Abstract
CDK4/6 inhibitors are now part of the standard armamentarium for hormone receptor-positive breast cancer. In this article, we review the biologic outcomes imposed by these drugs on cancer cells, determinants of response, mechanisms of intrinsic and acquired resistance, as well as combinatorial approaches emanating from mechanistic studies that may allow use of these agents to extend beyond breast cancer. In addition, we will address tumor-, imaging-, and blood-based pharmacodynamic biomarkers that can inform rationally designed trials as clinical development continues.
CELL CYCLE CONTROL, THE RETINOBLASTOMA PATHWAY, AND THE CANONICAL VIEW OF CDK4/6 INHIBITION
The mitotic cell cycle underlies the division of all somatic cells in the body. Because uncontrolled cell division is a hallmark of cancer, it has been posited that targeting the cell cycle could represent a universal mechanism for cancer therapy.1,2 The signaling pathways that control the cell cycle are complex, but most mitogenic and oncogenic signals act by driving cells through the G0/G1 to S-phase transition (Fig. 1). The key machinery of the cell cycle includes the cyclin-dependent kinases (CDKs) and the associated cyclins required for their catalytic activity.3 Cell cycle traversal during mid-G1 is governed by CDK4 or CDK6 kinase complexes that are highly responsive to a host of cancer-relevant perturbations.4,5 For example, multiple oncogenic factors hijack normal regulation of cyclin D1, which promotes CDK4/6 activity.6 In addition, genetic permutations directly target CDK4/6 activity, including amplification of genes that encode CDK4, CDK6, or cyclin D1, which have been observed in more than 15% of all tumors, as well as deletion, mutation, or methylation of CDKN2A, which encodes the endogenous CDK4/6 inhibitor p16INK4A.4 Thus, much of the oncogenic activity observed in the cell cycle is targeted at net CDK4/6 activity. This is also borne out because the retinoblastoma (RB) tumor suppressor is one of the key substrates of CDK4/67 and is also a commonly mutated tumor suppressor in a fashion that is mutually exclusive with CDK4/6 deregulation.8
FIGURE 1. Canonical G1/S Cell Cycle Control.

Activated retinoblastoma (RB) participates in the repression of an E2F-mediated transcriptional program required for S-phase progression. Mitogenic signals stimulate the assembly of cyclin D-CDK4/6 complexes that initiate RB phosphorylation, followed sequentially by cyclin E-CDK2-mediated phosphorylation that ultimately releases phosphorylated RB and allows E2F-mediated transcription. Cyclin D-CDK4/6 complexes may be inhibited by endogenous inhibitors, such as p16INK4A, or by mimetic drugs, including palbociclib, ribociclib, and abemaciclib.
Abbreviation: RB, retinoblastoma.
The canonical view of the RB pathway is presented in Figure 1, where the activation of CDKs 4 and 6 by multiple mechanisms is required for the initiation of RB phosphorylation. In the absence of phosphorylation, RB, in an activated state, elicits potent transcriptional repression over a gene expression program required for subsequent cell cycle progression.9 This list, which is ever evolving, includes genes required for DNA synthesis, mitosis, and cytokinesis.5 In essence, the RB activation state is a bottleneck for the cell cycle; when RB is activated and unphosphorylated, cells cannot progress through the cell cycle because of the absence of required proteins.
