Dysregulation of cell division resulting in aberrant cell proliferation is a key hallmark of cancer, making it a rational and important target for innovative anticancer drug development. Three selective cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitors are FDA and European Medicines Agency (EMA) approved for hormone receptor–positive/HER2-negative advanced breast cancer. A major emerging appreciation is that these inhibitors not only are cytostatic, but also play critical roles in the interaction between tumor cells and the host immune response. However, to trigger an effective immune response, lymphocytes must also proliferate. This review aims to assimilate our emerging understanding on the role of CDK4/6 inhibitors in cell-cycle control, as well as their biological effect on T cells and other key immune cells, and the confluence of preclinical evidence of augmentation of anticancer immunity by these drugs. We aim to provide a framework for understanding the role of the cell cycle in anticancer immunity, discussing ongoing clinical trials evaluating this concept and challenges for developing rational combinations with immunotherapy.

The mammalian cell cycle is a highly organized and regulated process that ensures duplication of genetic material and cell division (1). Key features of this process are cascades of growth-regulatory signals and signaling proteins that monitor genetic integrity. Proliferation depends on progression from the quiescent state (G0) through four distinct phases: G1 (the first gap phase), S-phase (DNA synthesis), G2 (the second gap phase), and M (mitosis)—which is controlled at checkpoints by cyclins and their associated cyclin-dependent kinases (CDK; ref. 2). CDKs 4 and 6 (CDK4/6) are fundamental drivers of the cell cycle and required for entry into, and progression through, G1.

Unsurprisingly, this intricate process is disrupted in most cancers (3), either as a result of mutations in upstream signaling pathways or by defects in genes encoding cell-cycle proteins (reviewed in ref. 4). Specific inhibitors of CDK4/6 have been touted as paradigm-shifting with recent FDA and European Medicines Agency (EMA) approval for three orally available inhibitors—palbociclib (PD-0332991; Ibrance; Pfizer), ribociclib (LEE011; Kisqali; Novartis), and abemaciclib (LY2835219; Verzenio; Lilly; refs. 5–7). In contrast to traditional chemotherapeutic agents, CDK4/6 inhibitors arrest progression through G1, promoting transient quiescence or inducing senescence, and have shown significant clinical benefit in combination with aromatase inhibitors; the selective estrogen receptor (ER) degrader fulvestrant; and tamoxifen (8).

Translational outputs from these ongoing trials have unexpectedly revealed effects of CDK4/6 inhibitors in several critical roles underpinning the interactions of cancer cells with the host immune system (9–11). The cell-cycle cascade couples two processes that are required for the generation of an effective adaptive immune response: clonal expansion and differentiation, and consequently, CDK inhibitors have the potential to participate in the decision between tolerance, anergy, and the promotion of antitumor immunity.

In this review, we discuss the biological functions of the CDK4–CDK6–Retinoblastoma (CDK4/6–Rb) axis—both when pathologically hijacked in cancer and physiologically in immune cells, with a view to providing a framework for understanding the role of the cell cycle in anticancer immunity. We discuss emerging preclinical and clinical data showing effects of CDK4/6 inhibition on promoting various aspects of antitumor immunity including enhancing antigen presentation, depleting immunosuppressive regulatory T cells, and ultimately shifting the balance toward the generation of an efficient antitumor immune response. We also ponder the challenges faced by ongoing clinical trials attempting to therapeutically target these together with immunotherapy.

Numerous detailed reviews of this pathway are available (4, 12). This review will therefore focus on the major principles of this axis. Quiescent cells in G0 can be triggered to re-enter the cell cycle through stimulation by a variety of mitogenic factors that activate intricate intracellular signaling networks that are “sensed” by the holoenzyme complex of Cyclin D and CDK4 and/or CDK6 (ref. 4; Fig. 1). Evolutionarily highly conserved, there are three mammalian Cyclin Ds that have overlapping functions in a cell-lineage–specific manner. These allosterically bind to and regulate CDK4/6—two highly homologous serine/threonine kinases that have unique functions that are cell-type specific as well as being tightly developmentally and temporally regulated (13, 14). CDK6 is expressed at high levels in hematopoietic cells (14), and Cdk6 deficiency is characterized by subtle defects in the hematopoietic system, such as defects in thymocyte development (13, 15). CDK6 is also a more robust kinase (as compared with CDK4) with distinct affinities for specific modulatory client proteins, and together, these two Cyclin D–dependent CDKs allow for the creation of a network of finely tuned interactions to regulate cell-cycle progression (16). Activation of the Cyclin D–CDK4/6 complex promotes progression from the G1 phase into S-phase by phosphorylating several cellular targets, of which the Rb protein is key (17). Rb phosphorylation attenuates its inhibition of transcription by the E2F family of transcription factors, leading to the commitment of the cell to DNA replication and progression through the cell cycle (18).

Figure 1.

Regulation and functions of cyclin D–CDK4/6 kinases. The Cyclin D–CDK4/6 holoenzyme complex (green boxe) acts as an environmental sensor responding dynamically to mitogenic signals (e.g., estrogen, Ras–Raf–MEK, EGFR, and PI3K signaling pathways), cytokines, and other cues. Upon stimulation, D-type cyclins accumulate in early G1 phase through both transcriptional and posttranscriptional mechanisms. The activated cyclin D–CDK4/6 complexes initiate the phosphorylation of pRb releasing E2F transcription factors, thereby driving the expression of genes required for cellular commitment to enter S-phase and ultimately mitotic cell division. Growth-inhibitory signals antagonize G1–S progression by upregulating CDK inhibitors of the INK4 family (p16INK4A, p15INKB, p18INK4C, and p19INK4D). Amplification of the CCND1 gene encoding for Cyclin D or its overexpression, loss of stoichiometric inhibitors of cyclin D–CDK4/6 (members of the INK4A family), or the loss of Rb in tumors aberrantly activate the Cyclin D–CDK4/6 complex, thereby driving dysregulated cell-cycle progression. Depending on the cell type, and other mitogenic transforming signals, Rb-positive cells undergo either quiescence or senescence when treated with CDK4/6 inhibitors. In contrast, cells without functional Rb are refractory to arrest by chemical inhibitors of CDK4/6.

Figure 1.

