Skip to main content
Erschienen in: Journal of Experimental & Clinical Cancer Research 1/2020

Open Access 01.12.2020 | Review

CDK4/6 inhibitors: a novel strategy for tumor radiosensitization

verfasst von: Yilan Yang, Jurui Luo, Xingxing Chen, Zhaozhi Yang, Xin Mei, Jinli Ma, Zhen Zhang, Xiaomao Guo, Xiaoli Yu

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2020

Abstract

Recently, the focus of enhancing tumor radiosensitivity has shifted from chemotherapeutics to targeted therapies. Cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors are a novel class of selective cell cycle therapeutics that target the cyclin D-CDK4/6 complex and induce G1 phase arrest. These agents have demonstrated favorable effects when used as monotherapy or combined with endocrine therapy and targeted inhibitors, stimulating further explorations of other combination strategies. Multiple preclinical studies have indicated that CDK4/6 inhibitors exhibit a synergistic effect with radiotherapy both in vitro and in vivo. The principal mechanisms of radiosensitization effects include inhibition of DNA damage repair, enhancement of apoptosis, and blockade of cell cycle progression, which provide the rationale for clinical use. CDK4/6 inhibitors also induce cellular senescence and promote anti-tumor immunity, which might represent potential mechanisms for radiosensitization. Several small sample clinical studies have preliminarily indicated that the combination of CDK4/6 inhibitors and radiotherapy exhibited well-tolerated toxicity and promising efficacy. However, most clinical trials in combined therapy remain in the recruitment stage. Further work is required to seek optimal radiotherapy-drug combinations. In this review, we describe the effects and underlying mechanisms of CDK4/6 inhibitors as a radiosensitizer and discuss previous clinical studies to evaluate the prospects and challenges of this combination.
Hinweise
Yilan Yang and Jurui Luo contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ATM
Ataxia telangiectasia mutated
ATRT
Atypical teratoid rhabdoid tumor
BSG
Brainstem Glioma
CCA
Cholangiocarcinoma
CDK4/6
Cyclin-dependent kinases 4 and 6
cGAS
Cyclic GMP-AMP synthase
CXCL1
Chemokine (C-X-C motif) ligand 1
DC
Dendritic cell
DDR
DNA damage repair
DEF
Radiation dose enhancement factor
DIPG
Diffuse intrinsic pontine glioma
DMF
Radiation dose modifying factor
DSB
Double-strand break
EAC
Esophageal adenocarcinoma
HCC
Hepatocellular carcinoma
HGG
High grade glioma
HNSCC
Head and neck squamous cell carcinoma
HR
Homologous recombination
IL-2
Interleukin-2
IL-6
Interleukin-6
IL-8
Interleukin-8
IMRT
Intensity modulated radiation therapy
IR
Ionizing radiation
MBC
Metastatic breast cancer
mTORC1
Mammalian target of rapamycin complex 1
NFAT
Nuclear factor of activated T cells
NHEJ
Non-homologous end joining
NPC
Nasopharyngeal carcinoma
NSCLC
Non-small cell lung cancer
p-H3
Phosphorylated histone 3
RB
Retinoblastoma
ROS
Reactive oxygen species
SA-β-gal
Senescence-associated β-galactosidase
SASP
Senescence-associated secretory phenotype
SBRT
Stereotactic body radiation therapy
SER
Sensitizer enhancement ratio
SER10
Sensitizer enhancement ratio at 10% survival
SER50
Sensitizer enhancement ratio at 50% survival
SF2
Survival fraction at 2 Gy
SSB
Single-strand break
STING
Stimulator of interferon genes
TNBC
Triple-negative breast cancer
Treg
Regulatory T cells
TSC2
Tuberous sclerosis complex 2

Background

Radiotherapy is one of the most important local control methods of malignant tumors. Approximately 50% of cancer patients receive radiotherapy during the treatment process [1]. However, the radioresistance of tumor cells limits the ability to reach a curative dose of radiation, which reduces radiotherapy efficacy and is more likely to cause local failures [1, 2]. Substantial efforts have been made to improve the radiosensitivity via various types of radiation modulators. The early-stage radiosensitizers are mostly chemotherapeutic agents, such as cisplatin and 5-fluorouracil, which have been demonstrated to enhance radiosensitivity in head and neck squamous cell carcinoma (HNSCC), nasopharyngeal carcinoma (NPC), and gastrointestinal cancers [3]. These chemotherapeutics exhibit radiosensitization effects by increasing radiation damage and inhibiting DNA repair process [4, 5]. However, due to the non-specific effects of chemotherapeutics, chemoradiation therapy increases the radiation toxicity of normal tissues as well [1]. Given the rapid development of targeted therapies, numerous studies of specific radiosensitizers are underway. CDK4/6 inhibitors are a novel class of selective cell cycle therapeutics that target the cyclin D-CDK4/6 complex and suppress activation of the downstream RB-E2F pathway, thereby blocking cell cycle progression and inhibiting the tumor cell proliferation [68]. Multiple preclinical studies have demonstrated the radiosensitization effects of CDK4/6 inhibitors in various cancer types. In this review, we focus on the molecular basis and the underlying mechanisms regarding the efficacy of CDK4/6 inhibitors with radiotherapy to provide a strong rationale for clinical utilization of the combined therapy.

Overview of CDK4/6 inhibitors

Role of CDK4/6 in G1-S transition

CDK4/6 plays a critical role in the G1-S checkpoint, which governs genome replication in the cell cycle [9, 10] (Fig. 1). From the classical view, the retinoblastoma (RB) protein, as a negative cell cycle regulator, binds to the transcription factor E2F to repress the transcriptional activity in the early G1 phase [9, 11]. Mitogenic stimuli induce increased expression levels of D-type cyclins, which form complexes with CDK4/6 to phosphorylate RB [9]. Hypophosphorylated RB partially relieves the inhibitory control of the E2F transcription factor family; promotes the expression of E2F target genes, such as cyclin E; and facilitates the G1 phase progression [9, 11, 12]. In the late G1 phase, CDK2 is activated as cyclin E levels increase. The continuous formation of the cyclin E-CDK2 complex hyperphosphorylates and inactivates RB proteins, which results in the release of E2F transcription factors, initiation of transcription, and increased expression of S phase genes [9, 12]. Thereby, the cell cycle proceeds from the late G1 to S phase normally. During the G1-S transition, CDK4/6 activity is regulated by Cip/Kip (p21cip1, p27kip1, and p57kip2) family and Ink4 family (p16Ink4a, p15Ink4b, p18Ink4c, and p19Ink4d) proteins. The Cip/Kip family broadly inhibits the cyclin-CDK complex activity, also known as pan-CDK inhibitors [10, 12]. Conversely, Ink4 family proteins specifically bind to CDK4/6 to inactivate the kinase, thus inducing the inhibition of RB phosphorylation and blockade of cell cycle progression [8, 9, 13]. CDK4/6 inhibitors exert similar tumor-suppressing functions as Ink4 family members [12]. All three CDK4/6 inhibitors bind to the ATP domain of CDK4/6, but their targets are slightly different [14] (Fig. 2).

Therapeutic applications of CDK4/6 inhibitors

The cyclin D-CDK4/6-RB pathway is frequently dysregulated in many cancers [12]. Moreover, increased levels of D-type cyclins and CDK4 are commonly observed [1517], which makes inhibitors targeting the cyclin D-CDK4/6-RB pathway ideal candidates for cancer therapeutics. When used as monotherapy, CDK4/6 inhibitors exhibit certain efficacy in solid tumors, such as non-small cell lung cancer (NSCLC) [18, 19] and mature teratoma [20]. Moreover, CDK4/6 inhibitors in combined therapies displayed better prospects. Palbociclib, ribociclib, and abemaciclib have been approved by the Food and Drug Administration for advanced hormone receptor-positive (HR+) breast cancer patients when combined with letrozole or fulvestrant [12, 13]. The combinatorial strategy presented a substantial improvement in progression-free survival with a well-tolerated toxicity profile in multiple clinical trials [2126]. The combination of CDK4/6 inhibitors and other targeted drugs also shows promising prospects. PI3K-AKT-mTOR and RAS-RAF-MEK-ERK pathway inhibitors both exhibit synergistic tumor suppression effects with CDK4/6 inhibitors in many preclinical models [2732]. An important mechanism is that activation of oncogenic PI3K and RAS pathways is correlated with the cyclin D-CDK4/6 complex [8, 33]. RAS pathway drives CCND1 mRNA transcription and enhances cyclin D-CDK4/6 activity [34]. Hence, PI3K and RAS pathway inhibitors in combination with CDK4/6 inhibitors produce a double-hit on cyclin D-CDK4/6 activity [9]. Moreover, CDK4/6, PI3K, and RAS pathways intersect via tuberous sclerosis complex 2 (TSC2)(Fig. 1), which is a negative regulator of mammalian target of rapamycin complex 1 (mTORC1) [9, 33]. The PI3K and RAS pathway activate mTOR signaling through phosphorylating TSC2, and CDK4/6 can directly bind to and phosphorylate TSC2 [33, 35]. Therefore, co-inhibition of the PI3K or RAS pathway with the CDK4/6 pathway leads to a synergistic suppression of TSC2 phosphorylation and thus decreases mTORC1 activity [36]. This action might represent another critical mechanism for the synergy of CDK4/6 inhibitors with PI3K or RAS pathway inhibitors.
Although CDK4/6 inhibitors have achieved some clinical success, the lack of predictive biomarkers is a major obstacle preventing CDK4/6 inhibitors from better efficacy. Currently, a majority of research focuses on the potential biomarkers of breast cancer, and the HR-positive status remains the only clinically used biomarker. However, HR expression levels as a biomarker showed no clear survival advantage in the phase III PALOMA-3 trial [13, 25]. Several potential biomarkers, such as CCND1, CDKN2A, and RB1, exhibit an association with the sensitivity of CDK4/6 inhibitors in preclinical studies [12, 13, 37]. The most reliable of these biomarkers is RB1 since amplification of CCND1 and loss of CDKN2A showed no difference in benefit in the phase II PALOMA-1/TRIO-18 study [38]. Loss of RB1 appears to induce resistance of CDK4/6 inhibitors in many in vitro studies, but this finding has yet to be confirmed clinically [12, 37]. Nevertheless, the lack of RB1 expression is rare in ER-positive breast cancer patients, leaving no appropriate biomarkers other than the HR status [39]. However, there are a few ongoing clinical trials specifically targeting the molecular drivers of tumors, such as Lung-MAP (NCT02154490) and SIGNATURE (NCT02187783) trials, in which patients are being allocated based on CCND, CDKN2A, or CDK4 aberrations. More data regarding the verification of various biomarkers will be available after the completion of these ongoing clinical trials.

