The recent clinical success of targeted therapies in melanoma directed at the oncogene BRAF validates the concept of targeting oncogenes. The p16-cyclin D-CDK4/6-retinoblastoma protein pathway (CDK4 pathway) is dysregulated in 90% of melanomas, and is, therefore, an obvious therapeutic target for this disease. The main outcome of CDK4 activation is the phosphorylation and, thus, inhibition of the retinoblastoma protein leading to G1–S cell-cycle transition. In addition, CDK4 directly phosphorylates other proteins that promote cell-cycle progression and inhibit both cell senescence and apoptosis. In preclinical studies, the response to CDK4 inhibition correlates with genomic changes that increase CDK4 activity, most notably where the tumor suppressor CDKN2A (p16INK4A) is deleted. A central question is whether melanomas with activating events in the CDK4 pathway have become “addicted” to this signaling pathway, in which case inhibition of CDK4 would not simply induce cell-cycle arrest but induce cell death and tumor regression. Recently, a number of selective CDK4/6 inhibitors have entered clinical trials, and these compounds are showing great promise in that they are well tolerated and show clinical benefit. This review discusses the CDK4 pathway, its dysregulation in melanoma, the consequences of CDK4 pathway inhibition, and potential novel combinational strategies for the treatment of melanoma. Clin Cancer Res; 19(19); 5320–8. ©2013 AACR.

Cyclin-dependent kinases (CDK) drive cell-cycle progression, control transcriptional processes, and, thus, regulate cell proliferation. Considering that aberrant entry into the cell cycle and uncontrolled cell proliferation is a hallmark of cancer (1), it is not surprising that dysregulation of CDKs plays a central role in tumorigenesis. The p16-cyclin D-CDK4/6-retinoblastoma protein (RB1) pathway (CDK4 pathway) promotes G1–S cell-cycle transition, and this pathway is commonly dysregulated in most cancers. In melanoma, the CDK4 pathway is associated with activating genomic alterations in more than 90% of cases (2, 3), and, in both human and mouse models of melanoma, activation of the CDK4 pathway potently cooperates with mutant BRAF or NRAS in transformation of melanocytes (4–6). Furthermore, the RAS/RAF/MEK/ERK pathway, which is dysregulated in 65% to 90% of metastatic melanoma (7), enhances CDK4 pathway signaling through increasing cyclin D1 expression (8, 9), and this increase is implicated in the transforming ability of both RAS and RAF (10).

The recent clinical success of targeted therapies directed at the oncogene BRAF in melanoma (11, 12) validate the concept of targeting oncogenes in this disease and pave the way to potentially target other activated signaling pathways. The prevalence of activating events in the CDK4 pathway in melanoma highlights it as a potential therapeutic target. A central question discussed further here is whether melanomas with these activating events are “addicted” to the pathway, such that inhibition of CDK4 specifically targets cancer cells by inducing G1-arrest or alternately leading to other cellular outcomes. Currently, there are several inhibitors with varying selectivity for specific members of the CDK family, but CDK4/6 inhibitors have emerged as the most attractive therapeutic option because of their ability to inhibit cell proliferation and minimal toxic effects. The pyridopyrimidine PD0332991 (PD991: palbociclib) is the most advanced in clinical development, and studies to date with this compound have shown favorable pharmacokinetics, absence of significant off-target toxicities, and, most importantly, evidence of target modulation in humans (13–15).

This review discusses the CDK4 pathway, its dysregulation in melanoma, the consequences of CDK4 pathway inhibition, and potential novel combinational strategies for the treatment of melanoma.

CDK4 pathway

The mammalian cell cycle is regulated by two key classes of molecules—the regulatory cyclins and the catalytic CDKs that form active heterodimers leading to phosphorylation of target proteins. CDK4 and its close homolog CDK6 are serine/threonine kinases that form heterodimers with D-type cyclins and are central regulators of G1–S transition of the cell cycle (Fig. 1). Together with cyclin E–CDK2 complexes, cyclin D–CDK4/6 complexes sequentially phosphorylate the retinoblastoma protein (RB1), diminishing its ability to suppress RNA polymerase I and III activity (16, 17) and gene transcription (Fig. 2). Hypophosphorylated RB1 modulates gene transcription through its action on suppressing the E2F family of transcription factors and by recruiting histone deacetylases to the promoters of genes required for S-phase entry (18). These inhibitory actions of hypophosphorylated RB1 lead to the modulation of ribosome biogenesis, which has an impact on protein synthesis and transcription factors that coordinate subsequent cell-cycle progression, nucleotide biosynthesis, DNA replication, mitotic progression, and DNA damage repair (19). Thus, CDK4/6-mediated deactivation of RB1 is critical for cell-cycle progression. This process is negatively regulated by the tumor suppressor p16INK4A, which specifically inhibits the assembly and activation of cyclin D–CDK4/6 complexes. Further control of this pathway is via a negative feedback loop whereby CDK4/6 inactivation of RB1 relieves RB1-mediated suppression of p16INK4A (20), leading to increased p16INK4A and subsequent reduction in CDK4/6 activity. Thus, this negative feedback loop effectively works as a natural brake on the activation of this pathway (Fig. 1). Increased p16INK4A expression due to loss of RB1 is also seen in HPV-infected cells, where the E7 protein leads to RB1 degradation, and in cells in which there is a genetic loss of RB1 (21). These data confirm that the regulatory feedback from RB1 to p16INK4A exists under both physiologic and pathologic conditions. Under these circumstances where there is a permanent loss of RB1, this loss renders the increased p16INK4A expression incapable of inhibiting the cell cycle. In normal cells, p16INK4A levels are usually low, allowing cell-cycle progression. Increased p16INK4A expression upon oncogenic signaling, DNA damage, and physiologic aging (22, 23) inhibits cell proliferation, and these three processes also trigger cellular senescence, indicating that p16INK4A-mediated cell-cycle arrest is likely necessary for induction of senescence.

Figure 1.

CDK4 pathway. In response to mitogenic signaling, CDK4 and CDK6 associate with cyclin D to form active complexes that phosphorylate the retinoblastoma protein (RB1). Subsequent phosphorylation of RB1 by CDK2–cyclin E complex allows transcription of E2F regulated genes and transcription of both ribosomal and small RNAs by RNA polymerases. Phosphorylation of RB1 also inhibits its ability to induce senescence. p16INK4A and p14ARF are transcribed from the same gene (CDKN2A) and regulate cell-cycle progression by inhibiting CDK4/6 and CDK2 activity, respectively. Induction of p14ARF inhibits HDM2-mediated degradation of p53, thus increasing expression of p21, which then inhibits CDK2/cyclin E activity. p16INK4A negatively regulates the cell cycle by specifically inhibiting the assembly and activation of cyclin D–CDK4/6 complexes. Further control of this pathway is via a negative feedback loop whereby CDK4/6 inactivation of RB1 relieves RB1-mediated suppression of p16INK4A. In addition to the canonical CDK4 pathway, active CDK4/6–cyclin D complexes can directly phosphorylate and regulate transcription factors, including Smad3, MYC, and FOXM1 as well as MEP50, a PRMT5 coregulatory factor, resulting in RB1-independent regulation of cell-cycle progression, senescence, and apoptosis.

Figure 1.