Regulation and functions of cyclin D–CDK4/6 kinases. The Cyclin D–CDK4/6 holoenzyme complex (green boxe) acts as an environmental sensor responding dynamically to mitogenic signals (e.g., estrogen, Ras–Raf–MEK, EGFR, and PI3K signaling pathways), cytokines, and other cues. Upon stimulation, D-type cyclins accumulate in early G1 phase through both transcriptional and posttranscriptional mechanisms. The activated cyclin D–CDK4/6 complexes initiate the phosphorylation of pRb releasing E2F transcription factors, thereby driving the expression of genes required for cellular commitment to enter S-phase and ultimately mitotic cell division. Growth-inhibitory signals antagonize G1–S progression by upregulating CDK inhibitors of the INK4 family (p16INK4A, p15INKB, p18INK4C, and p19INK4D). Amplification of the CCND1 gene encoding for Cyclin D or its overexpression, loss of stoichiometric inhibitors of cyclin D–CDK4/6 (members of the INK4A family), or the loss of Rb in tumors aberrantly activate the Cyclin D–CDK4/6 complex, thereby driving dysregulated cell-cycle progression. Depending on the cell type, and other mitogenic transforming signals, Rb-positive cells undergo either quiescence or senescence when treated with CDK4/6 inhibitors. In contrast, cells without functional Rb are refractory to arrest by chemical inhibitors of CDK4/6.

Close modal

In cancer, multiple components of the CDK4/6–Rb axis are commonly dysregulated (19). The Cyclin D1 gene (CCND1) represents the second most frequently amplified locus among all human cancer types (20), with the highest prevalence in well-differentiated and dedifferentiated liposarcoma (21), glioblastomas (22), breast cancer (23–25), non–small cell lung cancer (NSCLC), endometrial cancers, and pancreatic cancers (23). Copy-number variation or overexpression in at least one component of the cyclin D–CDK4/6 pathway is also common and seen in up to 75% of melanoma (26) and gliomas (27). Loss of the negative regulators of the pathway either by genomic deletions, loss-of-function point mutations, or promoter methylation are also frequent, with p16INK4A most commonly lost in breast cancers (28) and head and neck cancers (29). However, other than in mantle cell lymphoma, which is defined by a translocation involving CCND1 resulting in cyclin D1 overexpression (30), mutations in genes encoding for the pathway are less common than copy-number changes (31).

Transcription of Cyclin D and its assembly with CDK4/6 is highly dependent on mitogenic signaling and is therefore an important mechanism of Cyclin D–CDK4/6 upregulation in cancer (31). ER signaling upregulates cyclin D1 levels as well as other signaling pathways, which largely culminate in the upregulation of CDK4/6 activity (6, 32, 33). Other upstream oncogenic signal transduction pathways including the PI3K–AKT–mTOR, wnt/β-catenin, MAPK, and NF-κB pathways also significantly lead to the induction of cell-cycle proteins, and D-type Cyclins in particular (6, 34).

Single-agent CDK4/6 inhibitors in vitro are fundamentally cytostatic, causing downregulation of E2F target genes, loss of proliferation markers, and cell-cycle arrest in G1 (35). It can therefore be hypothesized that cancer cells that are addicted to mitogenic signaling pathways and have functional Rb are strongly dependent on Cyclin D–CDK4/6 and thereby more vulnerable to CDK4/6 inhibition; this has been elegantly demonstrated in vitro (36). However, only modest clinical benefit has been reported in the various unselected early-phase trials of single-agent CDK4/6 monotherapy including in NSCLC, glioblastoma, melanoma, colorectal, and ovarian cancers as well as in mantle cell lymphoma and advanced liposarcoma. One exception to this is abemaciclib (which at clinically efficacious doses also inhibits CDK9) and has shown potentially useful single-agent activity in breast cancer leading to licensing as monotherapy in previously treated advanced breast cancer patients (37). Furthermore, the randomized JUNIPER study (NCT02152631) will evaluate its monotherapy efficacy in NSCLC.

Given the striking dependence of activated Cyclin D–CDK4/6 complex on mitogenic signals, there has been substantial work developing synergistic combinations of signal transduction inhibitors together with CDK4/6 inhibitors. The most advanced combinations are those in with endocrine therapies in estrogen-positive breast cancer, which led to the first FDA approvals. The pivotal phase II PALOMA-1 (NCT 00721409) study randomized postmenopausal women with advanced ER+/HER2 breast cancer to either letrozole, an aromatase inhibitor that prevents estrogen induction of Cyclin D, in combination with palbociclib, or letrozole alone (38). The significant improvement of progression-free survival in the combination arm (20.2 months vs. 10.2 months for letrozole alone; HR, 0.48; P < 0.001) led to early provisional FDA approval of palbociclib. Subsequent larger phase III trials have not only confirmed these results (7, 39, 40), but also extended the proof of principle of synergy in combining other CDK4/6 inhibitors with the selective ER degrader, fulvestrant (41, 42), or the antiestrogen, tamoxifen (40, 43).

The concept that combinatorial therapy with signal transduction inhibitors will amplify the effectiveness of a CDK4/6 inhibitor is now being extended to other mitogenic pathways and other tumor types. For example, preclinical evidence to suggest CDK4/6 inhibitors enhance the effect of RAS–RAF–MEK pathway inhibition in RAS-driven NSCLC (44) and RAS/RAF-resistant malignant melanoma (45) has led to early-phase trials of combination therapy in KRAS-mutant NSCLC (NCT02022982) and NRAS- and BRAF-mutant melanoma (46, 47). Hyperactivation of the PI3K pathway has also been shown to stabilize the Cyclin D protein and the Cyclin D–CDK4/6 complex (48), and CDK4/6 inhibitors have been shown preclinically to sensitize PIK3CA-mutant breast cancer to PI3K inhibitors (49). Triplet combinations of CDK4/6 inhibitors together with hormone therapies and PI3K inhibitors are also ongoing in breast cancer (NCT03006172; refs. 50–52).

One really interesting observation that may be pivotal is that in addition to blocking cell proliferation, CDK4/6 inhibitors can induce senescence—an irreversible distinct cellular state characterized by the absence of proliferation markers, expression of tumor-suppressor genes, senescence-associated beta-galactosidase activity, and the presence of senescence-associated heterochromatin foci in multiple Rb-proficient cell lines (53, 54). The decision whether to transition from quiescence into senescence is the subject of much ongoing work, and the outcome appears to cell-type specific with downregulation of MDM2, redistribution of the chromatin-remodeling enzyme ATRX, repressions of oncogenes as well as upregulation of proteasomal homeostasis necessary for the shift to senescence (55–57). Senescent cells secrete a collection of inflammatory cytokines, chemokines, and proteinases, collectively referred to as the senescence-associated secretory phenotype (SASP) which recruits and activates distinct cells from the innate and adaptive immune system, such as macrophages and natural killer cells as well as T cells (58, 59). The SASP is one of the most profound features of senescence with the triggering of immune cell recruitment into the tumor (60, 61), although on the other hand, there are concerns that the inflammatory environment chronically stimulated by SASP could be protumorigenic (58).