If such a model is accurate, it follows that pharmacologically inhibiting CDK4/6 could have potent effects for blocking tumor cell division.3,4 Although the class of CDK4/6 inhibitors is expanding, palbociclib, ribociclib, and abemaciclib are the only currently clinically approved agents, all of which are highly selective for CDK4 and CDK6 relative to other CDK complexes.3,4 Classically, each of these agents arrests tumor cells in a G1-like state. Concordantly, they generally elicit a cytostatic, as opposed to cytotoxic, mechanism of action in solid tumor cell lines, animal models, and in patient samples.10–17 Ample preclinical data indicate that this activity in restraining cell division is mediated by the activation of RB.13,18,19 This model suggests that CDK4/6 inhibitors should have potent clinical activity against a wide variety of cancers; however, the evaluation of this class of agents in a multitude of tumor types has demonstrated a varying degree of clinical efficacy.3,20 This may be related to diverse biologic outcomes that may occur after CDK4/6 inhibitor-mediated quiescence. Some cell types may more easily adapt than others and readily demonstrate resistance, whereas other cells may convert to a senescent state, resulting in more long-term disease control.21–23
CLINICAL USE OF CDK4/6 INHIBITORS AND BIOMARKERS OF SENSITIVITY
CDK4/6 inhibitors as single agents are particularly effective in suppressing the proliferation of luminal-like breast cancer models.13 In addition, CDK4/6 inhibition can be combined effectively with endocrine therapies used in the management of breast cancer and can clearly exert activity in tumor models resistant to endocrine therapy.13,24,25 This preclinical efficacy has been validated in the clinic, which led to the U.S. Food and Drug Administration approvals for palbociclib, ribociclib, and abemaciclib in combination with endocrine therapy in the context of HR+/HER2− breast cancer,4,20 where they are highly effective in delaying progression of disease. In several cases, these treatments have improved overall survival26–29 compared with endocrine therapy alone. Abemaciclib has also demonstrated monotherapy activity.16,30
Despite these striking clinical results, biomarkers predicting activity of CDK4/6 inhibitors have been difficult to identify, other than preserved expression of the RB protein, a primary CDK4/6 target. In the PALOMA-1 study, the improvement afforded by combined palbociclib and letrozole compared with letrozole alone was similar in unselected patients and in those whose tumors harbored CCND1 amplification or CDKN2A loss.31 However, more recent analyses among a large number of cell lines and preclinical models of varying tumor types suggested that tumors with D-cyclin activating features (DCAF) might be particularly sensitive to CDK4/6 inhibition, including those with 3′UTR alterations that stabilize D-cyclin mRNAs and encoded proteins, as can occur in mantle cell lymphoma (MCL) or endometrial cancer. Other DCAF features include CCND2 or CCND3 amplification, as in multiple hematopoietic malignancies, or less commonly among solid tumors.32,33
CELL CYCLE PLASTICITY AND MECHANISMS OF INTRINSIC AND ACQUIRED RESISTANCE
Loss of RB has emerged as a mechanism of both intrinsic and acquired resistance. At the genetic level, RB1 loss is rare in advanced ER+ breast cancer, occurring in less than 10% of patients. Although a rare event, in one report, in 9 of 338 patients with RB1 loss who were subsequently treated with CDK4/6 inhibitors, progression-free survival (PFS) was only 3.6 months compared with 10.1 months for patients with intact RB1.34 Similarly, RB1 mutations have been reported in samples obtained from patients after development of acquired resistance to CDK4/6 inhibitors.35
However, RB1 mutation or loss does not account for most of the acquired resistance observed to date in breast cancer; other mechanisms must govern the evolution to a presumed CDK4-/6-independent state. In addition, in many other tumor types, it appears that a CDK4-/6-independent state is either preexisting or evolves rapidly,5,36 thereby limiting clinical efficacy of monotherapy. Work in mouse models has provided insight into mechanisms by which CDK4/6 inhibition is bypassed, demonstrating that cells in the mouse can adapt to the loss or reduction of CDK4/6 activity so that other cyclin and/or CDK complexes take their place and mediate the phosphorylation of RB, referred to as “cell cycle plasticity.”37,38
The ability to bypass pharmacologic CDK4/6 inhibition, whether intrinsic or acquired by cell cycle plasticity, occurs through two broad mechanisms. First, high levels of CDK4/6 and D-cyclin complexes can either titrate the pharmacologic inhibitor or escape inhibition and have been described in multiple preclinical and clinical settings. For example, FAT1, a putative tumor suppressor and a member of the cadherin superfamily that interacts with the Hippo signaling pathway, has recently been shown to regulate the expression of CDK6, and its loss may mediate resistance to CDK4/6 inhibitors. Knockout of FAT1 leads to the downregulation of the Hippo pathway and overexpression of CDK6.34 Genetic sequencing of 348 biopsies from patients who were subsequently treated with CDK4/6 inhibitor-based therapy revealed that FAT1 was mutated in approximately 6% of these patients.