CDK4 pathway. In response to mitogenic signaling, CDK4 and CDK6 associate with cyclin D to form active complexes that phosphorylate the retinoblastoma protein (RB1). Subsequent phosphorylation of RB1 by CDK2–cyclin E complex allows transcription of E2F regulated genes and transcription of both ribosomal and small RNAs by RNA polymerases. Phosphorylation of RB1 also inhibits its ability to induce senescence. p16INK4A and p14ARF are transcribed from the same gene (CDKN2A) and regulate cell-cycle progression by inhibiting CDK4/6 and CDK2 activity, respectively. Induction of p14ARF inhibits HDM2-mediated degradation of p53, thus increasing expression of p21, which then inhibits CDK2/cyclin E activity. p16INK4A negatively regulates the cell cycle by specifically inhibiting the assembly and activation of cyclin D–CDK4/6 complexes. Further control of this pathway is via a negative feedback loop whereby CDK4/6 inactivation of RB1 relieves RB1-mediated suppression of p16INK4A. In addition to the canonical CDK4 pathway, active CDK4/6–cyclin D complexes can directly phosphorylate and regulate transcription factors, including Smad3, MYC, and FOXM1 as well as MEP50, a PRMT5 coregulatory factor, resulting in RB1-independent regulation of cell-cycle progression, senescence, and apoptosis.

Close modal
Figure 2.

Retinoblastoma protein regulation of E2F target gene transcription and ribosomal RNA synthesis. A, activated hypophosphorylated RB1 binds to E2F transcriptional regulators, recruiting histone deacetylases, corepressors, and chromatin remodeling enzymes and, thus, suppressing target gene transcription. Phosphorylation of RB1 by activated CDK4/6 and CDK2 relieves inhibition and allows E2F target gene transcription. B, activated hypophosphorylated RB1 binds to the RNA polymerase I-specific factor UBF, inhibiting its association with the SL-1–RNA polymerase I complex, thereby inhibiting rRNA transcription. Phosphorylation of RB1 by activated CDK4/6 and CDK2 relieves inhibition and allows UBF to recruit the SL-1 complex and RNA polymerase I to initiate rDNA transcription.

Figure 2.

Retinoblastoma protein regulation of E2F target gene transcription and ribosomal RNA synthesis. A, activated hypophosphorylated RB1 binds to E2F transcriptional regulators, recruiting histone deacetylases, corepressors, and chromatin remodeling enzymes and, thus, suppressing target gene transcription. Phosphorylation of RB1 by activated CDK4/6 and CDK2 relieves inhibition and allows E2F target gene transcription. B, activated hypophosphorylated RB1 binds to the RNA polymerase I-specific factor UBF, inhibiting its association with the SL-1–RNA polymerase I complex, thereby inhibiting rRNA transcription. Phosphorylation of RB1 by activated CDK4/6 and CDK2 relieves inhibition and allows UBF to recruit the SL-1 complex and RNA polymerase I to initiate rDNA transcription.

Close modal

Although RB1 is considered the major substrate of the cyclin D–CDK4/6 complex, the RB1-related proteins p107 and p130 are also substrates and have a similar function to RB1 in regulating the cell cycle (24). In addition, there is increasing evidence that CDK4/6 independent of RB1 can regulate the cell cycle, survival, and senescence via direct phosphorylation of transcription factors, including Smad3 (25), MYC, and FOXM1 (26), or through targeting transcriptional coregulators including MEP50, a PRMT5 coregulatory factor (27).

CDK4 pathway in human melanoma

The early indications that the CDK4 pathway was likely a critical oncogenic pathway in melanoma were that the inhibitory mutations in CDKN2A or activating mutations in CDK4 in the germ line of humans led to an increase in the risk of developing melanoma well above any other cancer type (28). Loss of p16INK4A occurs in approximately 80% to 90% of metastatic tumors, most commonly via gene deletion (50–60%; ref. 29) but also via mutation (∼10%) and promoter methylation (20–30%; refs. 30–32). The loss of p16INK4A protein expression appears to progress from benign nevi being approximately 70% to 100% positive for p16INK4A protein, primary melanoma 30% to 80% positive, and metastatic disease only being 10% to 20% positive (33). The sequential loss of p16INK4A from benign nevi to metastatic melanoma indicates that this loss is vital in disease establishment and progression and reinforces the idea that activation of the CDK4 pathway is an early event and likely essential in melanoma progression.

P16INK4A specifically inhibits the assembly and activation of both cyclin D–CDK4 and cyclin D–CDK6 complexes; thus, the loss of p16INK4A in melanoma results in increased CDK4/6 signaling (34). In most cases, however, mutations in p16INK4A lead to decreased binding to CDK4 but not CDK6 (35, 36), underscoring a predominant role of CDK4 over CDK6 in the development of melanoma. Mutations in CDK4 (R24C) originally identified in melanoma-prone families (28) abolish the interaction with p16INK4A and, thus, render CDK4 constitutively active (37), further highlighting the importance of CDK4 activation in melanoma. Although CDK6 is not mutated in melanoma (38), and there is no focal amplification of the gene, CDK6 does lie within a large region that is amplified in at least 62% of tumors (29), suggesting it may play a role in this disease. Both CDK4 and CDK6, when complexed with cyclin D1, promote cell-cycle progression through the phosphorylation and deactivation of RB1. Cyclin D1 is a known oncogene in many tumor types, including melanoma. It is amplified in 18% of melanomas (29, 39), and, more importantly, increased signaling in the RAS/RAF/MEK/ERK pathway in BRAF- and NRAS-mutant melanomas triggers the expression of cyclin D1 through both gene transcription and protein stabilization (8, 9, 40). Finally, germ line inactivation of the RB1 gene also predisposes patients to melanoma (28). In summary, there are multiple gene deletions and mutations along the CDK4 pathway that increase signaling, leading to cell-cycle progression and an increased risk of developing malignant melanoma.

Models of melanoma with disrupted CDK4 pathway

The association of melanoma with loss of the CDKN2A gene prompted studies assessing the relative importance of the two distinct proteins, p16INK4A and p14ARF (p19ARF in mouse), that are encoded by this gene. Whereas p16INK4A inhibits CDK4/6 activity by maintaining RB1 in its hypophosphorylated, antiproliferative state (41), p14ARF inhibits HDM2-mediated ubiquitination and subsequent degradation of p53 (42); thus, loss of p14ARF inactivates p53 (Fig. 1). In mice with targeted deletion of the CDKN2A gene, which eliminates both p16INK4A and p19ARF (mouse homolog of p14ARF), spontaneous tumors developed at an early age, and these mice are highly sensitive to carcinogenic agents (43). Mice lacking p19ARF but expressing functional p16INK4A also develop tumors early in life (44), indicating disruption of p19ARF alone predisposes mice to tumorigenesis. Loss of p16INK4A in mice is not sufficient to induce melanoma but does mimic the melanoma predisposition seen in humans (45, 46). In these p16INK4A null mice, susceptibility to spontaneous melanoma was low and, similar to p19ARF loss, tumor incidence in these mice increased in response to carcinogen.