This raises several obvious questions about the clinical effect of these inhibitors on host immune cells, and whether this would hinder, or could be leveraged for combination therapies. In the following sections, we review the role of Cyclin D–CDK4/6 in immune cell expansion and differentiation, together with the emerging learnings from the translations studies of CDK4/6 inhibitors.

Mouse models provided the first clues to the physiologic roles of Cdk4 and Cdk6 in vivo, particularly with respect to the immune cell types that critically depend upon the Cyclin D–CDK4/6 pathway during development. Double-mutant mice lacking both Cdk4 and Cdk6 (Cdk4/6−/− mice) display late embryonic lethality accompanied by a defect in fetal hematopoiesis very similar to the phenotype observed in the triple D1/2/3-cyclins−/− mice, including multilineage hematopoietic abnormalities (13, 62).

Myeloid lineage

Myeloid cell development in preclinical models is entirely reliant on Cyclin D2– and Cyclin D3–driven CDK6 (13, 63), and all myeloid progenitor cells' populations were also severely reduced in Cdk4/6−/− double-mutant mice (13).

Not unsurprisingly, neutropenia has been the dose-limiting toxicity of both palbociclib and ribociclib, necessitating intermittent dosing schedules (64, 65). Abemaciclib, being a more potent inhibitor of CDK4 (as well as inhibiting CDK9 at clinically efficacious doses), seems distinct and causes much lower rates of neutropenia (Table 1) and can be dosed on a continuous schedule (66). An ongoing study of palbociclib is investigating whether a continuous dosing schedule (at 100 mg/day) is as effective and tolerable as the approved intermittent dosing schedule (NCT02630693). Clinical data on the changes in other myeloid cell subpopulations are however scarce at this time, chiefly as multiparameter data analysis of circulating immune cells or immune cells within the tumor microenvironment was not collected in the initial trials.

Table 1.

Myelosuppression seen in the initial early-phase trials of single-agent CDK4/6 inhibitors

PalbociclibRibociclibAbemaciclib
IC50 CDK4 9–11 nmol/L 10 nmol/L 2 nmol/L 
 CDK6 15 nmol/L 39 nmol/L 5 nmol/L 
Dosing  125 mg daily 600 mg daily 200 mg daily 
  3 weeks on, 1 week off 3 weeks on, 1 week off continuously 
Effects on immune cells All grades G3/4 All grades G3/4 All grades G3/4 
 Neutropenia 95% 54%a 46% 29%a 23% 10% 
 Leukopenia 68% 23% 48% 21% 25% 10% 
Trial ID NCT00141297 NCT01237236 NCT01394016 
PalbociclibRibociclibAbemaciclib
IC50 CDK4 9–11 nmol/L 10 nmol/L 2 nmol/L 
 CDK6 15 nmol/L 39 nmol/L 5 nmol/L 
Dosing  125 mg daily 600 mg daily 200 mg daily 
  3 weeks on, 1 week off 3 weeks on, 1 week off continuously 
Effects on immune cells All grades G3/4 All grades G3/4 All grades G3/4 
 Neutropenia 95% 54%a 46% 29%a 23% 10% 
 Leukopenia 68% 23% 48% 21% 25% 10% 
Trial ID NCT00141297 NCT01237236 NCT01394016 

aIndicates dose-limiting toxicity.

Lymphoid lineage

Following antigen exposure, quiescent lymphocytes require intense, prolonged, and repeated proliferation to establish a rapid immune response and generate immunologic memory. Upon stimulation, T cells exit G0 via an NF-κB–dependent pathway (67). Cyclin D is expressed in cells during G1, with significant upregulation of Cyclin D2 and Cyclin D3 and CDK6 during early and late G1 (refs. 68, 69; Fig. 2). Deletion of specific cyclins and CDKs in mice has identified CDK6 and Cyclin D3 to be the key players in hematopoietic stem cells' regulation, their proliferation, and subsequent commitment to the T-cell lineage (13, 15, 70–72). Loss of CDK6 leads to delayed G1 progression in lymphocytes, but critically, once a cell is committed to proliferation, other Cyclin-CDKs, particularly Cyclin E and CDK2, appear to compensate. As such, despite CDK6-mutant mice having lower numbers of thymocytes early on in development, they have normal/higher than normal levels of CD4+/CD8+ cells later in development (63). This is very consistent with the modest reductions in total lymphocyte numbers seen clinically with the unique differential potency of inhibition of abemaciclib likely to be responsible for the lack of appreciable leukopenia reported clinically for abemaciclib (Table 1; ref. 66).

Figure 2.

Cyclin D–CDK4/6 in T-cell activation, expansion, and differentiation. The figure shows relative levels of CDK4/6 and D-type cyclins at the various stages of the cell cycle as naïve T cells respond to antigen stimulation, enter the cell cycle, and undergo clonal expansion followed by maturation and subsequent differentiation. Lilac boxes highlight cells with high levels of CDK4/6 and D-type cyclins, whereas light green indicates cells with low levels. Stimulation of the T-cell receptor (TCR) together with costimulatory signals (e.g., CD28) leads to the induction of a number of cell-cycle activators, including CDK4/6 and D-type cyclins, which set off a signaling cascade permitting progression through the G1 phase of the cell cycle. Subsequent progression through S-phase is accompanied by downregulation of both CDK4/6 and D-type cyclins in an oscillating manner as cells undergo repeated cycles of cell division. As cells differentiate, Cyclin D remains low in the majority of T cells, with the exception of regulatory T cells (Treg), which retain high expression of both Cyclin D and CDK4/6 (74, 75). As such, although therapeutic targeting of CDK4/6 can theoretically slow T-cell proliferation, in vivo it has a preferential effect of promoting cell differentiation while specifically depleting Tregs (9, 10).

Figure 2.

Cyclin D–CDK4/6 in T-cell activation, expansion, and differentiation. The figure shows relative levels of CDK4/6 and D-type cyclins at the various stages of the cell cycle as naïve T cells respond to antigen stimulation, enter the cell cycle, and undergo clonal expansion followed by maturation and subsequent differentiation. Lilac boxes highlight cells with high levels of CDK4/6 and D-type cyclins, whereas light green indicates cells with low levels. Stimulation of the T-cell receptor (TCR) together with costimulatory signals (e.g., CD28) leads to the induction of a number of cell-cycle activators, including CDK4/6 and D-type cyclins, which set off a signaling cascade permitting progression through the G1 phase of the cell cycle. Subsequent progression through S-phase is accompanied by downregulation of both CDK4/6 and D-type cyclins in an oscillating manner as cells undergo repeated cycles of cell division. As cells differentiate, Cyclin D remains low in the majority of T cells, with the exception of regulatory T cells (Treg), which retain high expression of both Cyclin D and CDK4/6 (74, 75). As such, although therapeutic targeting of CDK4/6 can theoretically slow T-cell proliferation, in vivo it has a preferential effect of promoting cell differentiation while specifically depleting Tregs (9, 10).