Patients with FAT1 mutation had a PFS of only 2.4 months compared with 10.1 months for patients without FAT1 mutations.34 In preclinical models of acquired CDK4/6 inhibitor resistance, both CDK6 amplification39 and CDK6 overexpression via microRNA-mediated modulation of the transforming growth factor beta (TGF-β) pathway have been described; the latter has also been validated in samples from patients whose tumors demonstrated CDK4/6 inhibitor resistance.4 In addition, in KRAS-dependent models of pancreatic and lung cancer, adaptation to CDK4/6 inhibition has resulted in increased functional cyclin-CDK complexes that mediate resistance.41,42
Similarly, upstream alterations that likely modulate cyclin D1-CDK4/6 activity have also been observed among resistant tumors, including those activating the fibroblast growth factor receptor (FGFR) signaling pathway. Next-generation sequencing of circulating tumor DNA from patients enrolled in the MONALEESA-2 trial demonstrated that patients with FGFR1 amplification exhibited a shorter PFS compared with patients with wild-type FGFR1.43 Similarly, following progression on CDK4/6 inhibitors, FGFR1/2 amplification or activating mutations were identified in 14 of 34 (41%) postprogression specimens.43 FGFR-mediated signaling also played a role in resistance in KRAS-dependent models.42 In contrast, the role of mutations in other upstream proteins that could affect cyclin D1-CDK4/6 activity is less clear, including ESR1 and PIK3CA mutations. For example, in the PALOMA-3 study, the phase III clinical trial testing the combination of palbociclib and fulvestrant, there was no correlation between response and PFS and the presence of ESR1 or PIK3CA mutations, which indicated that palbociclib was equally active regardless of mutational status.44,45 Similar data were reported with ribociclib used as first-line therapy in hormone receptor-positive breast cancer (MONALEESA-2).46
Second, in addition to alterations in the expression of the target cyclin-CDK complexes, CDK2 can assume the role of CDK4/6 and therefore render tumor cells resistant.47 Cyclin E, a regulatory subunit of CDK2, is central to the initiation of DNA replication at the G1/S checkpoint. Cyclin E-CDK2 can also phosphorylate RB and can result in the activation of E2F target genes. Because cyclin E-CDK2 phosphorylation events are downstream of those mediated by cyclin D-CDK4/6, overexpression of cyclin E renders inhibition of CDK4/6 ineffective in inducing G1 arrest or subsequent growth inhibition. Hence, tumors with high levels of cyclin E are likely to be intrinsically resistant to CDK4/6 inhibition. Gene expression profiling on 302 archival formalin-fixed, paraffin-embedded samples from the PALOMA 3 trial revealed that in patients treated with combined palbociclib and fulvestrant, those with high cyclin E1 mRNA expression had a median PFS of 7.6 months compared with those with low cyclin E mRNA, who had a PFS of 14.1 months.48
Cyclin E is also post-translationally modified by neutrophil elastase-mediated proteolytic cleavage to generate the low molecular-weight isoforms of cyclin E (LMW-E) that are detected in many cancer types.49 LMW-E lacks the N-terminal cyclin E nuclear localization signal and promotes its accumulation in the cytoplasm. Compared with full-length cyclin E, the aberrant localization and unique stereochemistry of LMW-E dramatically alters the substrate specificity and selectivity of CDK2, increasing tumorigenicity in experimental models. Cytoplasmic LMW-E, which can be assessed by immunohistochemistry, is prognostic of poor survival independent of breast cancer subtype50,51 and also predicts lack of response to neoadjuvant chemotherapy in breast cancer.52 Immunohistochemical analysis of cytoplasmic cyclin E on archival tumor tissue from 109 hormone receptor-positive patients with advanced breast cancer who were treated with palbociclib as first line (+ letrozole) or second line (+ fulvestrant) endocrine therapy revealed that 49.5% of these patients were positive for cyclin E, which was associated with a worse prognosis.53 Specifically, patients with cytoplasmic cyclin E-positive tumors had a median PFS of 13.4 months compared with a PFS of 36.5 months in those with cytoplasmic negative tumors.