Preclinical studies of CDK4/6 inhibitors as radiosensitizers

In addition to intersecting with ER, PI3K, and RAS pathways, the cyclin D-CDK4/6-RB pathway is also involved in DNA damage repair (DDR) [40], which makes CDK4/6 inhibitors perfect candidates for tumor radiosensitization. Numerous in vitro and in vivo data have validated the radiosensitization effects of CDK4/6 inhibitors.

In vitro experimental data

CDK4/6 inhibitors enhance the radiosensitivity in multiple cell lines. The combination therapy significantly inhibits single-cell colony formation with a sensitizer enhancement ratio (SER) of 1.1–2.3 (Table 1). The radiosensitization effect is quite outstanding in malignant brain cancer cell lines. Two medulloblastoma cell lines, Daoy and ONS-76, were administered different doses of radiation following 48 h of treatment with palbociclib. The enhancement ratio ranged from 1.6 to 2.3 at 10% survival [41]. Moreover, the combination not only decreases colony numbers but also reduces the tumor sphere formation ability [45]. The tumor sphere formation ability is associated with cancer stem cell properties, which includes increased radiation resistance [50, 51]. Huang et al. [45] reported that the combination of 6 Gy radiation and 5 μM palbociclib dramatically reduced tumor sphere formation in Huh7 (hepatocellular carcinoma, HCC) cells compared to ionizing radiation (IR) alone. The combined therapy significantly inhibited radiation-induced cell growth and reduced radiation resistance.
Table 1
In vitro experimental data of radiosensitization effects
Authors
CDK4/6 inhibitor
Human cell line
Efficacy with radiotherapy
Whiteway et al. [41]
Palbociclib
Medulloblastoma
(Daoy)
(ONS-76)
SF2 a
SER10 b 1.6, SER50 c 1.5
SER10 2.3, SER50 2.3
Hashizume et al. [42]
Palbociclib
Intracranial ATRT d
(BT12)
(BT16)
DEF e 1.16–1.60
DEF 1.18–1.70
Whittaker et al. [43]
Palbociclib
Glioblastoma
(GBM-L1, HW1, RN1, BAH1)
Colony numbers drop to zero
Naz et al. [44]
Abemaciclib
NSCLC
(A549, H460, H820, H1975)
DMF f 1.30–1.71
Tao et al. [30]
Palbociclib and Trametinib
NSCLC
(A549, H460)
Cell survival fraction ↓
Apoptosis ↑
Huang et al. [45]
Palbociclib
HCC (Hep3B, Huh7)
CCA g (HuCCT1)
Tumor sphere numbers ↓
Colony numbers ↓
Xie et al. [46]
Palbociclib
NPC
(CNE-1)
(CNE-2)
SF2
SER 1.118–1.475
SER 1.10–1.20
Gottgens et al. [47]
Palbociclib
HNSCC
(UT-SCC-24A)
SER 1.36–1.60
Tai et al. [48]
Ribociclib
HNSCC
(OML1, OML1-R)
Colony numbers ↓
Cell viability ↓
Fernández-Aroca et al. [49]
Palbociclib
Breast cancer (MCF-7)
Lung cancer (A549)
Colorectal cancer (HCT116)
Cell survival fraction ↓
Li et al. [50]
Ribociclib and CA3
EAC h
(Flo-1 XTR)
Colony numbers ↓
a SF2, survival fraction at 2 Gy
b SER10, sensitizer enhancement ratio at 10% survival
c SER50, sensitizer enhancement ratio at 50% survival
d ATRT, atypical teratoid rhabdoid tumor
e DEF, radiation dose enhancement factor
f DMF, radiation dose modifying factor
g CCA, cholangiocarcinoma
h EAC, esophageal adenocarcinoma
Additionally, although palbociclib, ribociclib, and abemaciclib resemble each other structurally, the three drugs exhibit different radiosensitization effects. Naz et al. [44] demonstrated that palbociclib and ribociclib failed to promote radiation sensitivity in H460 (NSCLC) cells when added either pre- or post-IR treatment. In contrast, abemaciclib exhibited enhancement of radiosensitivity in the majority of NSCLC cell lines when added post-IR, including H460. CDK4/6 inhibitors probably have different radiosensitization effects, and the underlying mechanisms should be assessed in further investigations.

In vivo experimental data

In vivo preclinical studies have further verified the radiosensitization effects of CDK4/6 inhibitors, including prolonging the median survival time, reducing tumor volume, and delaying tumor regeneration (Table 2). Several in vivo studies demonstrated that palbociclib in combination with IR increased the median survival time by 1.2- to 3.3-fold in mice with brain malignancy xenografts [42, 43, 52]. The combined therapy also dramatically reduced the tumor weight and volume without no obvious systemic toxicity as assessed by mouse body weight [30, 45, 50]. The Ki-67 staining in xenografts was drastically reduced, revealing the reduction of tumor proliferation [30, 44, 50].
Table 2
In vivo experimental data of radiosensitization effects
Authors
CDK4/6 inhibitor
Xenograft
Efficacy with radiotherapy
Hashizume et al. [42]
Palbociclib
Intracranial ATRT
(BT12)
(BT16)
Glioblastoma
(GBM43)
Median survival time increased by 24 to 26 days (1.4-fold)
Median survival time increased by 31.5 to 34.5 days (3.3-fold)
Median survival time increased by 10 to 13.5 days (1.6-fold)
Whittaker et al. [43]
Palbociclib
Glioblastoma
(RN1)
Median survival time increased by 8 days
Naz et al. [44]
Abemaciclib
NSCLC
(H460)
Tumor regrowth delay 8 and 9 days, inhibit IR-induced tumor vasculogenesis
Tao et al. [30]
Palbociclib and Trametinib
NSCLC
(A549)
Tumor volume ↓
Proliferation ↓
Apoptosis ↑
Huang et al. [45]
Palbociclib
HCC
(Huh7)
Tumor volume ↓
Tumor weight ↓
Li et al. [50]
Ribociclib and CA3
EAC
(Flo-1 XTR)
Tumor volume ↓
Tumor weight ↓
Barton et al. [52]
Palbociclib
Ink4a-ARF- deficient BSG a mouse model
Median survival time increased by 10 days (19%)
a BSG, brainstem glioma

Mechanisms of CDK4/6 inhibitors as radiosensitizers

Inhibition of DNA damage repair

The impairment of DDR is an essential determinant of CDK4/6 inhibitor-associated radiosensitization in human cell lines. IR produces cytotoxic effects on DNA, which mainly results in DNA single-strand breaks (SSBs) and double-strand breaks (DSBs) [53]. Unrepaired and inaccurate-repaired DNA DSBs are the main cause of radiation-induced cell death [54]. Multiple preclinical studies have demonstrated that IR increases γ-H2AX and 53BP1 levels in a dose dependent manner and increases DNA DSBs [30, 42, 4447]. γ-H2AX is one of the earliest events of DDR with the induction of DSB [55]. The abundance of γ-H2AX foci reaches a peak at 30 min after IR and returns to baseline levels approximately 24 h post-IR [37]. In addition, 53BP1 plays a similar role to γ-H2AX in most cases [45], which is also a crucial hallmark of IR-induced DSBs [56]. Naz et al. [44] investigated γ-H2AX foci in H460 and H1299 (NSCLC) cells following 7.5 Gy irradiation. The γ-H2AX level rapidly increased in H460 (7.9-fold) and H1299 (6.7-fold) cells 0.5 h after IR compared to untreated control. Additional studies also demonstrated that various post-irradiated cell lines showed substantial increases in γ-H2AX and (or) 53BP1 foci, which eventually led to increased DSBs [37, 42, 4547, 49].
γ-H2AX and 53BP1 levels decrease over time, reflecting the dynamic process of DDR. The combination of IR and CDK4/6 inhibitors causes marked retention of γ-H2AX and (or) 53BP1 levels, thus delaying DSB repair to enhance radiotherapy efficacy [42, 4447]. With 7.5 Gy and 10 μM abemaciclib, γ-H2AX levels were increased 3.92-fold at 24 h in H460 cells. However, the levels were only 0.97-fold in H1299 cells (radiation-resistant), which was basically equivalent to the level before irradiation [44]. Huang et al. [45] reported similar findings in HuCCT1 (CCA), Huh7, and Hep3B (HCC) cells. These post-IR cells treated with 5 μM palbociclib sustained higher levels of γ-H2AX and 53BP1 at 24 h compared with DMSO-treated cells. Several other studies also supported this view that CDK4/6 inhibitors exhibited cellular radiosensitivity by increasing unrepaired DSBs and inducing the delayed repair kinetics of DSBs [42, 46, 47].
The combination of CDK4/6 inhibitors and IR not only increases DSBs and delays the DSB repair but also causes homologous recombination (HR) deficiency by decreasing the expression of Rad51 and ataxia telangiectasia mutated (ATM) kinase [57]. HR and non-homologous end joining (NHEJ) are the two principal pathways in DSB repair, and the latter plays a dominant role in IR-induced DDR [55, 58]. The DNA recombinase Rad51 is a pivotal component of the HR pathway, and the fraction of Rad51 foci increases during the HR process [47]. ATM kinase, the upstream regulator of Rad51, is activated to enhance HR in response to DSBs [55, 57]. In UT-SCC-24A (HNSCC) cells, palbociclib combined with IR reduced the expression of Rad51 approximately 3.5-fold [47]. Palbociclib also impaired ATM kinase activation by reducing phosphorylation of ATM kinase and its downstream targets with 10 Gy irradiation in both HCC and CCA cells [45]. However, impairment of the HR pathway appeared to be dependent on a functional p53 or RB status. Abemaciclib and IR decreased the formation of Rad51 foci markedly in H460 (p53-proficient) cells. Nevertheless, no significant changes were observed in H1299 (p53-deficient) cells [44]. A possible explanation was that p53 is a key effector in IR-induced cellular response and the lack of p53 was associated with increased radioresistance [59]. This hypothesis was confirmed by Fernández-Aroca et al. [49], who reported that p53 was a critical determinant of palbociclib-associated radiosensitivity. Dean et al. [37] shared similar findings in breast cancer given that the response of Rad51 to palbociclib with IR presented in an RB-dependent manner. Palbociclib pretreatment led to complete inhibition of Rad51 foci accumulation in MDA-MB-231 and Hs578t (RB-proficient) cells but not in MDA-MB-468 (RB-deficient) cells. Furthermore, 500 nM palbociclib caused an approximately 60% decrease in HR-mediated DSB repair and 2.5-fold increase in relative NHEJ activity, indicating that CDK4/6 inhibition augmented NHEJ efficiency [37]. However, current data are not available to demonstrate that CDK4/6 inhibitors with IR enhance NHEJ activity while weakening the HR efficiency. More studies are currently underway to elucidate this relationship.