Close modal

Commitment to specific cell fates and differentiation with transcriptional activation of specific gene expression programs are predominantly directed by CDK2 (73), and as such in vitro differentiation of naïve CD4+ and CD8+ T cells is not affected by CDK4/6 inhibitors (10). Specific T-cell subpopulations however may be much more selectively suppressed by CDK4/6 inhibition as suggested by flow cytometry analysis of circulating immune cells in preclinical models. In tumor-free mice, both abemaciclib and palbociclib significantly reduced FOXP3+ regulatory T cells (10) without affecting other cell subtypes, and this may relate to the higher levels of both Cyclin D and CDK4/6 (74, 75) or Rb1 present in these cells (76). Abemaciclib may also have additional epigenetic effects by selectively inhibiting the enzyme DNMT1 in regulatory T cells, resulting in overexpression of the negative regulator p21 (10). The effects of CDK4/6 inhibition on tumor-infiltrating lymphocytes may be more complex with both palbociclib and trilaciclib causing increased infiltration of T cells into lung tumors in an immunocompetent genetically engineered mouse model (GEMM). In this model, absolute numbers of CD4+ and CD8+ cells were unchanged, but proliferation of tumor-infiltrating FOXP3+ regulatory cells as well as immunosuppressive myeloid cells were significantly reduced, resulting in an increased percentage of effector cells within the tumor microenvironment (9).

In summary, the data thus far reveal that although therapeutic targeting of CDK4/6 can theoretically slow T-cell proliferation, in vivo it has a preferential effect of promoting cell differentiation while specifically depleting regulatory T cells (refs. 9, 10; Fig. 2).

Enhancing immune cell infiltration into tumor

Cell-cycle arrest and the induction of senescence lead to the activation of the SASP in a subset of cancer cells which can induce the recruitment of innate immune cells including macrophages, neutrophils, and natural killer cells into the tumor microenvironment where they are provoked into coordinately attacking tumors through both phagocytosis and direct cytotoxic killing (61, 66). The challenge remains in understanding how tumor cells are directed toward either reversible quiescence or a more stable senescence, as preliminary work in a small subset of abemaciclib-sensitive breast cancer cell lines suggests that genes encoding for the canonical SASP cytokines were not shown to be upregulated in the cell lines tested (11).

Enhancing antigen presentation

The initial studies suggesting links between CDK4/6 inhibition and the immune system came from an Rb-proficient transgenic mouse model of breast cancer (10). Treatment with abemaciclib caused not only cell stasis but also a significant decrease in tumor volume and reduced cell proliferation (10). Gene expression analysis has shown that in addition to downregulating genes related to cell cycle, mitotic, and E2F targets, abemaciclib also significantly upregulates genes responsible for antigen processing and presentation including MHC class I molecules (10). This was confirmed in vitro as well as in patient-derived xenografts. Strikingly, tumor cells treated with CDK4/6 inhibitors show a marked reduction of DNMT1, which decreases DNA methylation of genes that regulate immune function as well as endogenous retroviral genes. Expression of double-stranded RNA triggers “viral mimicry” stimulating the production of an IFN response (ref. 10; Fig. 3).

Figure 3.

To cycle or fight. Tumors with hyperactivation of the Cyclin D–CDK4/6 axis aberrantly progress through the cell cycle and effectively hide from the host immune system through multiple mechanisms including downregulation of MHC class I molecules. Treatment with CDK4/6 inhibitors both arrests cell cycle and promotes a “fight” mode, promoting antitumor immunity by stimulating antigen presentation through (i) the upregulation of MHC class I expression within tumors and (ii) the increase in proinflammatory cytokine secretion (e.g., IFNγ) either via inducing the SASP or other mechanism resulting in activation of dendritic cells and macrophages. CDK4/6 inhibitors also selectively deplete immunosuppressive regulatory T cells and change the cytokine milieu within the tumor microenvironment, thereby increasing effector T-cell infiltration into the tumor (9–11).

Figure 3.

To cycle or fight. Tumors with hyperactivation of the Cyclin D–CDK4/6 axis aberrantly progress through the cell cycle and effectively hide from the host immune system through multiple mechanisms including downregulation of MHC class I molecules. Treatment with CDK4/6 inhibitors both arrests cell cycle and promotes a “fight” mode, promoting antitumor immunity by stimulating antigen presentation through (i) the upregulation of MHC class I expression within tumors and (ii) the increase in proinflammatory cytokine secretion (e.g., IFNγ) either via inducing the SASP or other mechanism resulting in activation of dendritic cells and macrophages. CDK4/6 inhibitors also selectively deplete immunosuppressive regulatory T cells and change the cytokine milieu within the tumor microenvironment, thereby increasing effector T-cell infiltration into the tumor (9–11).

Close modal

Further support of this model comes from data from The Cancer Genome Atlas showing that breast cancers harboring CCND1 amplification (therefore enhanced CDK4/6 activity) display significantly lower expression of MHC class I molecules HLA-A, HLA-B, and HLA-C than nonamplified tumors (10).

Effects on cytokine milieu in the tumor microenvironment

In addition, Deng and colleagues used a small-molecule screen designed to identify targets that enhanced T-cell activity in the setting of PD-1 engagement and found that CDK4/6 inhibitors potently upregulated IL2 (9). Using siRNAs, they confirmed that CDK6, not CDK4, was responsible for the enhanced IL2 secretion supporting a predominant role for CDK6 in immune cell function. Careful dissection of the mechanistic basis of this effect found that CDK6 was an upstream regulator of Nuclear Factor of Activated T cells (NFAT) proteins which are critical in regulation of T-cell activation and function. CDK4/6 inhibition resulted in increased nuclear levels of NFAT and increased transcriptional activity ultimately resulting in a change in cytokine milieu within the tumor microenvironment and increased effector T-cell activity (9, 11). Levels of IL6, IL10, and IL23, three cytokines produced by immunosuppressive myeloid cells, were significantly reduced, whereas an increase of the Th1 chemokines CXCL9 and CXCL10 which govern the trafficking of effector cells to tumor sites was seen (9, 77).

Effects on PD-L1 and other coinhibitor molecule expression

PD-L1 protein abundance fluctuates during cell-cycle progression in multiple human cancer cell lines, peaking in M and early G1, with a sharp reduction in latter stages of the cell cycle. This is tightly regulated by Cyclin D–CDK4-mediated phosphorylation of the speckle-type POZ protein, a core component of the Cullin3-SPOP E3 ligase responsible for the proteasomal degradation of PD-L1 (78). Inhibition of CDK4/6 in this single article increases PD-L1 expression, but only in SPOP-proficient cancer cells. The story is far from complete though, as effects of CDK4/6 inhibition on the expression of other coinhibitory molecules, particularly on immune cells, are likely to be complex. For example in the GEMM mouse model used by Deng and colleagues, levels of the coinhibitory molecular PD-1 and CTLA-4 were reduced in both CD4+ and CD8+ T cells after CDK4/6 inhibition (9).