The majority of mutations in the CDKN2A gene specifically target and impair p16INK4A function, whereas mutations uniquely targeting p14ARF are rare. Of the mutations in the CDKN2A gene that pertain to both p16INK4A and p14ARF that have been tested, all show compromised p16INK4A function and a small subset shows compromised p14ARF function (47). Thus, loss of p16INK4A function in the majority of melanomas highlights the importance of this protein in tumor suppression, whereas the rare loss of p14ARF function raises questions about its role in this disease. Inactivation of p53 via mutation is a common event in human cancer (48); however, these events are low in melanoma (49, 50). Melanomas develop with high penetrance in mouse models where there is both loss of p53 and activation of the RAS/RAF/MEK/ERK pathway (51, 52), indicating that p53 inactivation can contribute to this disease. Further evidence for a role of p53 inactivation in melanoma progression is that high expression of HDM4 is found in 65% of melanomas. This protein inhibits p53 activity and, in a mouse model of melanoma driven by the oncogene NRAS, overexpression of MDM4 (mouse homolog of HDM4) enhanced tumorigenesis (53). Thus, p53 inactivation in melanoma in the majority of tumors is likely via HDM4 and thus negates the need for loss of p53 function through mutation or loss of p14ARF.

The common mutation in CDK4 (CDK4R24C) in melanoma inhibits the interaction with p16INK4A and thus leads to constitutive activation. Mice with a knockin of CDK4R24C display a similar phenotype to mice with p16INK4A loss, in that there is a low incidence of spontaneous melanoma that is augmented in response to carcinogen (37, 54, 55). Further evidence that CDK4 pathway activation may be necessary for melanoma progression is that both cyclin D1- and CDK4-null mice are relatively resistant to chemical-induced skin cancer (56, 57). Furthermore, in a model of human skin tissue regenerated with melanocytes, CDK4 overexpression or inhibition of p53, together with activated RAS, produced invasive human melanoma (5).

From studies in mice, it is clear that loss of p16INK4A or activation of CDK4 as single events is not sufficient to induce melanoma but does predispose animals to tumor formation, which aligns with the clinical data showing increased risk of developing melanoma in patients who have these genetic changes. Furthermore, systematic interrogation of the CDK4 pathway in melanocytes has shown that activation of the pathway, together with the introduction of telomerase reverse transcriptase (TERT), can lead to immortalization of these cells but is not sufficient for transformation. P16INK4A-deficient human melanocytes with inactivating mutations in both alleles of p16INK4A/p14ARF undergo delayed senescence but can be immortalized by the introduction of TERT (58). Furthermore, TERT, in combination with CDK4 overexpression or inhibition of RB1, immortalizes cells (59). Melanocytes are unique in that activation of the CDK4 pathway, in addition to expression of TERT, is required for immortalization, whereas introduction of TERT alone in most cell lines is sufficient.

Senescence

Similar to melanoma (60), benign nevi harbor activating mutations in BRAF (61) and, less frequently, NRAS (62). Unlike melanoma, however, nevi are composed of mainly senescent cells that are in cell-cycle arrest, have increased expression of p16INK4A, and express senescence biomarkers including SA-β-galactosidase (63). In human melanocytes, expression of BRAFV600E induces senescence (63) and, in mouse models, when BRAFV600E is expressed in the melanocytic lineage, nevi display the senescent phenotype (64, 65). Senescence was originally defined as an irreversible cell-cycle arrest and, thus, oncogene-induced senescence in melanocytes is thought to be an effective barrier preventing melanoma. CDK4 pathway activation, p53 inactivation (16, 66, 67), and, more recently, activation of the PI3K pathway (68) have been implicated in melanocytes escaping from senescence. Whether these senescent escape event(s) occur before, concurrently, or after the initiating senescent signal is, as yet, unresolved (69).

The role of both p16INK4A and p53 in overcoming senescence has been addressed in several mouse models. In BRAFV600E transgenic mice, progression from benign nevi to melanoma occurs with loss of both p16INK4A and loss of the senescent phenotype, indicating that p16INK4A is involved. In these mice, the frequency of melanoma is greatly increased when they are crossed with CDKN2A-null mice that have loss of both p16INK4A and p19ARF (64). Furthermore, in mice harboring both activated CDK4 and HRAS, there is enhanced susceptibility to melanoma compared with HRAS transgenic mice, and ultraviolet radiation increases the number of lesions and decreases the age of onset (70).

In both humans and mouse models, the loss of p16INK4A is associated with loss of the senescent phenotype, indicating that p16INK4A may play an active role in inducing senescence. Indeed, melanocytes overexpressing p16INK4A undergo a rapid cell-cycle arrest and induction of cellular senescence that is p53-independent but pRB1-dependent (71). Inhibition of this senescence response could be induced by overexpression of either CDK4 or CDK6 but not CDK2, showing that p16INK4A initiates a CDK4/6-dependent senescence program. Inhibition of CDK4/6 with the small-molecule inhibitor palbociclib induces senescence in melanoma cells (26), further supporting a role of the CDK4 pathway in overcoming senescence in melanoma. Recently, several CDK4/6 substrates were identified as possible mediators of senescence, including FOXM1 (26). FOXM1 is involved in regulating cell-cycle progression and the suppression of cellular senescence (72, 73). The CDK4/6-mediated suppression of senescence induced by FOXM1 was independent of pRB1 and due to direct phosphorylation of FOXM1, which increased its activity and stability. Senescence induced by knockdown of FOXM1, however, did not totally recapitulate the effect of the specific CDK4/6 inhibitor (26), indicating that CDK4/6 mediates robust senescence suppression through both pRB1-dependent and -independent mechanisms, which includes FOXM1 activation. The observation that interfering with the CDK4 pathway leads to a robust senescence response opens the option of targeting this pathway and exploiting senescence to induce melanoma regression (74, 75).

There is a potential pitfall in exploiting senescence induction as a therapeutic strategy for the treatment of melanoma: senescence, originally thought to be an irreversible cell state, may be reversible. In cell line models, including melanocytes, manipulating p53, cytokines, cellular metabolism, or the phosphoinositide 3 kinase (PI3K) pathway (69, 76) has been shown to reverse the senescent phenotype. Furthermore, the substantial number of melanocytic nevi contiguous with melanoma also suggests that some melanomas arise from these nevi and there is genetic evidence for this (68, 77). In addition, in a mouse model of BRAFV600E-driven nevi, acute depletion of PTEN in the nevi-induced melanoma (68). Whether the cell of origin from which melanoma arise within a nevus is a fully senescent cell and whether a senescent cell that is not replicating DNA can acquire genetic and/or epigenetic defects to become malignant is still debatable.

Clinical development of CDK4 inhibitors and opportunities for combination with MEK pathway inhibitors in melanoma

Genomic data indicating a central role of dysregulation of the CDK4 pathway in human cancer led to the development of small-molecule inhibitors of the kinase activity of CDK4/6. Early compounds including the flavone flavopiridol lacked selectivity for CDK4 over other CDKs, and, in the case of flavopiridol, the molecule inhibited CDK9/pTEFb and RNA polymerase II transcription (78). Although flavopiridol has been evaluated in phase II studies in melanoma, where it did not show clinical activity (79), it was not clear to what extent flavopiridol was able to inhibit CDK4 at the phase II dose and schedule. Furthermore, this study did not enrich for patients with genomic events known to activate CDK4. As such flavopiridol has not enabled clear conclusions to be drawn on the role of CDK4 inhibition as a treatment strategy for melanoma.

More recently, a number of more selective CDK4 inhibitors have entered clinical trials. Palbociclib is the most extensively evaluated compound in patients. It is able to inhibit CDK4 as shown by a reduction in RB1 phosphorylation, inhibit uptake of 18F-fluoro-l-thymidine (cell proliferation), and induce clinical responses in patients with mantle cell lymphoma that have cyclin-D1 translocations (15). Dose-limiting toxicity associated with palbociclib was myelosuppression consistent with on-target inhibition (13–15). Other CDK4/6 inhibitors in clinical development include LEE011, LY2835219, and P276-00. All these compounds show good selectivity for CDK4 in preclinical studies (Table 1; refs. 80–83).