Enhancement of apoptosis

As mentioned above, radiotherapy induces massive DSBs. As the intracellular stimulus, IR-induced DNA damage mediates most radiosensitivity-associated pro-apoptotic effects [60]. Several preclinical studies investigated apoptotic changes in combined therapy. Huang et al. [45] showed that 8 Gy irradiation plus 20 μM palbociclib resulted in remarkably increased DNA fragmentations (a hallmark of apoptosis) in HCC and CCA cells compared with monotherapy. Another study also reached similar conclusions in NPC cells, demonstrating that palbociclib treatment after irradiation prominently elevated the proportion of apoptotic cells in comparison with IR alone (CNE-1, 19.6% versus 10.685%; CNE-2, 21.655% versus 12.635%). Mechanistically, they demonstrated that the combination augmented the mitochondrial reactive oxygen species (ROS) level, which is regarded as an apoptosis mediator in radiotherapy or chemotherapy, thus enhancing apoptosis [46]. Interestingly, Hagen et al. [61] reported that palbociclib induced anti-apoptosis effects in irradiated MCF10A (normal human mammary epithelial cell) and MDA-MB-231 (triple-negative breast cancer, TNBC) cells by reducing cleaved PARP levels. The reason why the investigators reached the opposite conclusion remains unclear, but the radiotherapy-drug combination strategy and the p53 status of different cell lines could be an explanation.

Blockade of cell cycle progression

Another critical determinant of radiosensitivity is cell cycle arrest since CDK4/6 inhibitors induce significant G1-S arrest [11]. Meanwhile, G2-M phase cells are the most sensitive to radiation [4]. Tai et al. [48] found that 4 Gy irradiation in combination with ribociclib caused evident G1-S arrest in HNSCC cells as the ratio of OML1 cells in the G1 phase increased from 48.6 to 69.4% after treatment and similar effects were observed in radioresistant OML1-R cells. Xie et al. [46] demonstrated that concurrent palbociclib with IR and radiotherapy followed by palbociclib in NPC cells conspicuously increased the G2-M cell proportion and decreased radioresistant G1 cells, suggesting that the combination therapy also caused G2-M arrest. Furthermore, the combined regimens not only blocked G1-S and G2-M checkpoint but also suppressed mitosis. Gottgens et al. [47] investigated the changes in phosphorylated histone 3 (p-H3) at Ser10 in UT-SCC-24A (HNSCC) cells in response to IR and palbociclib. Phospho-H3 (Ser10) is associated with chromosome condensation, which is a major event during mitosis [62]. After IR alone, p-H3 (Ser10) levels dropped rapidly and returned to normal at 24 h. In contrast, p-H3 (Ser10) depleted quickly without rebound after combing palbociclib and IR, indicating a decreased number of mitotic cells and inhibition of mitosis. Intriguingly, Hagen et al. [61] reported that knockdown of CDK4 did not affect cell cycle as no marked changes in cell cycle distribution were observed after IR in shCDK4 cells. A possible explanation is that silencing CDK4 and CDK4/6 inhibitors display different outcomes in terms of cell cycle progression. In general, CDK4/6 inhibitors in combination with IR provide a strong blockade of cell cycle progression, which is a critical mechanism of radiosensitivity.

Other potential mechanisms

In addition to the classic radiosensitization mechanisms mentioned above, CDK4/6 inhibitors also induce other biological phenotypes, providing mechanistic foundations for the combination of CDK4/6 inhibitors and radiotherapy.
First, long-term exposure to CDK4/6 inhibitors induces the cellular senescence phenotype in many cancer cell lines, such as breast cancer [63], neuroblastoma [64], and melanoma [65]. It is not surprising as CDK4/6 inhibitors play a similar role to p16Ink4a, and the p16Ink4a-RB pathway is one of the most important mechanisms of cellular senescence [66, 67]. Persistent CDK4/6 inhibition suppresses RB phosphorylation and downstream transcription activities to induce an irreversible arrest of cell proliferation, which is also known as cellular senescence [68, 69]. After 8 days exposure of palbociclib, the fraction of senescence-associated β-galactosidase (SA-β-gal)-positive cells was significantly increased in 1205Lu (melanoma) cells [65]. Ribociclib also yielded similar results in neuroblastoma cell lines [64]. On the other hand, radiotherapy triggers premature senescence in solid tumor cell lines [70]. This finding is attributed to the fact that IR causes massive lesions in DNA, therefore activates the ATM-Chk2-p53-p21 axis (senescence-associated DDR pathway), leading to persistent cell cycle arrest and cellular senescence [68]. In addition, 8 or 10 Gy irradiation accelerated cellular senescence in TP53 wild-type tumor cell lines based on increased SA-β-gal positivity [70, 71]. Accordingly, radiotherapy and CDK4/6 inhibitors may synergistically induce tumor cell senescence and further inhibit tumor progression.
Senescent cells secrete inflammatory cytokines, chemokines, and growth factors, which collectively comprise the the senescence-associated secretory phenotype (SASP) [67, 68]. After treatment with palbociclib for 8 days, SASP components, such as interleukin-6 (IL-6), interleukin-8 (IL-8), and chemokine (C-X-C motif) ligand 1 (CXCL1), secreted by 1205Lu cells substantially increased [65]. Furthermore, radiation triggers the release of cytoplasmic DNA and activates the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, which plays a pivotal role in SASP production [72, 73]. Thus, the combination of CDK4/6 inhibitors and radiation could theoretically induce SASP, which affects the immune response [72]. On one hand, SASP recruits immune cells to stimulate the adaptive immune response and eliminate senescent tumor cells. On the other hand, SASP also attracts immunosuppressive cells and creates a protumorigenic environment [72]. Although SASP is usually regarded as a “double-edged sword”, it still provides a novel perspective for radiosensitization mechanisms. Thus, the combination of CDK4/6 inhibitors and radiotherapy may exert radiosensitization effects through immunomodulation.
Indeed, several investigators have demonstrated that CDK4/6 inhibitors exhibited direct immunostimulatory effects in both tumor and immune cells [74]. In tumor cells, CDK4/6 inhibitors suppressed the RB-E2F-DNMT1 axis, which activated endogenous retroviral elements and increased double-stranded RNA levels. This action subsequently induced a type III interferon response and upregulated tumor antigen presentation [75]. On the other hand, radiotherapy enhances MHC class I expression by activating the mTOR pathway [76]. Moreover, radiation elicits activation of dendritic cells (DCs) and enhances cross-presentation of tumor antigens [73, 76]. Gupta et al. [77] reported that the expression of CD70 and CD86 (co-stimulatory molecules) on DCs was significantly increased after 10 Gy irradiation. In immune cells, CDK4/6 inhibitors promoted T cell activation via enhancing nuclear factor of activated T cells (NFAT) transcriptional activity and interleukin-2 (IL-2) production [78, 79]. On the other hand, radiation-induced T cell activation has been demonstrated in several preclinical studies, which is mediated by the induction of viral mimicry and activation of the cGAS-STING pathway [8082]. Although IR recruits regulatory T cells (Treg) and other immunosuppressive cells to the tumor microenvironment, CDK4/6 inhibitors markedly reduce the proliferation of Tregs [75]. Collectively, CDK4/6 inhibitors and radiotherapy may synergistically exert an anti-tumor immune response by enhancing antigen presentation capacity and T cell activation. These potential mechanisms offer new perspectives for future exploration. The combination of CDK4/6 inhibitors and radiation may not only improve local tumor control but also enhance systemic disease control, providing the possibility for the triplet combination of CDK4/6 inhibitors, radiotherapy and immunotherapy.