Taken together, these studies illustrate the complex connection between immunity and cell-cycle regulation and constitute an exciting new area of research, which is likely to lead to significant anticancer therapeutic opportunities, and pharmacodynamic and translational outputs from the ongoing clinical trials are eagerly awaited. Combining CDK4/6 inhibitors together with immune checkpoint inhibitors enhances tumor regression in a number of immunocompetent preclinical mice models (9, 78). These effects seem to be at least in part to be tumor-intrinsic, as most potent upregulation of the antigen-processing machinery at a gene expression level occurred in CDK4/6-sensitive cell lines (10). In addition, there are hints that as cancers evolve and undergo immune-editing, thus becoming more immune-refractory, they may be increasingly dependent on Cyclin D–CDK4/6. Oh and colleagues studied a highly immune-refractory cancer and found that synaptonemal complex protein 3 (SCP3) is overexpressed in immune-edited cancer cells and upregulates the pluripotency transcriptional factor NANOG by hyperactivation of the Cyclin D–CDK4/6 axis. In this model, the combination of palbociclib together with adoptive cytotoxic cell transfer showed considerable therapeutic efficacy, suggesting a niche role for CDK4/6 inhibitors in immunotherapy combinations in the resistant/refractory setting (79).

The ongoing clinical trials testing combinations of CDK4/6 inhibitors with immune checkpoint inhibitors are listed in Table 2, but a few specific challenges in combining these are worth exploring. Understanding the temporal kinetics of pharmacodynamics effects of CDK4/6 inhibitors on the tumor microenvironment and the immune system would be key to optimizing sequencing of any combinations. Schaer and colleagues looked at the differences in antitumor responses when anti–PD-L1 therapy was given either concurrently, sequentially (after completion of CDK4/6 inhibitor), or in a phased (initiated after 1 week of CDK4/6 inhibitor) manner with abemaciclib (11). Surprisingly, concurrent administration of abemaciclib with immune checkpoint inhibitors showed no significant difference in the antitumor response compared with monotherapy. Sequential treatment was additive, but the phased regime was significantly synergistic, with complete responses seen in 2 of 10 mice (11) highlighting the importance of understanding the biology to direct scheduling of combinations. Furthermore, they analyzed the effect of transient versus continuous exposure to abemaciclib on primary T cells during T-cell receptor (TCR)–mediated expansion and found that the greatest effect in upregulating genes indicated of T-cell activation was seen with the continuous exposure. This may be pertinent when other CDK4/6 inhibitors that are given intermittently are considered for combinations. Moving forward, it will be absolutely imperative that proof-of-mechanism pharmacodynamics studies utilizing paired tumor biopsies as well as in-depth analyses of host immune responses be incorporated into these trials to maximize patient benefit.

Table 2.

Ongoing clinical trials combining CDK4/6 inhibitors with immune checkpoint inhibitors

Trial name/IDPhasePatient populationCombination agentsPrimary objective
Ribociclib + PDR001 in Breast Cancer and Ovarian Cancer/NCT03294694 HR+HER2 breast cancer Ribociclib MTD/RP2D 
  Epithelial ovarian cancer PDR001–PD-1 inhibitor  
   Fulvestrant  
PACT/NCT02791334 Solid tumor LY3300054—PD-1 Inhibitor DLT 
  Microsatellite instability-high (MSI-H) solid tumors Ramucirumab  
  Cutaneous melanoma Abemaciclib  
  Pancreatic cancer Merestinib  
  Breast cancer (HR+HER2  
A Study of Abemaciclib (LY2835219) in Participants With Non-Small Cell Lung Cancer or Breast Cancer/NCT02779751 NSCLC Abemaciclib AEs 
  HR+HER2 breast cancer Pembrolizumab  
PAVEMENT—A phase Ib study of palbociclib and avelumab in metastatic AR+ triple-negative breast cancer (NCT pending) Ib Androgen-receptor–positive triple-negative breast cancer Palbociclib R2PD 
   Avelumab  
Trial name/IDPhasePatient populationCombination agentsPrimary objective
Ribociclib + PDR001 in Breast Cancer and Ovarian Cancer/NCT03294694 HR+HER2 breast cancer Ribociclib MTD/RP2D 
  Epithelial ovarian cancer PDR001–PD-1 inhibitor  
   Fulvestrant  
PACT/NCT02791334 Solid tumor LY3300054—PD-1 Inhibitor DLT 
  Microsatellite instability-high (MSI-H) solid tumors Ramucirumab  
  Cutaneous melanoma Abemaciclib  
  Pancreatic cancer Merestinib  
  Breast cancer (HR+HER2  
A Study of Abemaciclib (LY2835219) in Participants With Non-Small Cell Lung Cancer or Breast Cancer/NCT02779751 NSCLC Abemaciclib AEs 
  HR+HER2 breast cancer Pembrolizumab  
PAVEMENT—A phase Ib study of palbociclib and avelumab in metastatic AR+ triple-negative breast cancer (NCT pending) Ib Androgen-receptor–positive triple-negative breast cancer Palbociclib R2PD 
   Avelumab  

Abbreviations: AEs, adverse events; DLT, dose-limiting toxicity; RP2D, recommended phase II dose.

Biomarker-driven patient selection is also likely to direct these combinations to the patients who are most likely to derive benefit; for example, if loss of MHC I expression or markers of T-cell exhaustion are seen, combinations of phased in pretreatment with CDK4/6 inhibitors given with immunotherapy may be therapeutically beneficial. Paraphrasing Shakespeare, much more remains to be learned about how a cell decides whether to cycle or to fight, and future work will reveal if the promise of combining CDK4/6 inhibitors with immunotherapy will be realized and validated.