Table 1.

Activity of CDK4/6 inhibitors in clinical trials

IC50Target inhibition (p-RB)Clinical trial involving melanoma patients
CDK4/6 inhibitorCDK4/D1CDK6/D3CDK1/BCDK2/ACDK7CDK9/T1In vitroIn vivo (xenografts)PhenotypeTrialTumor typeReferences
PD0332991 (palbociclib) ++++ ++++ Yes Yes G1 arrest, senescence apoptosis - NCT01037790 Phase II (recruiting) Recurrent and stage IV melanoma (26, 84, 86) 
LY2835219 ++++ ++++ Yes Yes G1 Arrest - NCT01394016 Phase I: MTD (recruiting) Advanced cancer (including melanoma) (80, 81) 
LEE011 ++++ ++++ Yes Yes G1 arrest, senescence apoptosis - NCT01237236 Phase I: LEE011 MTD (recruiting) Advanced solid tumors (including melanoma) (Sudha Parasuraman, Novartis; personal communication) 
          - NCT01781572 NRAS mutant melanoma  
          Phase Ib & II: LEE011+ MEK162 MTD (recruiting)   
          - NCT01777776 BRAF mutant melanoma  
          Phase I & II: LEE011 ± LGX818 MTD (recruiting)   
P276-00 ++++ +++ ++++ +++ ++ ++++ Yes Yes G1–G2 arrest, apoptosis - NCT00835419 Phase II (complete) Melanoma positive for cyclin D1 expression (82, 83) 
Flavopiridol ++++ ++++ ++++ ++++ +++ Yes NT G1–G2 arrest, apoptosis - NCT00005971 Phase II (completed; 44% SD, No OR) Metastatic melanoma (79, 101) 
          - NCT00003690 Phase I carboplatin/cisplatin/alvocidib + flavopiridol MTD (completed: 34%SD, no OR) Advanced solid tumors (including melanoma)  
IC50Target inhibition (p-RB)Clinical trial involving melanoma patients
CDK4/6 inhibitorCDK4/D1CDK6/D3CDK1/BCDK2/ACDK7CDK9/T1In vitroIn vivo (xenografts)PhenotypeTrialTumor typeReferences
PD0332991 (palbociclib) ++++ ++++ Yes Yes G1 arrest, senescence apoptosis - NCT01037790 Phase II (recruiting) Recurrent and stage IV melanoma (26, 84, 86) 
LY2835219 ++++ ++++ Yes Yes G1 Arrest - NCT01394016 Phase I: MTD (recruiting) Advanced cancer (including melanoma) (80, 81) 
LEE011 ++++ ++++ Yes Yes G1 arrest, senescence apoptosis - NCT01237236 Phase I: LEE011 MTD (recruiting) Advanced solid tumors (including melanoma) (Sudha Parasuraman, Novartis; personal communication) 
          - NCT01781572 NRAS mutant melanoma  
          Phase Ib & II: LEE011+ MEK162 MTD (recruiting)   
          - NCT01777776 BRAF mutant melanoma  
          Phase I & II: LEE011 ± LGX818 MTD (recruiting)   
P276-00 ++++ +++ ++++ +++ ++ ++++ Yes Yes G1–G2 arrest, apoptosis - NCT00835419 Phase II (complete) Melanoma positive for cyclin D1 expression (82, 83) 
Flavopiridol ++++ ++++ ++++ ++++ +++ Yes NT G1–G2 arrest, apoptosis - NCT00005971 Phase II (completed; 44% SD, No OR) Metastatic melanoma (79, 101) 
          - NCT00003690 Phase I carboplatin/cisplatin/alvocidib + flavopiridol MTD (completed: 34%SD, no OR) Advanced solid tumors (including melanoma)  

NOTE: IC50s should not be compared between compounds given different kinase assay conditions.

++++ IC50 < 100 nmol/L; +++ IC50 < 500 nmol/L; ++ IC50 < 5 μmol/L; + IC50 > 5 μmol/L.

Abbreviations: ?, Unknown; MTD, maximum tolerated dose; NT, not tested; OR, objective response; P-RB, phosphorylated retinoblastoma protein; SD, stable disease.

Single-agent activity of CDK4-inhibitors is well described in preclinical models of a variety of cancers including melanoma with inhibition of CDK4 leading to cell-cycle arrest, senescence, and/or cell death (26, 82, 84, 85). Activity appears to be greater where genomic changes increase activity of CDK4, most notably with higher activity seen where CDKN2A is deleted (86, 87), and resistance is associated with loss of RB1 (85). Intriguingly, preclinical studies indicate that, in some situations, inhibition of CDK4 will not only induce G1-arrest or senescence but also induce cell death (82, 84, 88). Whether these tumor cells are “addicted” to CDK4 signaling and, thus, CDK4 inhibition is a treatment strategy capable of inducing cell death and tumor regression will ultimately have to be evaluated in clinical trials. Nonetheless, as CDK4 is activated in conjunction with other signaling pathways, and as inhibition of CDK4 should not be assumed to be sufficient to obtain tumor regression and clinical response, there is significant interest in combining CDK4/6 inhibitors with other cancer therapeutics. Preclinical studies have suggested that CDK4 inhibition may reduce the cytotoxicity of chemotherapy by inducing a G1 arrest (89) and, therefore, greater attention has focused on combinations with inhibitors of other signaling pathways including the RAS/RAF/MEK/ERK pathway in melanoma (90) and estrogen-receptor signaling in breast cancer (86, 91, 92). The recent success of palbociclib in combination with letrozole in ER+/HER2 advanced breast cancer supports this idea of combining palbociclib with inhibitors of other signaling pathways and biomarker-based patient selection (92).

Activating mutations in RAS or BRAF clearly activate ERK-signaling and the CDK4 pathway, defining CDK4 as a component of the RAS/RAF/MEK/ERK signaling pathway. Therefore, the concept of combining BRAF or MEK inhibitors with CDK4 inhibitors is appealing. Kwong and colleagues have shown synergistic efficacy of the MEK-inhibitor GSK1120212 when combined with the CDK4-inhibitor palbociclib in NRAS-mutant melanoma (90). Furthermore, decreased clinical benefit from dabrafenib in BRAF-mutant melanoma patients was associated with p16INK4A deletion, indicating combination of a BRAF and CDK4 inhibitor would likely be superior to single-agent dabrafenib treatment (93). Further studies and clinical trials will determine if this approach is more broadly applicable to NRAS- and BRAF-mutant melanoma.

BRAF inhibitor resistance is associated with mechanisms that reactivate ERK signaling (94) and expression of cyclinD1 is increased in a proportion of these cases (95). These data suggest that the combination of a BRAF-inhibitor and CDK4 inhibitor is worthy of assessment in patients with acquired resistance to BRAF-inhibitors or, perhaps, even more appealing is to use the combination of BRAF/MEK and CDK4-inhibition upfront in an attempt to prevent or delay the emergence of resistance.