GUIDELINES FOR BIOMARKER IDENTIFICATION
As evident by these examples, biomarkers of resistance to CDK4/6 inhibitors are heterogeneous in nature and currently have been primarily examined in pretreatment biopsies, which are largely prognostic. To better develop biomarkers of resistance to CDK4/6 inhibitors, it is critical to first identify if patients are early or late progressors. To this end, it is reasonable to define time to progression in groups of patients based on the historic median PFS of 24.8 months for first-line therapy and 9.5 months for second-line therapy. Progressors can be subdivided into three groups that likely have distinct molecular characteristics (Fig. 2): (1) early progressors (6 months post-therapy for all patients); (2) intermediate progressors (6–24 months post-therapy for first-line therapy patients [Fig. 2A] and 6–9 months post-therapy for second-line therapy patients [Fig. 2B]); and (3) late progressors (. 24 months post-therapy for first-line therapy patients [Fig. 2A] and . 9 months post-therapy for second-line therapy patients [Fig. 2B]). Early progressors are considered to be intrinsically resistant to CDK4/6 inhibitors or endocrine therapy (first or second line) as single agents, whereas intermediate and late progressors are considered to have acquired resistance to the combination of CDK4/6 inhibitors and endocrine therapy. Having distinguished the early from the late progressors, the selected biomarkers must be assessed in matched pretreatment and post-progression biopsy material from these patients. By examining only the pretreatment samples, the biomarkers selected will likely identify the intrinsically resistant patient cohort. However, if postprogression matched samples are also assessed for the same biomarkers, novel insights into the utility of that specific biomarker for potentially predicting acquired resistance to CDK4/6 inhibitor-based therapy can be attained. When matched tumor samples (n = 25) from patients with breast cancer who progressed on palbociclib were examined for deregulation of cyclin E, estrogen receptor, DNA repair, and interleukin-6/STAT3 signaling, results revealed that these pathways were all altered compared with pretreatment tumor samples, which suggested that the alterations of these pathways were specific to palbociclib resistance.54
FIGURE 2. Analysis of Disease Progression.

Analysis of disease progression while on treatment in advanced breast cancer, ER-positive/HER-2 negative patients treated with (A) first-line (CDK4/6 inhibitors + aromatase inhibitor) or (B) second-line (CDK4/6 inhibitors + fulvestrant) therapy. The progressors in each treatment group are divided into intrinsic (early) or acquired (intermediate + late) resistance as described in the text.
Therefore, there is a continued need for development of reliable biomarkers to identify patients with resistance to endocrine therapy and CDK4/6 inhibitors as single agents and in combination that will be clinically meaningful to guide patient care. In addition, exploring mechanisms of disease resistance could considerably improve development of novel treatments effective in delaying or reversing resistance and ultimately improving survival.
COMBINATION THERAPIES TO ENHANCE THE ACTIVITY AND INDICATIONS FOR CDK4/6 INHIBITORS
How cell cycle plasticity is fundamentally controlled or can be predicted is unclear. However, the framework of resistance outlined provides a basis for combination therapy, with the hope of enhancing clinical efficacy of CDK4/6 inhibitors beyond HR+/HER2− breast cancer.
To expand the therapeutic efficacy of CDK4/6 inhibitors, multiple combinatorial approaches have been undertaken and are summarized in Table 1. These efforts largely fall among three different classes of agents: endocrine therapy, kinase inhibitors, and chemotherapy.
TABLE 1.