A.F.C. Okines reports receiving commercial research grants from Pfizer and speakers bureau honoraria from Roche. J.S. Lopez is a consultant/advisory board member for Basilea, Eisai, Genmab, Merck, and Novartis. No potential conflicts of interest were disclosed by the other authors.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Graña
X
,
Reddy
EP
. 
Cell cycle control in mammalian cells: role of cyclins, cyclin dependent kinases (CDKs), growth suppressor genes and cyclin-dependent kinase inhibitors (CKIs)
.
Oncogene
1995
;
11
:
211
20
.
2.
Gonzalez
MA
,
Tachibana
KE
,
Laskey
RA
,
Coleman
N
. 
Control of DNA replication and its potential clinical exploitation
.
Nat Rev Cancer
2005
;
5
:
135
41
.
3.
Hanahan
D
,
Weinberg
RA
. 
Hallmarks of cancer: the next generation
.
Cell
2011
;
144
:
646
74
.
4.
Sherr
CJ
,
Beach
D
,
Shapiro
GI
. 
Targeting CDK4 and CDK6: from discovery to therapy
.
Cancer Discov
2016
;
6
:
353
67
.
5.
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
:
425
39
.
6.
Finn
RS
,
Aleshin
A
,
Slamon
DJ
. 
Targeting the cyclin-dependent kinases (CDK) 4/6 in estrogen receptor-positive breast cancers
.
Breast Cancer Res
2016
;
18
:
17
.
7.
Hortobagyi
GN
,
Stemmer
SM
,
Burris
HA
,
Yap
YS
,
Sonke
GS
,
Paluch-Shimon
S
, et al
Ribociclib as first-line therapy for HR-positive, advanced breast cancer
.
N Engl J Med
2016
;
375
:
1738
48
.
8.
Finn
RS
,
Dering
J
,
Conklin
D
,
Kalous
O
,
Cohen
DJ
,
Desai
AJ
, et al
PD 0332991, a selective cyclin D kinase 4/6 inhibitor, preferentially inhibits proliferation of luminal estrogen receptor-positive human breast cancer cell lines in vitro
.
Breast Cancer Res
2009
;
11
:
R77
.
9.
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
:
216
33
.
10.
Goel
S
,
DeCristo
MJ
,
Watt
AC
,
BrinJones
H
,
Sceneay
J
,
Li
BB
, et al
CDK4/6 inhibition triggers anti-tumour immunity
.
Nature
2017
;
548
:
471
.
11.
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
:
2978
94
.
12.
Otto
T
,
Sicinski
P
. 
Cell cycle proteins as promising targets in cancer therapy
.
Nat Rev Cancer
2017
;
17
:
93
.
13.
Malumbres
M
,
Sotillo
Ro
,
Santamaŕa
D
,
Galán
J
,
Cerezo
A
,
Ortega
S
, et al
Mammalian cells cycle without the D-type cyclin-dependent kinases Cdk4 and Cdk6
.
Cell
2004
;
118
:
493
504
.
14.
Scheicher
R
,
Hoelbl-Kovacic
A
,
Bellutti
F
,
Tigan
A-S
,
Prchal-Murphy
M
,
Heller
G
, et al
CDK6 as a key regulator of hematopoietic and leukemic stem cell activation
.
Blood
2015
;
125
:
90
101
.
15.
Hu
MG
,
Deshpande
A
,
Enos
M
,
Mao
D
,
Hinds
EA
,
Hu
G-f
, et al
A requirement for cyclin-dependent kinase 6 in thymocyte development and tumorigenesis
.
Cancer Res
2009
;
69
:
810
8
.
16.
Hallett
ST
,
Pastok
MW
,
Morgan
RML
,
Wittner
A
,
Blundell
K
,
Felletar
I
, et al
Differential regulation of G1 CDK complexes by the Hsp90-Cdc37 chaperone system
.
Cell Rep
2017
;
21
:
1386
98
.
17.
Harbour
JW
,
Luo
RX
,
Santi
AD
,
Postigo
AA
,
Dean
DC
. 
Cdk phosphorylation triggers sequential intramolecular interactions that progressively block Rb functions as cells move through G1
.
Cell
1999
;
98
:
859
69
.
18.
Harbour
JW
,
Dean
DC
. 
The Rb/E2F pathway: expanding roles and emerging paradigms
.
Genes Dev
2000
;
14
:
2393
409
.
19.
Nevins
JR
. 
The Rb/E2F pathway and cancer
.
Hum Mol Genet
2001
;
10
:
699
703
.
20.
Beroukhim
R
,
Mermel
CH
,
Porter
D
,
Wei
G
,
Raychaudhuri
S
,
Donovan
J
, et al
The landscape of somatic copy-number alteration across human cancers
.
Nature
2010
;
463
:
899
.
21.
Crago
AM
,
Singer
S
. 
Clinical and molecular approaches to well differentiated and dedifferentiated liposarcoma
.
Curr Opin Oncol
2011
;
23
:
373
8
.
22.
Schmidt
EE
,
Ichimura
K
,
Reifenberger
G
,
Collins
VP
. 
CDKN2 (p16/MTS1) gene deletion or CDK4 amplification occurs in the majority of glioblastomas
.
Cancer Res
1994
;
54
:
6321
4
.
23.
Musgrove
EA
,
Caldon
CE
,
Barraclough
J
,
Stone
A
,
Sutherland
RL
. 
Cyclin D as a therapeutic target in cancer
.
Nat Rev Cancer
2011
;
11
:
558
72
.
24.
Cancer Genome Atlas N
. 
Comprehensive molecular portraits of human breast tumours
.
Nature
2012
;
490
:
61
70
.
25.
An
HX
,
Beckmann
MW
,
Reifenberger
G
,
Bender
HG
,
Niederacher
D
. 
Gene amplification and overexpression of CDK4 in sporadic breast carcinomas is associated with high tumor cell proliferation
.
Am J Pathol
1999
;
154
:
113
8
.
26.
Young
RJ
,
Waldeck
K
,
Martin
C
,
Foo
JH
,
Cameron
DP
,
Kirby
L
, et al
Loss of CDKN2A expression is a frequent event in primary invasive melanoma and correlates with sensitivity to the CDK4/6 inhibitor PD0332991 in melanoma cell lines
.
Pigment Cell Melanoma Res
2014
;
27
:
590
600
.
27.
Brennan
CW
,
Verhaak
RG
,
McKenna
A
,
Campos
B
,
Noushmehr
H
,
Salama
SR
, et al
The somatic genomic landscape of glioblastoma
.
Cell
2013
;
155
:
462
77
.
28.
Geradts
J
,
Wilson
PA
. 
High frequency of aberrant p16(INK4A) expression in human breast cancer
.
Am J Pathol
1996
;
149
:
15
20
.
29.
Reed
AL
,
Califano
J
,
Cairns
P
,
Westra
WH
,
Jones
RM
,
Koch
W
, et al