The concept of combining inhibitors of BRAF or MEK with inhibitors of CDK4 raises the possibility of potential antagonism. As discussed earlier, it is proposed that activation of CDK4 through loss of p16INK4A is the central mechanism to overcome NRAS- or BRAF-mediated oncogene-induced senescence to allow melanomas to develop. Therefore, any reduction in ERK signaling may attenuate the induction of senescence associated with inhibition of CDK4. However, could BRAF/MEK inhibition and CDK4 inhibition be used sequentially and interleafed such that when ERK-pathway inhibition is removed CDK4 inhibition be applied to promote senescence in surviving cells? Given the recent observation that removing BRAF/MEK inhibition may lead to temporary hyperactivation of ERK-signaling (96), this approach might be successful in driving oncogene-induced senescence in advanced BRAF- and NRAS-mutant melanoma. In addition, the potential immunosuppressive effects of CDK4 inhibition (97, 98) need to be considered, especially in light of the recent clinical success of inhibitory T-cell checkpoint blockade in patients with advanced melanoma (99, 100).

Finally, based on preclinical studies, patients most likely to respond to CDK4 inhibition are those that have increased CDK4 signaling through loss of CDKN2A, whereas loss of RB1 would predict resistance. The degree of CDK4 inhibition could be assessed by measuring reduction in phosphorylated RB1, cell proliferation by Ki67 staining, or 18F-fluoro-l-thymidine by PET scans, and cell senescence via β-galactosidase staining. The identification of CDK4-inhibitor-response biomarkers that correlate with clinical benefit will facilitate clinical evaluation of candidate CDK4 inhibitors and dosing regimens as monotherapy or in combination with other therapeutics.

With the introduction of a number of highly selective CDK4/6 inhibitors into clinical trials, the field is now poised to definitively assess whether CDK4, clearly a central mediator of oncogenic signaling in melanoma cells, is a drug target that is able to improve survival and quality of life in patients with melanoma.

G.A. McArthur received commercial research funding from Pfizer, Novartis, and Millennium. K.E. Sheppard received commercial research funding from Pfizer.

Conception and design: K.E. Sheppard, G.A. McArthur

Development of methodology: G.A. McArthur

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): K.E. Sheppard, G.A. McArthur

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): K.E. Sheppard, G.A. McArthur

Writing, review, and/or revision of the manuscript: K.E. Sheppard, G.A. McArthur

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): K.E. Sheppard

Study supervision: G.A. McArthur

The work was financially supported by grants from the National Health and Medical Research Council (NHMRC) of Australia grant no. 10002655 to G.A. McArthur and grant no. 1042986 to G.A. McArthur and K.E. Sheppard. G.A. McArthur is a recipient of an NHMRC practitioner fellowship.