Clinical studies of CDK4/6 inhibitors in combination with radiotherapy

Multiple preclinical data suggest a potential synergistic effect when CDK4/6 inhibitors and radiotherapy are administered concurrently. However, safety and efficacy still require further investigations. Currently, there are six ongoing clinical trials (Table 3) as well as several small sample clinical studies (Table 4), which mostly target HR+/HER2- metastatic breast cancer (MBC) patients. The preliminary results [8386] propose that no significantly increased toxicity is observed, indicating that the combined therapy is promising as a novel strategy. Ippolito et al. [84] reported that hematological toxicity neutropenia is most common among all adverse events. In total, 60% of patients experienced grade 3 or greater neutropenia before combination treatment, suggesting that we should carefully evaluate previous toxicity to prevent reoccurrence. Figura et al. [85] retrospectively analyzed 42 lesions in 15 brain MBC patients. Two lesions (5%) developed radiation necrosis, and both of them underwent four previous RT courses before the occurrence of radionecrosis, indicating the significance of cautious assessment of the treatment plan margins. Chowdhary et al. [86] reported that concomitant treatment with palbociclib and RT resulted in grade 1 or 2 toxicity and notably relieved pain. Also, no local failure was noted in evaluable follow-up patients.
Table 3
Clinical trials with CDK4/6 inhibitors in combination with IR
Cancer Type
Phase
Arm
n
Status
NCT
HGG a, DIPGb, bithalamic HGG
I/II
Ribociclib + IR
24
Active, not recruiting
NCT02607124
Glioma (HGG, DIPG et al.)
I
Ribociclib + Everolimus + IR
24
Recruiting
NCT03355794
Locally advanced HNSCC
I/II
Palbociclib + Cetuximab + IMRT c
33
Recruiting
NCT03024489
HPV-unrelated HNSCC
II
Palbociclib + Cetuximab or Cisplatin + IMRT
29
Recruiting
NCT03389477
Bone metastatic breast cancer
(HR+/HER2-)
II
Palbociclib + Hormone therapy + IR
42
Recruiting
NCT03691493
Metastatic breast cancer (HR+/HER2-)
II
Palbociclib + Letrozole ± SBRT d
204
Not yet recruiting
NCT04220476
a HGG, high-grade glioma
b DIPG, diffuse intrinsic pontine glioma
c IMRT, intensity-modulated radiation therapy
d SBRT, stereotactic body radiation therapy
Table 4
Case reports and clinical studies with CDK4/6 inhibitors in combination with IR
Authors
Patients
Arm
Toxicity
Efficacy
Hans et al. [83]
Metastatic breast cancer
(HR+/HER2-)
n = 5
Palbociclib + Fulvestrant + palliative IR
Digestive toxicity: mucositis (grade 1 = 20%, grade 2 = 20%)
Hematological toxicity: grade 3 neutropenia = 40%, grade 3 anemia = 20%, grade 3 thrombopenia = 40%
No skin toxicity
Symptom control and pain relief (100%)
Ippolito et al. [84]
Metastatic breast cancer
(HR+)
n = 16
Palbociclib or Ribociclib + palliative IR
Hematological toxicity: neutropenia (grade 2 = 12.5%, grade 3 = 25%, grade 4 = 6.3%)
Absent
Figura et al. [85]
Brain metastatic breast cancer
(HR+)
n = 15
lesions = 42
Palbociclib or Abemaciclib + SBRT
Radionecrosis (2 lesions, 5%)
No neurologic toxicity or scalp toxicity
Median OS 36.7 months
Six-month local control (88%)
Six-month distant brain control (61%)
Chowd-hary et al. [86]
Metastatic breast cancer
(HR+/HER2-)
n = 16
Palbociclib + Fulvestrant or Letrozole + palliative IR
Hematological toxicity: grade 1 = 87.5%, grade 2 = 12.5%
No grade 2 or higher cutaneous, neurologic, or gastrointestinal toxicity
Local control and pain relief (100%)
Given these results, the combined treatment seems well-tolerated, whereas severe adverse effects may be a concern. A breast cancer patient with supraclavicular lymph node metastasis was reported to develop grade 3 radiation esophagitis and dermatitis after receiving palbociclib with 40 Gy in 20 fractions to the left neck [87]. Another case was a breast cancer patient with bone metastases at the left iliac bone and first sacral vertebrae. This patient experienced radiation-induced grade 3 enterocolitis after administration of palbociclib and 30 Gy in 10 fractions to pelvic bones, which might be related to the over-sensitization after palbociclib administration [88]. Hence, combination therapy should be used cautiously until more data are available. In addition, the patient’s condition should be thoroughly evaluated and treated individually before the utilization of combination regimens.

Conclusions

CDK4/6 inhibitors have greatly changed the treatment landscape of HR-positive breast cancer patients, stimulating further explorations of combination therapy. Significant preclinical data have demonstrated the radiosensitization effects of CDK4/6 inhibitors via inhibiting DDR, enhancing apoptosis, and blocking cell cycle progression. CDK4/6 inhibitors also induce cellular senescence and promote anti-tumor immunity, which might be potential mechanisms for clinical radiosensitization. Although several clinical studies have presented well-tolerated toxicity and promising efficacy for combination therapy, safety remains a major concern. Radiotherapy is currently used as palliative treatment in clinical trials, and most patients have already received irradiation in previous treatment processes. Therefore, re-irradiation is likely to cause cumulative toxicity and severe adverse events. In addition to palliative treatment, radiotherapy can also be used as a postoperative adjuvant treatment in combination with CDK4/6 inhibitors. Several studies demonstrated that CDK4/6 inhibitors were not only efficacious for advanced or metastatic breast cancer patients but also showed promise for early breast cancer patients [89, 90]. Thus, combination therapy still exhibits promising application prospects despite the risks of severe adverse events. In summary, our data provide a strong rationale for the clinical application of CDK4/6 inhibitors as radiation modifiers. However, more work remains to be done to achieve optimal clinical impacts.