High frequency of p16 (CDKN2/MTS-1/INK4A) inactivation in head and neck squamous cell carcinoma
.
Cancer Res
1996
;
56
:
3630
3
.
30.
Rosenwald
A
,
Wright
G
,
Wiestner
A
,
Chan
WC
,
Connors
JM
,
Campo
E
, et al
The proliferation gene expression signature is a quantitative integrator of oncogenic events that predicts survival in mantle cell lymphoma
.
Cancer Cell
2003
;
3
:
185
97
.
31.
Hunter
T
,
Pines
J
. 
Cyclins and cancer II: cyclin D and CDK inhibitors come of age
.
Cell
1994
;
79
:
573
82
.
32.
Zwijsen
RM
,
Wientjens
E
,
Klompmaker
R
,
van der Sman
J
,
Bernards
R
,
Michalides
RJ
. 
CDK-independent activation of estrogen receptor by cyclin D1
.
Cell
1997
;
88
:
405
15
.
33.
Musgrove
EA
,
Sutherland
RL
. 
Cell cycle control by steroid hormones
.
Semin Cancer Biol
1994
;
5
:
381
9
.
34.
Ewen
ME
. 
Relationship between Ras pathways and cell cycle control
.
Prog Cell Cycle Res
2000
;
4
:
1
17
.
35.
Fry
DW
,
Harvey
PJ
,
Keller
PR
,
Elliott
WL
,
Meade
M
,
Trachet
E
, et al
Specific inhibition of cyclin-dependent kinase 4/6 by PD 0332991 and associated antitumor activity in human tumor xenografts
.
Mol Cancer Ther
2004
;
3
:
1427
38
.
36.
Gong
X
,
Litchfield
LM
,
Webster
Y
,
Chio
L-C
,
Wong
SS
,
Stewart
TR
, et al
Genomic aberrations that activate D-type cyclins are associated with enhanced sensitivity to the CDK4 and CDK6 inhibitor abemaciclib
.
Cancer Cell
2017
;
32
:
761
76
.
e6
.
37.
Dickler
MN
,
Tolaney
SM
,
Rugo
HS
,
Cortés
J
,
Diéras
V
,
Patt
D
, et al
MONARCH 1, a phase II study of abemaciclib, a CDK4 and CDK6 inhibitor, as a single agent, in patients with refractory HR+/HER2− metastatic breast cancer
.
Clin Cancer Res
2017
;
23
:
5218
24
.
38.
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
:
25
35
.
39.
Finn
RS
,
Martin
M
,
Rugo
HS
,
Jones
S
,
Im
S-A
,
Gelmon
K
, et al
Palbociclib and letrozole in advanced breast cancer
.
N Engl J Med
2016
;
375
:
1925
36
.
40.
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
:
3638
46
.
41.
Turner
NC
,
Ro
J
,
André
F
,
Loi
S
,
Verma
S
,
Iwata
H
, et al
Palbociclib in hormone-receptor–positive advanced breast cancer
.
N Engl J Med
2015
;
373
:
209
19
.
42.
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
:
2875
84
.
43.
Tripathy
D
,
Sohn
J
,
Im
S-A
,
Colleoni
M
,
Franke
F
,
Bardia
A
, et al
.
First-line ribociclib vs. placebo with goserelin and tamoxifen or a non-steroidal aromatase inhibitor in premenopausal women with hormone receptor-positive, HER2-negative advanced breast cancer: results from the randomized phase III MONALEESA-7 trial [abstract]
. In: 
Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5–9
;
San Antonio, TX
.
Abstract nr GS2-05
.
44.
Zhou
J
,
Zhang
S
,
Chen
X
,
Zheng
X
,
Yao
Y
,
Lu
G
, et al
Palbociclib, a selective CDK4/6 inhibitor, enhances the effect of selumetinib in RAS-driven non-small cell lung cancer
.
Cancer Lett
2017
;
408
:
130
7
.
45.
Yadav
V
,
Burke
TF
,
Huber
L
,
Van Horn
RD
,
Zhang
Y
,
Buchanan
SG
, et al
The CDK4/6 inhibitor LY2835219 overcomes vemurafenib resistance resulting from MAPK reactivation and cyclin D1 upregulation
.
Mol Cancer Ther
2014
;
13
:
2253
63
.
46.
Schuler
MH
,
Ascierto
PA
,
De Vos
FYFL
,
Postow
MA
,
Van Herpen
CML
,
Carlino
MS
, et al
Phase 1b/2 trial of ribociclib+binimetinib in metastatic NRAS-mutant melanoma: safety, efficacy, and recommended phase 2 dose (RP2D)
.
J Clin Oncol
35s
, 
2017
(
suppl; abstr
9519
).
47.
Taylor
M
,
Sosman
J
,
Gonzalez
R
,
Carlino
M
,
Kittaneh
M
,
Lolkema
M
, et al
Phase Ib/II study of LEE011 (CDK4/6 inhibitor) and LGX818 (BRAF inhibitor) in BRAF-mutant melanoma
.
Ann Oncol
2014
:
25
(
suppl_4
):
iv374
iv393
.
48.
Diehl
JA
,
Cheng
M
,
Roussel
MF
,
Sherr
CJ
. 
Glycogen synthase kinase-3beta regulates cyclin D1 proteolysis and subcellular localization
.
Genes Dev
1998
;
12
:
3499
511
.
49.
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
:
136
49
.
50.
Lim
JSJ
,
Asghar
US
,
Diamantis
N
,
Ward
SE
,
Parmar
M
,
Purchase
B
, et al
,
editors
. 
A phase I trial of selective PI3K inhibitor taselisib (tas) plus palbociclib (palb) with and without endocrine therapy incorporating pharmacodynamic (PD) studies in patients (pts) with advanced cancers
.
J Clin Oncol
; 
2017
;
35
:
2573
.
51.
Bardia
A
,
Modi
S
,
Oliveira
M
,
Campone
M
,
Ma
B
,
Dirix
L
, et al
Triplet therapy with ribociclib, everolimus, and exemestane in women with HR+/HER2–advanced breast cancer [abstract]
. 
In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8–12; San Antonio, TX
.
Philadelphia (PA)
:
AACR
;
Cancer Res 2016;76(4 Suppl):Abstract nr P6-13-01.
52.
Juric
D
,
Ismail-Khan
R
,
Campone
M
,
García-Estévez
L
,
Becerra
C
,
De Boer
R
, et al
Phase Ib/II study of ribociclib and alpelisib and letrozole in ER+, HER2–breast cancer: safety, preliminary efficacy and molecular analysis [abstract]
. 
In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8–12; San Antonio, TX
.
Philadelphia (PA)
:
AACR
;
Cancer Res 2016;76(4 Suppl):Abstract nr P3-14-01
.
53.
Ewald
JA
,
Desotelle
JA
,
Wilding
G
,