1.
Hanahan
D
,
Weinberg
RA
. 
Hallmarks of cancer: the next generation
.
Cell
2011
;
144
:
646
74
.
2.
Walker
GJ
,
Flores
JF
,
Glendening
JM
,
Lin
AH
,
Markl
ID
,
Fountain
JW
. 
Virtually 100% of melanoma cell lines harbor alterations at the DNA level within CDKN2A, CDKN2B, or one of their downstream targets
.
Genes Chromosomes Cancer
1998
;
22
:
157
63
.
3.
Curtin
JA
,
Fridlyand
J
,
Kageshita
T
,
Patel
HN
,
Busam
KJ
,
Kutzner
H
, et al
Distinct sets of genetic alterations in melanoma
.
N Engl J Med
2005
;
353
:
2135
47
.
4.
Kannan
K
,
Sharpless
NE
,
Xu
J
,
O'Hagan
RC
,
Bosenberg
M
,
Chin
L
. 
Components of the Rb pathway are critical targets of UV mutagenesis in a murine melanoma model
.
Proc Natl Acad Sci U S A
2003
;
100
:
1221
5
.
5.
Chudnovsky
Y
,
Adams
AE
,
Robbins
PB
,
Lin
Q
,
Khavari
PA
. 
Use of human tissue to assess the oncogenic activity of melanoma-associated mutations
.
Nat Genet
2005
;
37
:
745
9
.
6.
Monahan
KB
,
Rozenberg
GI
,
Krishnamurthy
J
,
Johnson
SM
,
Liu
W
,
Bradford
MK
, et al
Somatic p16(INK4a) loss accelerates melanomagenesis
.
Oncogene
2010
;
29
:
5809
17
.
7.
Chin
L
,
Garraway
LA
,
Fisher
DE
. 
Malignant melanoma: genetics and therapeutics in the genomic era
.
Genes Dev
2006
;
20
:
2149
82
.
8.
Filmus
J
,
Robles
AI
,
Shi
W
,
Wong
MJ
,
Colombo
LL
,
Conti
CJ
. 
Induction of cyclin D1 overexpression by activated ras
.
Oncogene
1994
;
9
:
3627
33
.
9.
Albanese
C
,
Johnson
J
,
Watanabe
G
,
Eklund
N
,
Vu
D
,
Arnold
A
, et al
Transforming p21ras mutants and c-Ets-2 activate the cyclin D1 promoter through distinguishable regions
.
J Biol Chem
1995
;
270
:
23589
97
.
10.
Liu
JJ
,
Chao
JR
,
Jiang
MC
,
Ng
SY
,
Yen
JJ
,
Yang-Yen
HF
. 
Ras transformation results in an elevated level of cyclin D1 and acceleration of G1 progression in NIH 3T3 cells
.
Mol Cell Biol
1995
;
15
:
3654
63
.
11.
Flaherty
KT
,
Puzanov
I
,
Kim
KB
,
Ribas
A
,
McArthur
GA
,
Sosman
JA
, et al
Inhibition of mutated, activated BRAF in metastatic melanoma
.
N Engl J Med
2010
;
363
:
809
19
.
12.
Chapman
PB
,
Hauschild
A
,
Robert
C
,
Haanen
JB
,
Ascierto
P
,
Larkin
J
, et al
Improved survival with vemurafenib in melanoma with BRAF V600E mutation
.
N Engl J Med
2011
;
364
:
2507
16
.
13.
Schwartz
GK
,
LoRusso
PM
,
Dickson
MA
,
Randolph
SS
,
Shaik
MN
,
Wilner
KD
, et al
Phase I study of PD 0332991, a cyclin-dependent kinase inhibitor, administered in 3-week cycles (Schedule 2/1)
.
Br J Cancer
2011
;
104
:
1862
8
.
14.
Flaherty
KT
,
Lorusso
PM
,
Demichele
A
,
Abramson
VG
,
Courtney
R
,
Randolph
SS
, et al
Phase I, dose-escalation trial of the oral cyclin-dependent kinase 4/6 inhibitor PD 0332991, administered using a 21-day schedule in patients with advanced cancer
.
Clin Cancer Res
2012
;
18
:
568
76
.
15.
Leonard
JP
,
LaCasce
AS
,
Smith
MR
,
Noy
A
,
Chirieac
LR
,
Rodig
SJ
, et al
Selective CDK4/6 inhibition with tumor responses by PD0332991 in patients with mantle cell lymphoma
.
Blood
2012
;
119
:
4597
607
.
16.
Hannan
KM
,
Hannan
RD
,
Smith
SD
,
Jefferson
LS
,
Lun
M
,
Rothblum
LI
. 
Rb and p130 regulate RNA polymerase I transcription: Rb disrupts the interaction between UBF and SL-1
.
Oncogene
2000
;
19
:
4988
99
.
17.
Sutcliffe
JE
,
Brown
TR
,
Allison
SJ
,
Scott
PH
,
White
RJ
. 
Retinoblastoma protein disrupts interactions required for RNA polymerase III transcription
.
Mol Cell Biol
2000
;
20
:
9192
202
.
18.
Shapiro
GI
. 
Cyclin-dependent kinase pathways as targets for cancer treatment
.
J Clin Oncol
2006
;
24
:
1770
83
.
19.
Knudsen
ES
,
Knudsen
KE
. 
Tailoring to RB: tumour suppressor status and therapeutic response
.
Nat Rev Cancer
2008
;
8
:
714
24
.
20.
Li
Y
,
Nichols
MA
,
Shay
JW
,
Xiong
Y
. 
Transcriptional repression of the D-type cyclin-dependent kinase inhibitor p16 by the retinoblastoma susceptibility gene product pRb
.
Cancer Res
1994
;
54
:
6078
82
.
21.
Dyson
N
,
Howley
PM
,
Munger
K
,
Harlow
E
. 
The human papilloma virus-16 E7 oncoprotein is able to bind to the retinoblastoma gene product
.
Science
1989
;
243
:
934
7
.
22.
Bartkova
J
,
Rezaei
N
,
Liontos
M
,
Karakaidos
P
,
Kletsas
D
,
Issaeva
N
, et al
Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints
.
Nature
2006
;
444
:
633
7
.
23.
Ressler
S
,
Bartkova
J
,
Niederegger
H
,
Bartek
J
,
Scharffetter-Kochanek
K
,
Jansen-Durr
P
, et al
p16INK4A is a robust in vivo biomarker of cellular aging in human skin
.
Aging Cell
2006
;
5
:
379
89
.
24.
Weinberg
RA
. 
The retinoblastoma protein and cell cycle control
.
Cell
1995
;
81
:
323
30
.
25.
Matsuura
I
,
Denissova
NG
,
Wang
G
,
He
D
,
Long
J
,
Liu
F
. 
Cyclin-dependent kinases regulate the antiproliferative function of Smads
.
Nature
2004
;
430
:
226
31
.
26.
Anders
L
,
Ke
N
,
Hydbring
P
,
Choi
YJ
,
Widlund
HR
,
Chick
JM
, et al
A systematic screen for CDK4/6 substrates links FOXM1 phosphorylation to senescence suppression in cancer cells
.
Cancer Cell
2011
;
20
:
620
34
.
27.
Aggarwal
P
,
Vaites
LP
,
Kim
JK
,
Mellert
H
,
Gurung
B
,
Nakagawa
H
, et al
Nuclear cyclin D1/CDK4 kinase regulates CUL4 expression and triggers neoplastic growth via activation of the PRMT5 methyltransferase
.
Cancer Cell
2010
;
18
:
329
40
.
28.
Tsao
H
,
Chin
L
,
Garraway
LA
,
Fisher
DE
. 
Melanoma: from mutations to medicine
.
Genes Dev
2012
;
26
:
1131
55
.
30.
Hocker
T
,
Tsao
H
. 
Ultraviolet radiation and melanoma: a systematic review and analysis of reported sequence variants
.
Hum Mutat
2007
;
28
:
578
88
.
31.
Freedberg
DE
,
Rigas
SH
,
Russak
J
. 
Frequent p16-independent inactivation of p14ARF in human melanoma
.
J Natl Cancer Inst
2008
;
100
:
784
95
.
32.
Jonsson
A
,
Tuominen
R
,
Grafstrom
E
,
Hansson
J
,
Egyhazi
S
. 
High frequency of p16(INK4A) promoter methylation in NRAS-mutated cutaneous melanoma
.
J Invest Dermatol
2010
;
130
:
2809
17
.
33.
Sanki
A
,
Li
W
,
Colman
M
. 
Reduced expression of p16 and p27 is correlated with tumour progression in cutaneous melanoma
.
Pathology
2007
;
39
:
551
7
.
34.
Fung
C
,
Pupo
GM
,
Scolyer
RA
,
Kefford
RF
,
Rizos
H
. 
p16(INK) (4a) deficiency promotes DNA hyper-replication and genetic instability in melanocytes
.
Pigment Cell Melanoma Res
2013
;
26
:
236
46
.
35.
McKenzie
HA
,
Fung
C
,
Becker
TM
,
Irvine
M
,
Mann
GJ
,
Kefford
RF
, et al
Predicting functional significance of cancer-associated p16(INK4a) mutations in CDKN2A
.
