Palumbo, P.; Di Sibio, A.; Arrigoni, F.; Clemente, A.; Bafile, A.; Gravina, G.L.; Cappabianca, S.; Barile, A.; et al. Apparent diffusion coefficient magnetic resonance imaging (ADC-MRI) in the axillary breast cancer lymph node metastasis detection: A narrative review. Gland Surg. 2020, 9, 2225–2234.
  35. Reginelli, A.; Silvestro, G.; Fontanella, G.; Sangiovanni, A.; Conte, M.; Nuzzo, I.; Calvanese, M.; Traettino, M.; Ferraioli, P.; Grassi, R.; et al. Validation of DWI in assessment of radiotreated bone metastases in elderly patients. Int. J. Surg. 2016, 33, S148–S153.
  36. Mathiassen, S.G.; De Zio, D.; Cecconi, F. Autophagy and the Cell Cycle: A Complex Landscape. Front. Oncol. 2017, 7, 51.
  37. Brown, N.E.; Jeselsohn, R.; Bihani, T.; Hu, M.G.; Foltopoulou, P.; Kuperwasser, C.; Hinds, P.W. Cyclin D1 activity regulates autophagy and senescence in the mammary epithelium. Cancer Res. 2012, 72, 6477–6489.
  38. Pirtoli, L.; Belmonte, G.; Toscano, M.; Tini, P.; Miracco, C. Cyclin D1 Co-localizes with Beclin-1 in Glioblastoma Recurrences: A Clue to a Therapy-induced, Autophagy-mediated Degradative Mechanism? Anticancer Res. 2016, 36, 4057–4062.
  39. Chen, G.; Ding, X.F.; Bouamar, H.; Pressley, K.; Sun, L.Z. Everolimus induces G(1) cell cycle arrest through autophagy-mediated protein degradation of cyclin D1 in breast cancer cells. Am. J. Physiol. Cell. Physiol. 2019, 317, C244–C252.
  40. Pirtoli, L.; Belmonte, G.; Toscano, M.; Tini, P.; Miracco, C. Comment on “Everolimus induces G1 cell cycle arrest through autophagy-mediated protein degradation of cyclin D1 in breast cancer cells”. Am. J. Physiol. Physiol. 2020, 318, C448–C449.
  41. Puyol, M.; Martín, A.; Dubus, P.; Mulero, F.; Pizcueta, P.; Khan, G.; Guerra, C.; Santamaría, D.; Barbacid, M. A synthetic lethal interaction between K-Ras oncogenes and Cdk4 unveils a therapeutic strategy for non-small cell lung carcinoma. Cancer Cell. 2010, 18, 63–73.
  42. Prokhorova, E.A.; Egorshina, A.Y.; Zhivotovsky, B.; Kopeina, G.S. The DNA-damage response and nuclear events as regulators of nonapoptotic forms of cell death. Oncogene 2019, 39, 1–16.
  43. Pirtoli, L.; Cevenini, G.; Tini, P.; Vannini, M.; Oliveri, G.; Marsili, S.; Mourmouras, V.; Rubino, G.; Miracco, C. The prognostic role of Beclin 1 protein expression in high-grade gliomas. Autophagy 2009, 5, 930–936.
  44. Matthew-Onabanjo, A.N.; Janusis, J.; Mercado-Matos, J.; Carlisle, A.; Kim, D.; Levine, F.; Cruz-Gordillo, P.; Richards, R.; Lee, M.J.; Shaw, L.M. Beclin 1 Promotes Endosome Recruitment of Hepatocyte Growth Factor Tyrosine Kinase Substrate to Suppress Tumor Proliferation. Cancer Res. 2019, 80, 249–262.
More
Information
Subjects: Oncology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 284
Revisions: 2 times (View History)
Update Date: 12 Aug 2021
1000/1000