Jarrard
DF
. 
Therapy-induced senescence in cancer
.
J Natl Cancer Inst
2010
;
102
:
1536
46
.
54.
Munoz-Espin
D
,
Serrano
M
. 
Cellular senescence: from physiology to pathology
.
Nat Rev Mol Cell Biol
2014
;
15
:
482
96
.
55.
Kovatcheva
M
,
Liao
W
,
Klein
ME
,
Robine
N
,
Geiger
H
,
Crago
AM
, et al
ATRX is a regulator of therapy induced senescence in human cells
.
Nat Commun
2017
;
8
:
386
.
56.
Kovatcheva
M
,
Liu
DD
,
Dickson
MA
,
Klein
ME
,
O'Connor
R
,
Wilder
FO
, et al
MDM2 turnover and expression of ATRX determine the choice between quiescence and senescence in response to CDK4 inhibition
.
Oncotarget
2015
;
6
:
8226
43
.
57.
Miettinen
TP
,
Peltier
J
,
Hartlova
A
,
Gierlinski
M
,
Jansen
VM
,
Trost
M
, et al
Thermal proteome profiling of breast cancer cells reveals proteasomal activation by CDK4/6 inhibitor palbociclib
.
EMBO J
2018
;
37
.
58.
Coppe
JP
,
Desprez
PY
,
Krtolica
A
,
Campisi
J
. 
The senescence-associated secretory phenotype: the dark side of tumor suppression
.
Annu Rev Pathol
2010
;
5
:
99
118
.
59.
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
:
9
20
.
60.
Burton
DGA
,
Stolzing
A
. 
Cellular senescence: Immunosurveillance and future immunotherapy
.
Ageing Res Rev
2018
;
43
:
17
25
.
61.
Xue
W
,
Zender
L
,
Miething
C
,
Dickins
RA
,
Hernando
E
,
Krizhanovsky
V
, et al
Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas
.
Nature
2007
;
445
:
656
60
.
62.
Kozar
K
,
Ciemerych
MA
,
Rebel
VI
,
Shigematsu
H
,
Zagozdzon
A
,
Sicinska
E
, et al
Mouse development and cell proliferation in the absence of D-cyclins
.
Cell
2004
;
118
:
477
91
.
63.
Sicinska
E
,
Lee
Y-M
,
Gits
J
,
Shigematsu
H
,
Yu
Q
,
Rebel
VI
, et al
Essential role for cyclin D3 in granulocyte colony-stimulating factor-driven expansion of neutrophil granulocytes
.
Mol Cell Biol
2006
;
26
:
8052
60
.
64.
DeMichele
A
,
Clark
AS
,
Tan
KS
,
Heitjan
DF
,
Gramlich
K
,
Gallagher
M
, et al
CDK 4/6 inhibitor palbociclib (PD0332991) in Rb+ advanced breast cancer: phase II activity, safety, and predictive biomarker assessment
.
Clin Cancer Res
2015
;
21
:
995
1001
.
65.
Infante
JR
,
Cassier
PA
,
Gerecitano
JF
,
Witteveen
PO
,
Chugh
R
,
Ribrag
V
, et al
A phase I study of the cyclin-dependent kinase 4/6 inhibitor ribociclib (LEE011) in patients with advanced solid tumors and lymphomas
.
Clin Cancer Res
2016
;
22
:
5696
705
.
66.
Patnaik
A
,
Rosen
LS
,
Tolaney
SM
,
Tolcher
AW
,
Goldman
JW
,
Gandhi
L
, 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
53
.
67.
Doi
TS
,
Takahashi
T
,
Taguchi
O
,
Azuma
T
,
Obata
Y
. 
NF-κB RelA-deficient lymphocytes: normal development of T cells and B cells, impaired production of IgA and IgG1 and reduced proliferative responses
.
J Exp Med
1997
;
185
:
953
62
.
68.
Ajchenbaum
F
,
Ando
K
,
DeCaprio
J
,
Griffin
J
. 
Independent regulation of human D-type cyclin gene expression during G1 phase in primary human T lymphocytes
.
J Biol Chem
1993
;
268
:
4113
9
.
69.
Meyerson
M
,
Harlow
E
. 
Identification of G1 kinase activity for cdk6, a novel cyclin D partner
.
Mol Cell Biol
1994
;
14
:
2077
86
.
70.
Sicinska
E
,
Aifantis
I
,
Le Cam
L
,
Swat
W
,
Borowski
C
,
Yu
Q
, et al
Requirement for cyclin D3 in lymphocyte development and T cell leukemias
.
Cancer Cell
2003
;
4
:
451
61
.
71.
Hu
MG
,
Deshpande
A
,
Schlichting
N
,
Hinds
EA
,
Mao
C
,
Dose
M
, et al
CDK6 kinase activity is required for thymocyte development
.
Blood
2011
;
117
:
6120
31
.
72.
Appleman
LJ
,
Berezovskaya
A
,
Grass
I
,
Boussiotis
VA
. 
CD28 costimulation mediates T cell expansion via IL-2-independent and IL-2-dependent regulation of cell cycle progression
.
J Immunol
2000
;
164
:
144
51
.
73.
Chunder
N
,
Wang
L
,
Chen
C
,
Hancock
WW
,
Wells
AD
. 
Cyclin-dependent kinase 2 controls peripheral immune tolerance
.
J Immunol
2012
;
189
:
5659
66
.
74.
De Simone
M
,
Arrigoni
A
,
Rossetti
G
,
Gruarin
P
,
Ranzani
V
,
Politano
C
, et al
Transcriptional landscape of human tissue lymphocytes unveils uniqueness of tumor-infiltrating T regulatory cells
.
Immunity
2016
;
45
:
1135
47
.
75.
Konig
S
,
Probst-Kepper
M
,
Reinl
T
,
Jeron
A
,
Huehn
J
,
Schraven
B
, et al
First insight into the kinome of human regulatory T cells
.
PLoS One
2012
;
7
:
e40896
.
76.
Heng
TS
,
Painter
MW
,
Elpek
K
,
Lukacs-Kornek
V
,
Mauermann
N
,
Turley
SJ
, et al
The Immunological Genome Project: networks of gene expression in immune cells
.
Nat Immunol
2008
;
9
:
1091
.
77.
Teo
ZL
,
Versaci
S
,
Dushyanthen
S
,
Caramia
F
,
Savas
P
,
Mintoff
CP
, et al
Combined CDK4/6 and PI3Kα inhibition is synergistic and immunogenic in triple-negative breast cancer
.
Cancer Res
2017
;
77
:
6340
52
.
78.
Zhang
J
,
Bu
X
,
Wang
H
,
Zhu
Y
,
Geng
Y
,
Nihira
NT
, et al
Cyclin D–CDK4 kinase destabilizes PD-L1 via cullin 3–SPOP to control cancer immune surveillance
.
Nature
2018
;
553
:
91
.
79.
Oh
SJ
,
Cho
H
,
Kim
S
,
Noh
KH
,
Song
KH
,
Lee
HJ
, et al
Targeting cyclin D-CDK4/6 sensitizes immune-refractory cancer by blocking the SCP3-NANOG axis
.
Cancer Res
2018
;
78
:
2638
53
.