Representative Combinations of CDK4/6 Inhibitors With Hormonal Agents, Signal Transduction Inhibitors, and Chemotherapy
| Combination | Representative Clinical Studies | NCT Identifier |
|---|---|---|
| + Estrogen inhibition (breast cancer) | FDA-approved | |
| + Estrogen inhibition (gyn malignancies) | Abemaciclib With Letrozole in Recurrent or Persistent Endometrial Cancer | NCT03675893 |
| Ribociclib and Letrozole Treatment in Ovarian Cancer | NCT03673124 | |
| Palbociclib Plus Letrozole Treatment After Progression to Second Line Chemotherapy for Women With ER/PR-positive Ovarian Cancer. (LACOG1018) | NCT03936270 | |
| + Androgen inhibition (prostate cancer) | A Study of Abiraterone Acetate Plus Prednisone With or Without Abemaciclib (LY2835219) in Participants With Prostate Cancer | NCT03706365 |
| Enzalutamide With and Without Ribociclib for Metastatic, Castrate- Resistant, Chemotherapy-Naive Prostate Cancer That Retains RB Expression | NCT02555189 | |
| A Phase II Study of Androgen Deprivation Therapy With or Without Palbociclib in RB-Positive Metastatic Prostate Cancer | NCT02059213 | |
| + MTOR inhibitor | Ribociclib and Everolimus in Treating Children With Recurrent or Refractory Malignant Brain Tumors | NCT03387020 |
| Phase II Trial of Ribociclib and Everolimus in Advanced Dedifferentiated Liposarcoma (DDL) and Leiomyosarcoma (LMS) | NCT03114527 | |
| A Study of LEE011 With Everolimus in Patients With Advanced Neuroendocrine Tumors | NCT03070301 | |
| + MEK inhibitor | Study of Safety and Efficacy of Ribociclib and Trametinib in Patients With Metastatic or Advanced Solid Tumors | NCT02703571 |
| A Phase Ib/II Study of LEE011 in Combination With MEK162 in Patients With NRAS Mutant Melanoma | NCT01781572 | |
| Study of the CDK4/6 Inhibitor Palbociclib (PD-0332991) in Combination With the MEK Inhibitor Binimetinib (MEK162) for Patients With Advanced KRAS Mutant Non-Small Cell Lung Cancer | NCT03170206 | |
| Binimetinib and Palbociclib or TAS-102 in Treating Patients With KRAS and NRAS Mutant Metastatic or Unresectable Colorectal Cancer | NCT03981614 | |
| + PI3K inhibitor | Study of the CDK4/6 Inhibitor Palbociclib (PD-0332991) in Combination With the PI3K/mTOR Inhibitor Gedatolisib (PF-05212384) for Patients With Advanced Squamous Cell Lung, Pancreatic, Head & Neck and Other Solid Tumors | NCT03065062 |
| PIPA: Combination of PI3 Kinase Inhibitors and PAlbociclib (PIPA) | NCT02389842 | |
| + Tyrosine kinase inhibitor | A Study of Abemaciclib in Combination With Sunitinib in Metastatic Renal Cell Carcinoma | NCT03905889 |
| + ALK inhibitor | Study of Safety and Efficacy of LEE011 and Ceritinib in Patients With ALK- positive Nonsmall Cell Lung Cancer. | NCT02292550 |
| + EGFR inhibitor | Study of Safety and Efficacy of LEE011 and Ceritinib in Patients With ALK- positive Nonsmall Cell Lung Cancer. | NCT02292550 |
| Palbociclib and Cetuximab in Metastatic Colorectal Cancer | NCT03446157 | |
| PD 0332991 and Cetuximab in Patients With Incurable SCCHN | NCT02101034 | |
| + Taxane | LEE011 (Ribociclib) in Combination With Docetaxel Plus Prednisone in mCRPC | NCT02494921 |
| Dose-Escalation Study of Palbociclib + Nab-Paclitaxel in mPDAC | NCT02501902 | |
| + Platinum | Palbociclib With Cisplatin or Carboplatin in Advanced Solid Tumors | NCT02897375 |
| Ribociclib (Ribociclib (LEE-011)) With Platinum-based Chemotherapy in Recurrent Platinum Sensitive Ovarian Cancer | NCT03056833 | |
| Phase II Trial Evaluating the Efficacy of Palbociclib in Combination With Carboplatin for the Treatment of Unresectable Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma | NCT03194373 | |
| + Anthracycline | Ribociclib and Doxorubicin in Treating Patients With Metastatic or Advanced Soft Tissue Sarcomas That Cannot Be Removed by Surgery | NCT03009201 |
| + Gemcitabine | Ribociclib and Gemcitabine Hydrochloride in Treating Patients With Advanced Solid Tumors or Lymphoma | NCT02414724 |
| + Combination chemotherapy | PD-0332991, 5-FU, and Oxaliplatin for Advanced Solid Tumor Malignancies | NCT01522989 |
| Study of Palbociclib Combined With Chemotherapy in Pediatric Patients With Recurrent/Refractory Solid Tumors | NCT03709680 |
Abbreviation: FDA, U.S. Food and Drug Administration.