Acknowledgements

Not applicable.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Moding EJ, Kastan MB, Kirsch DG. Strategies for optimizing the response of cancer and normal tissues to radiation. Nat Rev Drug Discov. 2013;12(7):526–42.PubMedPubMedCentralCrossRef Moding EJ, Kastan MB, Kirsch DG. Strategies for optimizing the response of cancer and normal tissues to radiation. Nat Rev Drug Discov. 2013;12(7):526–42.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Salehifar E, Hosseinimehr SJ. The use of cyclooxygenase-2 inhibitors for improvement of efficacy of radiotherapy in cancers. Drug Discov Today. 2016;21(4):654–62.PubMedCrossRef Salehifar E, Hosseinimehr SJ. The use of cyclooxygenase-2 inhibitors for improvement of efficacy of radiotherapy in cancers. Drug Discov Today. 2016;21(4):654–62.PubMedCrossRef
3.
Zurück zum Zitat Negi P, Kingsley PA, Srivastava H, Sharma SK. Three weekly versus weekly Cisplatin as radiosensitizer in head and neck cancer: a decision dilemma. Asian Pac J Cancer Prev. 2016;17(4):1617–23.PubMedCrossRef Negi P, Kingsley PA, Srivastava H, Sharma SK. Three weekly versus weekly Cisplatin as radiosensitizer in head and neck cancer: a decision dilemma. Asian Pac J Cancer Prev. 2016;17(4):1617–23.PubMedCrossRef
4.
Zurück zum Zitat Seiwert TY, Salama JK, Vokes EE. The concurrent chemoradiation paradigm--general principles. Nat Clin Pract Oncol. 2007;4(2):86–100.PubMedCrossRef Seiwert TY, Salama JK, Vokes EE. The concurrent chemoradiation paradigm--general principles. Nat Clin Pract Oncol. 2007;4(2):86–100.PubMedCrossRef
5.
Zurück zum Zitat Buckley AM, Lynam-Lennon N, O'Neill H, O'Sullivan J. Targeting hallmarks of cancer to enhance radiosensitivity in gastrointestinal cancers. Nat Rev Gastroenterol Hepatol. 2020;17(5):298–313.PubMedCrossRef Buckley AM, Lynam-Lennon N, O'Neill H, O'Sullivan J. Targeting hallmarks of cancer to enhance radiosensitivity in gastrointestinal cancers. Nat Rev Gastroenterol Hepatol. 2020;17(5):298–313.PubMedCrossRef
6.
Zurück zum Zitat Bosco EE, Knudsen ES. RB in breast cancer: at the crossroads of tumorigenesis and treatment. Cell Cycle. 2007;6(6):667–71.PubMedCrossRef Bosco EE, Knudsen ES. RB in breast cancer: at the crossroads of tumorigenesis and treatment. Cell Cycle. 2007;6(6):667–71.PubMedCrossRef
8.
Zurück zum Zitat VanArsdale T, Boshoff C, Arndt KT, Abraham RT. Molecular pathways: targeting the Cyclin D-CDK4/6 Axis for cancer treatment. Clin Cancer Res. 2015;21(13):2905–10.PubMedCrossRef VanArsdale T, Boshoff C, Arndt KT, Abraham RT. Molecular pathways: targeting the Cyclin D-CDK4/6 Axis for cancer treatment. Clin Cancer Res. 2015;21(13):2905–10.PubMedCrossRef
9.
10.
Zurück zum Zitat Lim S, Kaldis P. Cdks, cyclins and CKIs: roles beyond cell cycle regulation. Development. 2013;140(15):3079–93.PubMedCrossRef Lim S, Kaldis P. Cdks, cyclins and CKIs: roles beyond cell cycle regulation. Development. 2013;140(15):3079–93.PubMedCrossRef
12.
Zurück zum Zitat O'Leary B, Finn RS, Turner NC. Treating cancer with selective CDK4/6 inhibitors. Nat Rev Clin Oncol. 2016;13(7):417–30.PubMedCrossRef O'Leary B, Finn RS, Turner NC. Treating cancer with selective CDK4/6 inhibitors. Nat Rev Clin Oncol. 2016;13(7):417–30.PubMedCrossRef
13.
Zurück zum Zitat Lynce F, Shajahan-Haq AN, Swain SM. CDK4/6 inhibitors in breast cancer therapy: current practice and future opportunities. Pharmacol Ther. 2018;191:65–73.PubMedPubMedCentralCrossRef Lynce F, Shajahan-Haq AN, Swain SM. CDK4/6 inhibitors in breast cancer therapy: current practice and future opportunities. Pharmacol Ther. 2018;191:65–73.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Klein ME, Kovatcheva M, Davis LE, Tap WD, Koff A. CDK4/6 inhibitors: the mechanism of action may not be as simple as once thought. Cancer Cell. 2018;34(1):9–20.PubMedPubMedCentralCrossRef Klein ME, Kovatcheva M, Davis LE, Tap WD, Koff A. CDK4/6 inhibitors: the mechanism of action may not be as simple as once thought. Cancer Cell. 2018;34(1):9–20.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Fumagalli D, Wilson TR, Salgado R, Lu X, Yu J, O'Brien C, et al. Somatic mutation, copy number and transcriptomic profiles of primary and matched metastatic estrogen receptor-positive breast cancers. Ann Oncol. 2016;27(10):1860–6.PubMedCrossRef Fumagalli D, Wilson TR, Salgado R, Lu X, Yu J, O'Brien C, et al. Somatic mutation, copy number and transcriptomic profiles of primary and matched metastatic estrogen receptor-positive breast cancers. Ann Oncol. 2016;27(10):1860–6.PubMedCrossRef
16.
Zurück zum Zitat Lundgren K, Holm K, Nordenskjold B, Borg A, Landberg G. Gene products of chromosome 11q and their association with CCND1 gene amplification and tamoxifen resistance in premenopausal breast cancer. Breast Cancer Res. 2008;10(5):R81.PubMedPubMedCentralCrossRef Lundgren K, Holm K, Nordenskjold B, Borg A, Landberg G. Gene products of chromosome 11q and their association with CCND1 gene amplification and tamoxifen resistance in premenopausal breast cancer. Breast Cancer Res. 2008;10(5):R81.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Finn RS, Liu Y, Zhu Z, Martin M, Rugo HS, Dieras V, et al. Biomarker analyses of response to cyclin-dependent kinase 4/6 inhibition and endocrine therapy in women with treatment-naive metastatic breast cancer. Clin Cancer Res. 2020;26(1):110–21.PubMedCrossRef Finn RS, Liu Y, Zhu Z, Martin M, Rugo HS, Dieras V, et al. Biomarker analyses of response to cyclin-dependent kinase 4/6 inhibition and endocrine therapy in women with treatment-naive metastatic breast cancer. Clin Cancer Res. 2020;26(1):110–21.PubMedCrossRef
18.
Zurück zum Zitat Gopalan PK, Pinder MC, Chiappori A, Ivey AM, Villegas AG, Kaye FJ. A phase II clinical trial of the CDK 4/6 inhibitor palbociclib (PD 0332991) in previously treated, advanced non-small cell lung cancer (NSCLC) patients with inactivated CDKN2A. J Clin Oncol. 2014;32(15_suppl):8077.CrossRef Gopalan PK, Pinder MC, Chiappori A, Ivey AM, Villegas AG, Kaye FJ. A phase II clinical trial of the CDK 4/6 inhibitor palbociclib (PD 0332991) in previously treated, advanced non-small cell lung cancer (NSCLC) patients with inactivated CDKN2A. J Clin Oncol. 2014;32(15_suppl):8077.CrossRef
19.
Zurück zum Zitat Goldman JW, Gandhi L, Patnaik A, Rosen LS, Hilton JF, Papadopoulos KP, et al. Clinical activity of LY2835219, a novel cell cycle inhibitor selective for CDK4 and CDK6, in patients with non-small cell lung cancer. J Clin Oncol. 2014;32(15_suppl):8026.CrossRef Goldman JW, Gandhi L, Patnaik A, Rosen LS, Hilton JF, Papadopoulos KP, et al. Clinical activity of LY2835219, a novel cell cycle inhibitor selective for CDK4 and CDK6, in patients with non-small cell lung cancer. J Clin Oncol. 2014;32(15_suppl):8026.CrossRef
20.
Zurück zum Zitat Narayan V, Hwang WT, Lal P, Rosen MA, Gallagher M, O'Dwyer PJ, et al. Cyclin-dependent kinase 4/6 inhibition for the treatment of unresectable mature teratoma: long-term follow-up of a phase II study. Clin Genitourin Cancer. 2016;14(6):504–10.PubMedCrossRef Narayan V, Hwang WT, Lal P, Rosen MA, Gallagher M, O'Dwyer PJ, et al. Cyclin-dependent kinase 4/6 inhibition for the treatment of unresectable mature teratoma: long-term follow-up of a phase II study. Clin Genitourin Cancer. 2016;14(6):504–10.PubMedCrossRef
21.
Zurück zum Zitat Tripathy D, Im SA, Colleoni M, Franke F, Bardia A, Harbeck N, 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(7):904–15.PubMedCrossRef Tripathy D, Im SA, Colleoni M, Franke F, Bardia A, Harbeck N, 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(7):904–15.PubMedCrossRef
22.
Zurück zum Zitat Sledge GW Jr, Toi M, Neven P, Sohn J, Inoue K, Pivot X, 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(25):2875–84.PubMedCrossRef Sledge GW Jr, Toi M, Neven P, Sohn J, Inoue K, Pivot X, 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(25):2875–84.PubMedCrossRef
23.
Zurück zum Zitat Slamon DJ, Neven P, Chia S, Fasching PA, De Laurentiis M, Im SA, 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(24):2465–72.PubMedCrossRef Slamon DJ, Neven P, Chia S, Fasching PA, De Laurentiis M, Im SA, 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(24):2465–72.PubMedCrossRef
24.
Zurück zum Zitat Goetz MP, Toi M, Campone M, Sohn J, Paluch-Shimon S, Huober J, et al. MONARCH 3: Abemaciclib as initial therapy for advanced breast cancer. J Clin Oncol. 2017;35(32):3638–46.PubMedCrossRef Goetz MP, Toi M, Campone M, Sohn J, Paluch-Shimon S, Huober J, et al. MONARCH 3: Abemaciclib as initial therapy for advanced breast cancer. J Clin Oncol. 2017;35(32):3638–46.PubMedCrossRef
25.
Zurück zum Zitat Cristofanilli M, Turner NC, Bondarenko I, Ro J, Im SA, Masuda N, 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(4):425–39.PubMedCrossRef Cristofanilli M, Turner NC, Bondarenko I, Ro J, Im SA, Masuda N, 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(4):425–39.PubMedCrossRef