Hum Mutat
2010
;
31
:
692
701
.
36.
Jones
R
,
Ruas
M
,
Gregory
F
,
Moulin
S
,
Delia
D
,
Manoukian
S
, et al
A CDKN2A mutation in familial melanoma that abrogates binding of p16INK4a to CDK4 but not CDK6
.
Cancer Res
2007
;
67
:
9134
41
.
37.
Rane
SG
,
Cosenza
SC
,
Mettus
RV
,
Reddy
EP
. 
Germ line transmission of the Cdk4(R24C) mutation facilitates tumorigenesis and escape from cellular senescence
.
Mol Cell Biol
2002
;
22
:
644
56
.
38.
Shennan
MG
,
Badin
AC
,
Walsh
S
,
Summers
A
,
From
L
,
McKenzie
M
, et al
Lack of germline CDK6 mutations in familial melanoma
.
Oncogene
2000
;
19
:
1849
52
.
39.
Gast
A
,
Scherer
D
,
Chen
B
,
Bloethner
S
,
Melchert
S
,
Sucker
A
, et al
Somatic alterations in the melanoma genome: a high-resolution array-based comparative genomic hybridization study
.
Genes Chromosomes Cancer
2010
;
49
:
733
45
.
40.
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
.
41.
Serrano
M
,
Hannon
GJ
,
Beach
D
. 
A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4
.
Nature
1993
;
366
:
704
7
.
42.
Freedman
DA
,
Wu
L
,
Levine
AJ
. 
Functions of the MDM2 oncoprotein
.
Cell Mol Life Sci
1999
;
55
:
96
107
.
43.
Serrano
M
,
Lee
H
,
Chin
L
,
Cordon-Cardo
C
,
Beach
D
,
DePinho
RA
. 
Role of the INK4a locus in tumor suppression and cell mortality
.
Cell
1996
;
85
:
27
37
.
44.
Kamijo
T
,
Zindy
F
,
Roussel
MF
,
Quelle
DE
,
Downing
JR
,
Ashmun
RA
, et al
Tumor suppression at the mouse INK4a locus mediated by the alternative reading frame product p19ARF
.
Cell
1997
;
91
:
649
59
.
45.
Krimpenfort
P
,
Quon
KC
,
Mooi
WJ
,
Loonstra
A
,
Berns
A
. 
Loss of p16Ink4a confers susceptibility to metastatic melanoma in mice
.
Nature
2001
;
413
:
83
6
.
46.
Sharpless
NE
,
Bardeesy
N
,
Lee
KH
,
Carrasco
D
,
Castrillon
DH
,
Aguirre
AJ
, et al
Loss of p16Ink4a with retention of p19Arf predisposes mice to tumorigenesis
.
Nature
2001
;
413
:
86
91
.
47.
Rizos
H
,
Darmanian
AP
,
Holland
EA
,
Mann
GJ
,
Kefford
RF
. 
Mutations in the INK4a/ARF melanoma susceptibility locus functionally impair p14ARF
.
J Biol Chem
2001
;
276
:
41424
34
.
48.
Vousden
KH
,
Prives
C
. 
Blinded by the light: the growing complexity of p53
.
Cell
2009
;
137
:
413
31
.
49.
Chin
L
. 
The genetics of malignant melanoma: lessons from mouse and man
.
Nat Rev Cancer
2003
;
3
:
559
70
.
50.
Hodis
E
,
Watson
IR
,
Kryukov
GV
,
Arold
ST
,
Imielinski
M
,
Theurillat
JP
, et al
A landscape of driver mutations in melanoma
.
Cell
2012
;
150
:
251
63
.
51.
Bardeesy
N
,
Bastian
BC
,
Hezel
A
,
Pinkel
D
,
DePinho
RA
,
Chin
L
. 
Dual inactivation of RB and p53 pathways in RAS-induced melanomas
.
Mol Cell Biol
2001
;
21
:
2144
53
.
52.
Sharpless
NE
,
Kannan
K
,
Xu
J
,
Bosenberg
MW
,
Chin
L
. 
Both products of the mouse Ink4a/Arf locus suppress melanoma formation in vivo
.
Oncogene
2003
;
22
:
5055
9
.
53.
Gembarska
A
,
Luciani
F
,
Fedele
C
,
Russell
EA
,
Dewaele
M
,
Villar
S
, et al
MDM4 is a key therapeutic target in cutaneous melanoma
.
Nat Med
2012
;
18
:
1239
47
.
54.
Sotillo
R
,
Dubus
P
,
Martin
J
,
de la Cueva
E
,
Ortega
S
,
Malumbres
M
, et al
Wide spectrum of tumors in knock-in mice carrying a Cdk4 protein insensitive to INK4 inhibitors
.
EMBO J
2001
;
20
:
6637
47
.
55.
Sotillo
R
,
Garcia
JF
,
Ortega
S
,
Martin
J
,
Dubus
P
,
Barbacid
M
, et al
Invasive melanoma in Cdk4-targeted mice
.
Proc Natl Acad Sci U S A
2001
;
98
:
13312
7
.
56.
Zou
X
,
Ray
D
,
Aziyu
A
,
Christov
K
,
Boiko
AD
,
Gudkov
AV
, et al
Cdk4 disruption renders primary mouse cells resistant to oncogenic transformation, leading to Arf/p53-independent senescence
.
Genes Dev
2002
;
16
:
2923
34
.
57.
Robles
AI
,
Rodriguez-Puebla
ML
,
Glick
AB
,
Trempus
C
,
Hansen
L
,
Sicinski
P
, et al
Reduced skin tumor development in cyclin D1-deficient mice highlights the oncogenic ras pathway in vivo
.
Genes Dev
1998
;
12
:
2469
74
.
58.
Sviderskaya
EV
,
Gray-Schopfer
VC
,
Hill
SP
,
Smit
NP
,
Evans-Whipp
TJ
,
Bond
J
, et al
p16/cyclin-dependent kinase inhibitor 2A deficiency in human melanocyte senescence, apoptosis, and immortalization: possible implications for melanoma progression
.
J Natl Cancer Inst
2003
;
95
:
723
32
.
59.
Gray-Schopfer
VC
,
Cheong
SC
,
Chong
H
,
Chow
J
,
Moss
T
,
Abdel-Malek
ZA
, et al
Cellular senescence in naevi and immortalisation in melanoma: a role for p16?
Br J Cancer
2006
;
95
:
496
505
.
60.
Davies
H
,
Bignell
GR
,
Cox
C
,
Stephens
P
,
Edkins
S
,
Clegg
S
, et al
Mutations of the BRAF gene in human cancer
.
Nature
2002
;
417
:
949
54
.
61.
Pollock
PM
,
Harper
UL
,
Hansen
KS
,
Yudt
LM
,
Stark
M
,
Robbins
CM
, et al
High frequency of BRAF mutations in nevi
.
Nat Genet
2003
;
33
:
19
20
.
62.
Bauer
J
,
Curtin
JA
,
Pinkel
D
,
Bastian
BC
. 
Congenital melanocytic nevi frequently harbor NRAS mutations but no BRAF mutations
.
J Invest Dermatol
2007
;
127
:
179
82
.
63.
Michaloglou
C
,
Vredeveld
LC
,
Soengas
MS
. 
BRAFE600-associated senescence-like cell cycle arrest of human naevi
.
Nature
2005
;
436
:
720
4
.
64.
Goel
VK
,
Ibrahim
N
,
Jiang
G
,
Singhal
M
,
Fee
S
,
Flotte
T
, et al
Melanocytic nevus-like hyperplasia and melanoma in transgenic BRAFV600E mice
.
Oncogene
2009
;
28
:
2289
98
.
65.
Dhomen
N
,
Reis-Filho
JS
,
da Rocha Dias
S
,
Hayward
R
,
Savage
K
,
Delmas
V
, et al
Oncogenic Braf induces melanocyte senescence and melanoma in mice
.
Cancer Cell
2009
;
15
:
294
303
.
66.
Lin
AW
,
Barradas
M
,
Stone
JC
,
van Aelst
L
,
Serrano
M
,
Lowe
SW
. 
Premature senescence involving p53 and p16 is activated in response to constitutive MEK/MAPK mitogenic signaling
.
Genes Dev
1998
;
12
:
3008
19
.
67.
Haferkamp
S
,
Tran
SL
,
Becker
TM
,
Scurr
LL
,
Kefford
RF
,
Rizos
H
. 
The relative contributions of the p53 and pRb pathways in oncogene-induced melanocyte senescence
.
Aging
2009
;
1
:
542
56
.
68.
Vredeveld
LC
,
Possik
PA
,
Smit
MA
,
Meissl
K
,
Michaloglou
C
,
Horlings
HM
, et al
Abrogation of BRAFV600E-induced senescence by PI3K pathway activation contributes to melanomagenesis
.
Genes Dev
2012
;
26
:
1055
69
.
69.
Kuilman
T
,
Michaloglou
C
,
Mooi
WJ
,
Peeper
DS
. 
The essence of senescence
.
Genes Dev
2010
;
24
:
2463
79
.
70.
Hacker
E
,
Muller
HK
,
Irwin
N
,
Gabrielli
B
,
Lincoln
D
,
Pavey
S
, et al
Spontaneous and UV radiation-induced multiple metastatic melanomas in Cdk4R24C/R24C/TPras mice
.
Cancer Res
2006
;
66
:
2946
52
.
71.