26.
Zurück zum Zitat Finn RS, Martin M, Rugo HS, Jones S, Im SA, Gelmon K, et al. Palbociclib and Letrozole in advanced breast cancer. N Engl J Med. 2016;375(20):1925–36.CrossRefPubMed Finn RS, Martin M, Rugo HS, Jones S, Im SA, Gelmon K, et al. Palbociclib and Letrozole in advanced breast cancer. N Engl J Med. 2016;375(20):1925–36.CrossRefPubMed
27.
Zurück zum Zitat Wong CH, Ma BBY, Hui CWC, Lo KW, Hui EP, Chan ATC. Preclinical evaluation of ribociclib and its synergistic effect in combination with alpelisib in non-keratinizing nasopharyngeal carcinoma. Sci Rep. 2018;8(1):8010.PubMedPubMedCentralCrossRef Wong CH, Ma BBY, Hui CWC, Lo KW, Hui EP, Chan ATC. Preclinical evaluation of ribociclib and its synergistic effect in combination with alpelisib in non-keratinizing nasopharyngeal carcinoma. Sci Rep. 2018;8(1):8010.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Vora SR, Juric D, Kim N, Mino-Kenudson M, Huynh T, Costa C, et al. CDK 4/6 inhibitors sensitize PIK3CA mutant breast cancer to PI3K inhibitors. Cancer Cell. 2014;26(1):136–49.PubMedPubMedCentralCrossRef Vora SR, Juric D, Kim N, Mino-Kenudson M, Huynh T, Costa C, et al. CDK 4/6 inhibitors sensitize PIK3CA mutant breast cancer to PI3K inhibitors. Cancer Cell. 2014;26(1):136–49.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Teo ZL, Versaci S, Dushyanthen S, Caramia F, Savas P, Mintoff CP, et al. Combined CDK4/6 and PI3Kalpha inhibition is synergistic and immunogenic in triple-negative breast cancer. Cancer Res. 2017;77(22):6340–52.PubMedCrossRef Teo ZL, Versaci S, Dushyanthen S, Caramia F, Savas P, Mintoff CP, et al. Combined CDK4/6 and PI3Kalpha inhibition is synergistic and immunogenic in triple-negative breast cancer. Cancer Res. 2017;77(22):6340–52.PubMedCrossRef
30.
Zurück zum Zitat Tao Z, Le Blanc JM, Wang C, Zhan T, Zhuang H, Wang P, et al. Coadministration of trametinib and palbociclib radiosensitizes KRAS-mutant non-small cell lung cancers in vitro and in vivo. Clin Cancer Res. 2016;22(1):122–33.PubMedCrossRef Tao Z, Le Blanc JM, Wang C, Zhan T, Zhuang H, Wang P, et al. Coadministration of trametinib and palbociclib radiosensitizes KRAS-mutant non-small cell lung cancers in vitro and in vivo. Clin Cancer Res. 2016;22(1):122–33.PubMedCrossRef
31.
Zurück zum Zitat Chen SH, Gong X, Zhang Y, Van Horn RD, Yin T, Huber L, et al. RAF inhibitor LY3009120 sensitizes RAS or BRAF mutant cancer to CDK4/6 inhibition by abemaciclib via superior inhibition of phospho-RB and suppression of cyclin D1. Oncogene. 2018;37(6):821–32.PubMedCrossRef Chen SH, Gong X, Zhang Y, Van Horn RD, Yin T, Huber L, et al. RAF inhibitor LY3009120 sensitizes RAS or BRAF mutant cancer to CDK4/6 inhibition by abemaciclib via superior inhibition of phospho-RB and suppression of cyclin D1. Oncogene. 2018;37(6):821–32.PubMedCrossRef
32.
Zurück zum Zitat Ziemke EK, Dosch JS, Maust JD, Shettigar A, Sen A, Welling TH, et al. Sensitivity of KRAS-mutant colorectal cancers to combination therapy that cotargets MEK and CDK4/6. Clin Cancer Res. 2016;22(2):405–14.PubMedCrossRef Ziemke EK, Dosch JS, Maust JD, Shettigar A, Sen A, Welling TH, et al. Sensitivity of KRAS-mutant colorectal cancers to combination therapy that cotargets MEK and CDK4/6. Clin Cancer Res. 2016;22(2):405–14.PubMedCrossRef
33.
Zurück zum Zitat Zacharek SJ, Xiong Y, Shumway SD. Negative regulation of TSC1-TSC2 by mammalian D-type cyclins. Cancer Res. 2005;65(24):11354–60.PubMedCrossRef Zacharek SJ, Xiong Y, Shumway SD. Negative regulation of TSC1-TSC2 by mammalian D-type cyclins. Cancer Res. 2005;65(24):11354–60.PubMedCrossRef
34.
Zurück zum Zitat Imperial R, Toor OM, Hussain A, Subramanian J, Masood A. Comprehensive pancancer genomic analysis reveals (RTK)-RAS-RAF-MEK as a key dysregulated pathway in cancer: its clinical implications. Semin Cancer Biol. 2019;54:14–28.PubMedCrossRef Imperial R, Toor OM, Hussain A, Subramanian J, Masood A. Comprehensive pancancer genomic analysis reveals (RTK)-RAS-RAF-MEK as a key dysregulated pathway in cancer: its clinical implications. Semin Cancer Biol. 2019;54:14–28.PubMedCrossRef
35.
Zurück zum Zitat Ma L, Chen Z, Erdjument-Bromage H, Tempst P, Pandolfi PP. Phosphorylation and functional inactivation of TSC2 by Erk implications for tuberous sclerosis and cancer pathogenesis. Cell. 2005;121(2):179–93.PubMedCrossRef Ma L, Chen Z, Erdjument-Bromage H, Tempst P, Pandolfi PP. Phosphorylation and functional inactivation of TSC2 by Erk implications for tuberous sclerosis and cancer pathogenesis. Cell. 2005;121(2):179–93.PubMedCrossRef
36.
Zurück zum Zitat Roux PP, Ballif BA, Anjum R, Gygi SP, Blenis J. Tumor-promoting phorbol esters and activated Ras inactivate the tuberous sclerosis tumor suppressor complex via p90 ribosomal S6 kinase. Proc Natl Acad Sci U S A. 2004;101(37):13489–94.PubMedPubMedCentralCrossRef Roux PP, Ballif BA, Anjum R, Gygi SP, Blenis J. Tumor-promoting phorbol esters and activated Ras inactivate the tuberous sclerosis tumor suppressor complex via p90 ribosomal S6 kinase. Proc Natl Acad Sci U S A. 2004;101(37):13489–94.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Dean JL, McClendon AK, Knudsen ES. Modification of the DNA damage response by therapeutic CDK4/6 inhibition. J Biol Chem. 2012;287(34):29075–87.PubMedPubMedCentralCrossRef Dean JL, McClendon AK, Knudsen ES. Modification of the DNA damage response by therapeutic CDK4/6 inhibition. J Biol Chem. 2012;287(34):29075–87.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Finn RS, Crown JP, Lang I, Boer K, Bondarenko IM, Kulyk SO, 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(1):25–35.PubMedCrossRef Finn RS, Crown JP, Lang I, Boer K, Bondarenko IM, Kulyk SO, 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(1):25–35.PubMedCrossRef
39.
Zurück zum Zitat Cancer Genome Atlas N. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490(7418):61–70.CrossRef Cancer Genome Atlas N. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490(7418):61–70.CrossRef
41.
Zurück zum Zitat Whiteway SL, Harris PS, Venkataraman S, Alimova I, Birks DK, Donson AM, et al. Inhibition of cyclin-dependent kinase 6 suppresses cell proliferation and enhances radiation sensitivity in medulloblastoma cells. J Neuro-Oncol. 2013;111(2):113–21.CrossRef Whiteway SL, Harris PS, Venkataraman S, Alimova I, Birks DK, Donson AM, et al. Inhibition of cyclin-dependent kinase 6 suppresses cell proliferation and enhances radiation sensitivity in medulloblastoma cells. J Neuro-Oncol. 2013;111(2):113–21.CrossRef
42.
Zurück zum Zitat Hashizume R, Zhang A, Mueller S, Prados MD, Lulla RR, Goldman S, et al. Inhibition of DNA damage repair by the CDK4/6 inhibitor palbociclib delays irradiated intracranial atypical teratoid rhabdoid tumor and glioblastoma xenograft regrowth. Neuro-Oncology. 2016;18(11):1519–28.PubMedPubMedCentral Hashizume R, Zhang A, Mueller S, Prados MD, Lulla RR, Goldman S, et al. Inhibition of DNA damage repair by the CDK4/6 inhibitor palbociclib delays irradiated intracranial atypical teratoid rhabdoid tumor and glioblastoma xenograft regrowth. Neuro-Oncology. 2016;18(11):1519–28.PubMedPubMedCentral
43.
Zurück zum Zitat Whittaker S, Madani D, Joshi S, Chung SA, Johns T, Day B, et al. Combination of palbociclib and radiotherapy for glioblastoma. Cell Death Dis. 2017;3:17033.CrossRef Whittaker S, Madani D, Joshi S, Chung SA, Johns T, Day B, et al. Combination of palbociclib and radiotherapy for glioblastoma. Cell Death Dis. 2017;3:17033.CrossRef
44.
Zurück zum Zitat Naz S, Sowers A, Choudhuri R, Wissler M, Gamson J, Mathias A, et al. Abemaciclib, a selective CDK4/6 inhibitor, enhances the radiosensitivity of non-small cell lung cancer in vitro and in vivo. Clin Cancer Res. 2018;24(16):3994–4005.PubMedPubMedCentralCrossRef Naz S, Sowers A, Choudhuri R, Wissler M, Gamson J, Mathias A, et al. Abemaciclib, a selective CDK4/6 inhibitor, enhances the radiosensitivity of non-small cell lung cancer in vitro and in vivo. Clin Cancer Res. 2018;24(16):3994–4005.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Huang CY, Hsieh FS, Wang CY, Chen LJ, Chang SS, Tsai MH, et al. Palbociclib enhances radiosensitivity of hepatocellular carcinoma and cholangiocarcinoma via inhibiting ataxia telangiectasia-mutated kinase-mediated DNA damage response. Eur J Cancer. 2018;102:10–22.PubMedCrossRef Huang CY, Hsieh FS, Wang CY, Chen LJ, Chang SS, Tsai MH, et al. Palbociclib enhances radiosensitivity of hepatocellular carcinoma and cholangiocarcinoma via inhibiting ataxia telangiectasia-mutated kinase-mediated DNA damage response. Eur J Cancer. 2018;102:10–22.PubMedCrossRef
46.
Zurück zum Zitat Xie X, Zheng W, Chen T, Lin W, Liao Z, Liu J, et al. CDK4/6 inhibitor palbociclib amplifies the radiosensitivity to nasopharyngeal carcinoma cells via mediating apoptosis and suppressing DNA damage repair. Onco Targets Ther. 2019;12:11107–17.PubMedPubMedCentralCrossRef Xie X, Zheng W, Chen T, Lin W, Liao Z, Liu J, et al. CDK4/6 inhibitor palbociclib amplifies the radiosensitivity to nasopharyngeal carcinoma cells via mediating apoptosis and suppressing DNA damage repair. Onco Targets Ther. 2019;12:11107–17.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Gottgens EL, Bussink J, Leszczynska KB, Peters H, Span PN, Hammond EM. Inhibition of CDK4/CDK6 enhances radiosensitivity of HPV negative head and neck squamous cell carcinomas. Int J Radiat Oncol Biol Phys. 2019;105(3):548–58.PubMedCrossRef Gottgens EL, Bussink J, Leszczynska KB, Peters H, Span PN, Hammond EM. Inhibition of CDK4/CDK6 enhances radiosensitivity of HPV negative head and neck squamous cell carcinomas. Int J Radiat Oncol Biol Phys. 2019;105(3):548–58.PubMedCrossRef
48.
Zurück zum Zitat Tai TS, Lin PM, Wu CF, Hung SK, Huang CI, Wang CC, et al. CDK4/6 inhibitor LEE011 is a potential radiation-sensitizer in head and neck squamous cell carcinoma: an in vitro study. Anticancer Res. 2019;39(2):713–20.PubMedCrossRef Tai TS, Lin PM, Wu CF, Hung SK, Huang CI, Wang CC, et al. CDK4/6 inhibitor LEE011 is a potential radiation-sensitizer in head and neck squamous cell carcinoma: an in vitro study. Anticancer Res. 2019;39(2):713–20.PubMedCrossRef
49.
Zurück zum Zitat Fernandez-Aroca DM, Roche O, Sabater S, Pascual-Serra R, Ortega-Muelas M, Sanchez Perez I, et al. P53 pathway is a major determinant in the radiosensitizing effect of Palbociclib: implication in cancer therapy. Cancer Lett. 2019;451:23–33.PubMedCrossRef Fernandez-Aroca DM, Roche O, Sabater S, Pascual-Serra R, Ortega-Muelas M, Sanchez Perez I, et al. P53 pathway is a major determinant in the radiosensitizing effect of Palbociclib: implication in cancer therapy. Cancer Lett. 2019;451:23–33.PubMedCrossRef
50.
Zurück zum Zitat Li F, Xu Y, Liu B, Singh PK, Zhao W, Jin J, et al. YAP1-mediated CDK6 activation confers radiation resistance in esophageal cancer - rationale for the combination of YAP1 and CDK4/6 inhibitors in esophageal cancer. Clin Cancer Res. 2019;25(7):2264–77.PubMedCrossRef Li F, Xu Y, Liu B, Singh PK, Zhao W, Jin J, et al. YAP1-mediated CDK6 activation confers radiation resistance in esophageal cancer - rationale for the combination of YAP1 and CDK4/6 inhibitors in esophageal cancer. Clin Cancer Res. 2019;25(7):2264–77.PubMedCrossRef
51.
Zurück zum Zitat Ogawa K, Yoshioka Y, Isohashi F, Seo Y, Yoshida K, Yamazaki H. Radiotherapy targeting cancer stem cells: current views and future perspectives. Anticancer Res. 2013;33(3):747–54.PubMed Ogawa K, Yoshioka Y, Isohashi F, Seo Y, Yoshida K, Yamazaki H. Radiotherapy targeting cancer stem cells: current views and future perspectives. Anticancer Res. 2013;33(3):747–54.PubMed
52.
Zurück zum Zitat Barton KL, Misuraca K, Cordero F, Dobrikova E, Min HD, Gromeier M, et al. PD-0332991, a CDK4/6 inhibitor, significantly prolongs survival in a genetically engineered mouse model of brainstem glioma. PLoS One. 2013;8(10):e77639.PubMedPubMedCentralCrossRef Barton KL, Misuraca K, Cordero F, Dobrikova E, Min HD, Gromeier M, et al. PD-0332991, a CDK4/6 inhibitor, significantly prolongs survival in a genetically engineered mouse model of brainstem glioma. PLoS One. 2013;8(10):e77639.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Goldstein M, Kastan MB. The DNA damage response: implications for tumor responses to radiation and chemotherapy. Annu Rev Med. 2015;66:129–43.PubMedCrossRef Goldstein M, Kastan MB. The DNA damage response: implications for tumor responses to radiation and chemotherapy. Annu Rev Med. 2015;66:129–43.PubMedCrossRef
54.
Zurück zum Zitat Morgan MA, Lawrence TS. Molecular pathways: overcoming radiation resistance by targeting DNA damage response pathways. Clin Cancer Res. 2015;21(13):2898–904.PubMedPubMedCentralCrossRef Morgan MA, Lawrence TS. Molecular pathways: overcoming radiation resistance by targeting DNA damage response pathways. Clin Cancer Res. 2015;21(13):2898–904.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Willers H, Gheorghiu L, Liu Q, Efstathiou JA, Wirth LJ, Krause M, et al. DNA damage response assessments in human tumor samples provide functional biomarkers of radiosensitivity. Semin Radiat Oncol. 2015;25(4):237–50.PubMedPubMedCentralCrossRef Willers H, Gheorghiu L, Liu Q, Efstathiou JA, Wirth LJ, Krause M, et al. DNA damage response assessments in human tumor samples provide functional biomarkers of radiosensitivity. Semin Radiat Oncol. 2015;25(4):237–50.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Gudkov AV, Komarova EA. The role of p53 in determining sensitivity to radiotherapy. Nat Rev Cancer. 2003;3(2):117–29.PubMedCrossRef Gudkov AV, Komarova EA. The role of p53 in determining sensitivity to radiotherapy. Nat Rev Cancer. 2003;3(2):117–29.PubMedCrossRef
60.
Zurück zum Zitat Pavlopoulou A, Bagos PG, Koutsandrea V, Georgakilas AG. Molecular determinants of radiosensitivity in normal and tumor tissue: a bioinformatic approach. Cancer Lett. 2017;403:37–47.PubMedCrossRef Pavlopoulou A, Bagos PG, Koutsandrea V, Georgakilas AG. Molecular determinants of radiosensitivity in normal and tumor tissue: a bioinformatic approach. Cancer Lett. 2017;403:37–47.PubMedCrossRef
61.
Zurück zum Zitat Hagen KR, Zeng X, Lee MY, Tucker Kahn S, Harrison Pitner MK, Zaky SS, et al. Silencing CDK4 radiosensitizes breast cancer cells by promoting apoptosis. Cell Div. 2013;8(1):10.PubMedPubMedCentralCrossRef Hagen KR, Zeng X, Lee MY, Tucker Kahn S, Harrison Pitner MK, Zaky SS, et al. Silencing CDK4 radiosensitizes breast cancer cells by promoting apoptosis. Cell Div. 2013;8(1):10.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Preuss U, Landsberg G, Scheidtmann KH. Novel mitosis-specific phosphorylation of histone H3 at Thr11 mediated by Dlk/ZIP kinase. Nucleic Acids Res. 2003;31(3):878–85.PubMedPubMedCentralCrossRef Preuss U, Landsberg G, Scheidtmann KH. Novel mitosis-specific phosphorylation of histone H3 at Thr11 mediated by Dlk/ZIP kinase. Nucleic Acids Res. 2003;31(3):878–85.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Thangavel C, Dean JL, Ertel A, Knudsen KE, Aldaz CM, Witkiewicz AK, et al. Therapeutically activating RB: reestablishing cell cycle control in endocrine therapy-resistant breast cancer. Endocr Relat Cancer. 2011;18(3):333–45.PubMedPubMedCentralCrossRef Thangavel C, Dean JL, Ertel A, Knudsen KE, Aldaz CM, Witkiewicz AK, et al. Therapeutically activating RB: reestablishing cell cycle control in endocrine therapy-resistant breast cancer. Endocr Relat Cancer. 2011;18(3):333–45.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Rader J, Russell MR, Hart LS, Nakazawa MS, Belcastro LT, Martinez D, et al. Dual CDK4/CDK6 inhibition induces cell-cycle arrest and senescence in neuroblastoma. Clin Cancer Res. 2013;19(22):6173–82.PubMedCrossRef Rader J, Russell MR, Hart LS, Nakazawa MS, Belcastro LT, Martinez D, et al. Dual CDK4/CDK6 inhibition induces cell-cycle arrest and senescence in neuroblastoma. Clin Cancer Res. 2013;19(22):6173–82.PubMedCrossRef
65.
Zurück zum Zitat Yoshida A, Lee EK, Diehl JA. Induction of therapeutic senescence in Vemurafenib-resistant melanoma by extended inhibition of CDK4/6. Cancer Res. 2016;76(10):2990–3002.PubMedPubMedCentralCrossRef Yoshida A, Lee EK, Diehl JA. Induction of therapeutic senescence in Vemurafenib-resistant melanoma by extended inhibition of CDK4/6. Cancer Res. 2016;76(10):2990–3002.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Munoz-Espin D, Serrano M. Cellular senescence: from physiology to pathology. Nat Rev Mol Cell Biol. 2014;15(7):482–96.CrossRefPubMed Munoz-Espin D, Serrano M. Cellular senescence: from physiology to pathology. Nat Rev Mol Cell Biol. 2014;15(7):482–96.CrossRefPubMed
68.
Zurück zum Zitat Mavrogonatou E, Pratsinis H, Kletsas D. The role of senescence in cancer development. Semin Cancer Biol. 2020;62:182–91.PubMedCrossRef Mavrogonatou E, Pratsinis H, Kletsas D. The role of senescence in cancer development. Semin Cancer Biol. 2020;62:182–91.PubMedCrossRef
69.
Zurück zum Zitat Hernandez-Segura A, Nehme J, Demaria M. Hallmarks of cellular senescence. Trends Cell Biol. 2018;28(6):436–53.PubMedCrossRef Hernandez-Segura A, Nehme J, Demaria M. Hallmarks of cellular senescence. Trends Cell Biol. 2018;28(6):436–53.PubMedCrossRef
70.
Zurück zum Zitat Mirzayans R, Scott A, Cameron M, Murray D. Induction of accelerated senescence by gamma radiation in human solid tumor-derived cell lines expressing wild-type TP53. Radiat Res. 2005;163(1):53–62.PubMedCrossRef Mirzayans R, Scott A, Cameron M, Murray D. Induction of accelerated senescence by gamma radiation in human solid tumor-derived cell lines expressing wild-type TP53. Radiat Res. 2005;163(1):53–62.PubMedCrossRef