Haferkamp
S
,
Becker
TM
,
Scurr
LL
,
Kefford
RF
,
Rizos
H
. 
p16INK4a-induced senescence is disabled by melanoma-associated mutations
.
Aging Cell
2008
;
7
:
733
45
.
72.
Kalinichenko
VV
,
Major
ML
,
Wang
X
,
Petrovic
V
,
Kuechle
J
,
Yoder
HM
, et al
Foxm1b transcription factor is essential for development of hepatocellular carcinomas and is negatively regulated by the p19ARF tumor suppressor
.
Genes Dev
2004
;
18
:
830
50
.
73.
Alvarez-Fernandez
M
,
Medema
RH
. 
Novel functions of FoxM1: from molecular mechanisms to cancer therapy
.
Front Oncol
2013
;
3
:
30
.
74.
Ventura
A
,
Kirsch
DG
,
McLaughlin
ME
,
Tuveson
DA
,
Grimm
J
,
Lintault
L
, et al
Restoration of p53 function leads to tumour regression in vivo
.
Nature
2007
;
445
:
661
5
.
75.
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
.
76.
Kaplon
J
,
Zheng
L
,
Meissl
K
,
Chaneton
B
,
Selivanov
VA
,
Mackay
G
, et al
A key role for mitochondrial gatekeeper pyruvate dehydrogenase in oncogene-induced senescence
.
Nature
2013
;
498
:
109
12
.
77.
Dadzie
OE
,
Yang
S
,
Emley
A
,
Keady
M
,
Bhawan
J
,
Mahalingam
M
. 
RAS and RAF mutations in banal melanocytic aggregates contiguous with primary cutaneous melanoma: clues to melanomagenesis
.
Br J Dermatol
2009
;
160
:
368
75
.
78.
Chao
SH
,
Price
DH
. 
Flavopiridol inactivates P-TEFb and blocks most RNA polymerase II transcription in vivo
.
J Biol Chem
2001
;
276
:
31793
9
.
79.
Burdette-Radoux
S
,
Tozer
RG
,
Lohmann
RC
,
Quirt
I
,
Ernst
DS
,
Walsh
W
, et al
Phase II trial of flavopiridol, a cyclin dependent kinase inhibitor, in untreated metastatic malignant melanoma
.
Invest New Drugs
2004
;
22
:
315
22
.
80.
Sanchez-Martinez
C
,
Gelbert
LM
,
Shannon
H
,
De Dios
A
,
Staton
B
,
Ajamie
TT
, et al
LY2835219, a potent oral inhibitor of the cyclin-dependent kinases 4 and 6 (CDK4/6) that crosses the blood-brain barrier and demonstrates in vivo activity against intracranial human brain tumor xenografts [abstract]
. In:
Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics
; 
2011 Nov 12–16
;
San Francisco, CA. Philadelphia (PA)
:
AACR
;
Mol Cancer Ther
2011
;
10
(
11 Suppl
):
Abstract nr B234
.
81.
Gelbert
LM
,
Cai
S
,
Lin
X
,
Sanchez-Martinez
C
,
Prado
M
,
Lallena
MJ
, et al
Identification and characterization of LY2835219: A potent oral inhibitor of the cyclin-dependent kinases 4 and 6 (CDK4/6) with broad in vivo antitumor activity [abstract]
. In:
Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics
; 
2011 Nov 12-16
;
San Francisco, CA. Philadelphia (PA)
:
AACR
;
Mol Cancer Ther
2011
;
10
(
11 Suppl
):
Abstract nr B233
.
82.
Joshi
KS
,
Rathos
MJ
,
Joshi
RD
,
Sivakumar
M
,
Mascarenhas
M
,
Kamble
S
, et al
In vitro antitumor properties of a novel cyclin-dependent kinase inhibitor, P276-00
.
Mol Cancer Ther
2007
;
6
:
918
25
.
83.
Joshi
KS
,
Rathos
MJ
,
Mahajan
P
,
Wagh
V
,
Shenoy
S
,
Bhatia
D
, et al
P276-00, a novel cyclin-dependent inhibitor induces G1-G2 arrest, shows antitumor activity on cisplatin-resistant cells and significant in vivo efficacy in tumor models
.
Mol Cancer Ther
2007
;
6
:
926
34
.
84.
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
.
85.
Dean
JL
,
Thangavel
C
,
McClendon
AK
,
Reed
CA
,
Knudsen
ES
. 
Therapeutic CDK4/6 inhibition in breast cancer: key mechanisms of response and failure
.
Oncogene
2010
;
29
:
4018
32
.
86.
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
.
87.
McArthur
GA
,
Young
RJ
,
Sheppard
KE
,
Mar
V
,
Waldeck
K
,
Fox
SB
, et al
Clinical significance of genomic alterations of the CDK4-pathway and sensitivity to the CDK4 inhibitor PD 0332991 in melanoma
.
J Clin Oncol
2012
;
30
:
suppl; abstr 8520
.
88.
Thoms
HC
,
Dunlop
MG
,
Stark
LA
. 
p38-mediated inactivation of cyclin D1/cyclin-dependent kinase 4 stimulates nucleolar translocation of RelA and apoptosis in colorectal cancer cells
.
Cancer Res
2007
;
67
:
1660
9
.
89.
Roberts
PJ
,
Bisi
JE
,
Strum
JC
,
Combest
AJ
,
Darr
DB
,
Usary
JE
, et al
Multiple roles of cyclin-dependent kinase 4/6 inhibitors in cancer therapy
.
J Natl Cancer Inst
2012
;
104
:
476
87
.
90.
Kwong
LN
,
Costello
JC
,
Liu
H
,
Jiang
S
,
Helms
TL
,
Langsdorf
AE
, et al
Oncogenic NRAS signaling differentially regulates survival and proliferation in melanoma
.
Nat Med
2012
;
18
:
1503
10
.
91.
Sutherland
RL
,
Musgrove
EA
. 
CDK inhibitors as potential breast cancer therapeutics: new evidence for enhanced efficacy in ER+ disease
.
Breast Cancer Res
2009
;
11
:
112
.
92.
Finn
RS
,
Crown
JP
,
Boer
K
,
Lang
I
,
Parikh
RJ
,
Breazna
A
, et al
Results of a randomized phase 2 study of PD0332991, a cyclin-dependent kinase (cdk) 4/6 inhibitor, in combination with letrozole vs letrozole alone for first-line treatment of ER+/HER2-advanced breast cancer
.
Ann Oncol
2012
;
23
Suppl 2
:
ii43
ii5
.
93.
Nathanson
KL
,
Martin
A
,
Letrero
R
,
D'Andrea
KP
,
O'Day
S
,
Infante
JR
, et al
Tumor genetic analyses of patients with metastatic melanoma treated with the BRAF inhibitor GSK2118436 (GSK436)
.
J Clin Oncol
2011
;
29
:
8501
.
94.
Sullivan
RJ
,
Flaherty
KT
. 
Resistance to BRAF-targeted therapy in melanoma
.
Eur J Cancer
2013
;
49
:
1297
304
.
95.
Trunzer
K
,
Pavlick
AC
,
Schuchter
L
,
Gonzalez
R
,
McArthur
GA
,
Hutson
TE
, et al
Pharmacodynamic effects and mechanisms of resistance to vemurafenib in patients with metastatic melanoma
.
J Clin Oncol
2013
;
31
:
1767
74
.
96.
Das Thakur
M
,
Salangsang
F
,
Landman
AS
,
Sellers
WR
,
Pryer
NK
,
Levesque
MP
, et al
Modelling vemurafenib resistance in melanoma reveals a strategy to forestall drug resistance
.
Nature
2013
;
494
:
251
5
.
97.
Kovalev
GI
,
Franklin
DS
,
Coffield
VM
,
Xiong
Y
,
Su
L
. 
An important role of CDK inhibitor p18(INK4c) in modulating antigen receptor-mediated T cell proliferation
.
J Immunol
2001
;
167
:
3285
92
.
98.
Sawai
CM
,
Freund
J
,
Oh
P
,
Ndiaye-Lobry
D
,
Bretz
JC
,
Strikoudis
A
, et al
Therapeutic targeting of the cyclin D3:CDK4/6 complex in T cell leukemia
.
Cancer Cell
2012
;
22
:
452
65
.
99.
Hamid
O
,
Robert
C
,
Daud
A
,
Hodi
FS
,
Hwu
WJ
,
Kefford
R
, et al
Safety and tumor responses with lambrolizumab (Anti-PD-1) in melanoma
.
N Engl J Med
2013
;
369
:
134
44
.
100.
Wolchok
JD
,
Kluger
H
,
Callahan
MK
,
Postow
MA
,
Rizvi
NA
,
Lesokhin
AM
, et al
Nivolumab plus ipilimumab in advanced melanoma
.
N Engl J Med
2013
;
369
:
122
33
.