71.
Zurück zum Zitat Jones KR, Elmore LW, Jackson-Cook C, Demasters G, Povirk LF, Holt SE, et al. p53-dependent accelerated senescence induced by ionizing radiation in breast tumour cells. Int J Radiat Biol. 2005;81(6):445–58.PubMedCrossRef Jones KR, Elmore LW, Jackson-Cook C, Demasters G, Povirk LF, Holt SE, et al. p53-dependent accelerated senescence induced by ionizing radiation in breast tumour cells. Int J Radiat Biol. 2005;81(6):445–58.PubMedCrossRef
72.
Zurück zum Zitat Faget DV, Ren Q, Stewart SA. Unmasking senescence: context-dependent effects of SASP in cancer. Nat Rev Cancer. 2019;19(8):439–53.PubMedCrossRef Faget DV, Ren Q, Stewart SA. Unmasking senescence: context-dependent effects of SASP in cancer. Nat Rev Cancer. 2019;19(8):439–53.PubMedCrossRef
73.
Zurück zum Zitat Weichselbaum RR, Liang H, Deng L, Fu YX. Radiotherapy and immunotherapy: a beneficial liaison? Nat Rev Clin Oncol. 2017;14(6):365–79.PubMedCrossRef Weichselbaum RR, Liang H, Deng L, Fu YX. Radiotherapy and immunotherapy: a beneficial liaison? Nat Rev Clin Oncol. 2017;14(6):365–79.PubMedCrossRef
75.
Zurück zum Zitat Goel S, DeCristo MJ, Watt AC, BrinJones H, Sceneay J, Li BB, et al. CDK4/6 inhibition triggers anti-tumour immunity. Nature. 2017;548(7668):471–5.PubMedPubMedCentralCrossRef Goel S, DeCristo MJ, Watt AC, BrinJones H, Sceneay J, Li BB, et al. CDK4/6 inhibition triggers anti-tumour immunity. Nature. 2017;548(7668):471–5.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Sharabi AB, Lim M, DeWeese TL, Drake CG. Radiation and checkpoint blockade immunotherapy: radiosensitisation and potential mechanisms of synergy. Lancet Oncol. 2015;16(13):e498–509.CrossRefPubMed Sharabi AB, Lim M, DeWeese TL, Drake CG. Radiation and checkpoint blockade immunotherapy: radiosensitisation and potential mechanisms of synergy. Lancet Oncol. 2015;16(13):e498–509.CrossRefPubMed
77.
Zurück zum Zitat Gupta A, Probst HC, Vuong V, Landshammer A, Muth S, Yagita H, et al. Radiotherapy promotes tumor-specific effector CD8+ T cells via dendritic cell activation. J Immunol. 2012;189(2):558–66.PubMedCrossRef Gupta A, Probst HC, Vuong V, Landshammer A, Muth S, Yagita H, et al. Radiotherapy promotes tumor-specific effector CD8+ T cells via dendritic cell activation. J Immunol. 2012;189(2):558–66.PubMedCrossRef
78.
Zurück zum Zitat Deng J, Wang ES, Jenkins RW, Li S, Dries R, Yates K, et al. CDK4/6 inhibition augments antitumor immunity by enhancing T-cell activation. Cancer Discov. 2018;8(2):216–33.PubMedCrossRef Deng J, Wang ES, Jenkins RW, Li S, Dries R, Yates K, et al. CDK4/6 inhibition augments antitumor immunity by enhancing T-cell activation. Cancer Discov. 2018;8(2):216–33.PubMedCrossRef
79.
Zurück zum Zitat Schaer DA, Beckmann RP, Dempsey JA, Huber L, Forest A, Amaladas N, et al. The CDK4/6 inhibitor Abemaciclib induces a T cell inflamed tumor microenvironment and enhances the efficacy of PD-L1 checkpoint blockade. Cell Rep. 2018;22(11):2978–94.PubMedCrossRef Schaer DA, Beckmann RP, Dempsey JA, Huber L, Forest A, Amaladas N, et al. The CDK4/6 inhibitor Abemaciclib induces a T cell inflamed tumor microenvironment and enhances the efficacy of PD-L1 checkpoint blockade. Cell Rep. 2018;22(11):2978–94.PubMedCrossRef
80.
Zurück zum Zitat Lhuillier C, Rudqvist N-P, Elemento O, Formenti SC, Demaria S. Radiation therapy and anti-tumor immunity: exposing immunogenic mutations to the immune system. Genome Med. 2019;11(1):40.PubMedPubMedCentralCrossRef Lhuillier C, Rudqvist N-P, Elemento O, Formenti SC, Demaria S. Radiation therapy and anti-tumor immunity: exposing immunogenic mutations to the immune system. Genome Med. 2019;11(1):40.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Lugade AA, Moran JP, Gerber SA, Rose RC, Frelinger JG, Lord EM. Local radiation therapy of B16 melanoma tumors increases the generation of tumor antigen-specific effector cells that traffic to the tumor. J Immunol. 2005;174(12):7516–23.PubMedCrossRef Lugade AA, Moran JP, Gerber SA, Rose RC, Frelinger JG, Lord EM. Local radiation therapy of B16 melanoma tumors increases the generation of tumor antigen-specific effector cells that traffic to the tumor. J Immunol. 2005;174(12):7516–23.PubMedCrossRef
82.
Zurück zum Zitat Lee Y, Auh SL, Wang Y, Burnette B, Wang Y, Meng Y, et al. Therapeutic effects of ablative radiation on local tumor require CD8+ T cells: changing strategies for cancer treatment. Blood. 2009;114(3):589–95.PubMedPubMedCentralCrossRef Lee Y, Auh SL, Wang Y, Burnette B, Wang Y, Meng Y, et al. Therapeutic effects of ablative radiation on local tumor require CD8+ T cells: changing strategies for cancer treatment. Blood. 2009;114(3):589–95.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Hans S, Cottu P, Kirova YM. Preliminary results of the association of Palbociclib and radiotherapy in metastatic breast cancer patients. Radiother Oncol. 2018;126(1):181.PubMedCrossRef Hans S, Cottu P, Kirova YM. Preliminary results of the association of Palbociclib and radiotherapy in metastatic breast cancer patients. Radiother Oncol. 2018;126(1):181.PubMedCrossRef
84.
Zurück zum Zitat Ippolito E, Greco C, Silipigni S, Dell'Aquila E, Petrianni GM, Tonini G, et al. Concurrent radiotherapy with palbociclib or ribociclib for metastatic breast cancer patients: preliminary assessment of toxicity. Breast. 2019;46:70–4.PubMedCrossRef Ippolito E, Greco C, Silipigni S, Dell'Aquila E, Petrianni GM, Tonini G, et al. Concurrent radiotherapy with palbociclib or ribociclib for metastatic breast cancer patients: preliminary assessment of toxicity. Breast. 2019;46:70–4.PubMedCrossRef
85.
Zurück zum Zitat Figura NB, Potluri TK, Mohammadi H, Oliver DE, Arrington JA, Robinson TJ, et al. CDK 4/6 inhibitors and stereotactic radiation in the management of hormone receptor positive breast cancer brain metastases. J Neuro-Oncol. 2019;144(3):583–9.CrossRef Figura NB, Potluri TK, Mohammadi H, Oliver DE, Arrington JA, Robinson TJ, et al. CDK 4/6 inhibitors and stereotactic radiation in the management of hormone receptor positive breast cancer brain metastases. J Neuro-Oncol. 2019;144(3):583–9.CrossRef
86.
Zurück zum Zitat Chowdhary M, Sen N, Chowdhary A, Usha L, Cobleigh MA, Wang D, et al. Safety and efficacy of Palbociclib and radiation therapy in patients with metastatic breast cancer: initial results of a novel combination. Adv Radiat Oncol. 2019;4(3):453–7.PubMedPubMedCentralCrossRef Chowdhary M, Sen N, Chowdhary A, Usha L, Cobleigh MA, Wang D, et al. Safety and efficacy of Palbociclib and radiation therapy in patients with metastatic breast cancer: initial results of a novel combination. Adv Radiat Oncol. 2019;4(3):453–7.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Messer JA, Ekinci E, Patel TA, Teh BS. Enhanced dermatologic toxicity following concurrent treatment with palbociclib and radiation therapy: a case report. Rep Pract Oncol Radiother. 2019;24(3):276–80.PubMedPubMedCentralCrossRef Messer JA, Ekinci E, Patel TA, Teh BS. Enhanced dermatologic toxicity following concurrent treatment with palbociclib and radiation therapy: a case report. Rep Pract Oncol Radiother. 2019;24(3):276–80.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Kawamoto T, Shikama N, Sasai K. Severe acute radiation-induced enterocolitis after combined palbociclib and palliative radiotherapy treatment. Radiother Oncol. 2019;131:240–1.PubMedCrossRef Kawamoto T, Shikama N, Sasai K. Severe acute radiation-induced enterocolitis after combined palbociclib and palliative radiotherapy treatment. Radiother Oncol. 2019;131:240–1.PubMedCrossRef
89.
Zurück zum Zitat Hurvitz SA, Martin M, Press MF, Chan D, Fernandez-Abad M, Petru E, 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(3):566–80.PubMedCrossRef Hurvitz SA, Martin M, Press MF, Chan D, Fernandez-Abad M, Petru E, 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(3):566–80.PubMedCrossRef
90.
Zurück zum Zitat Colwell J. Abemaciclib shows promise for early breast cancer. Cancer Discov. 2017;7(2):119–20.CrossRef Colwell J. Abemaciclib shows promise for early breast cancer. Cancer Discov. 2017;7(2):119–20.CrossRef
Metadaten
Titel
CDK4/6 inhibitors: a novel strategy for tumor radiosensitization
verfasst von
Yilan Yang
Jurui Luo
Xingxing Chen
Zhaozhi Yang
Xin Mei
Jinli Ma
Zhen Zhang
Xiaomao Guo
Xiaoli Yu
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2020
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-020-01693-w

Weitere Artikel der Ausgabe 1/2020

Journal of Experimental & Clinical Cancer Research 1/2020 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.