Similar to combined CDK4/6 inhibition and endocrine therapies in breast cancer, it has been shown that CDK4/6 inhibitors will cooperate with androgen-deprivation strategies in preclinical models of prostate cancer55 and with estrogen depletion in specific gynecologic malignancies. Whether there is clinical benefit of the combination of CDK4/6 inhibition and these approaches is being tested in several clinical trials (Table 1).
In addition to endocrine therapy, a host of kinase inhibitors have been shown to positively interact with CDK4/6 inhibitors. In many such contexts, the kinase inhibitor is targeting an oncogenic pathway present in the associated tumor type.
Examples include the combination of EGFR and CDK4/6 inhibition in EGFR-mutant lung cancer or MEK inhibition with CDK4/6 inhibition in a RAS-driven tumor.42,56–58 Considerable preclinical data support a wide spectrum of such combination therapies that either build off U.S. Food and Drug Administration-approved targeted therapies or represent new combination approaches to target a specific cancer (Table 1). Although many of these strategies are strongly supported by preclinical data, emerging clinical results remain uneven and will require randomized studies to clarify the contribution of CDK4/6 inhibition. In addition, toxicity of certain combinations represents a substantial challenge.
Chemotherapy and CDK4/6 inhibition at face value would appear to be antagonistic, because most chemotherapy kills tumor cells based on ongoing cell cycle progression and mitotic division. CDK4/6 inhibition has been shown to antagonize responses to chemotherapy in RB-expressing cells.59,60 However, most of these studies investigated short-term endpoints and did not necessarily model how to best combine the agents for efficacy. A number of recent studies have suggested that CDK4/6 inhibitors could be used metronomically or as maintenance strategies following different chemotherapy regimens.12,61–64 The nature of the positive interaction of chemotherapy and CDK4/6 inhibition is complex. RB regulates the expression of many factors associated with DNA repair or recovery from mitotic stress,9 and therefore, if RB is active, it could prevent recovery from chemotherapy-mediated damage. Conversely, it is well known that chemotherapy will elicit mechanisms to downregulate CDK activity and could therefore cooperate with CDK4/6 inhibition. Irrespective of these mechanisms, clinical studies with combination strategies have produced some degree of efficacy, and multiple trials are ongoing (Table 1).65
NONCANONICAL EFFECTS OF ACTIVATING THE RB-PATHWAY AND NEW VULNERABILITIES WITH CDK4/6 INHIBITION
With the extensive research in the activity of CDK4/6 inhibitors, it is not surprising that new discoveries are being made that could be clinically actionable beyond effects on the cell cycle.5,21,66 Analysis of gene expression data from cells and tumors in which the RB pathway is activated by CDK4/6 inhibition revealed the induction of an immunologic gene expression program, including genes involved in antigen presentation and interferon response, which suggests that the tumor cell may become more susceptible to immunologic surveillance.67–71 Parallel studies demonstrated that systemic CDK4/6 inhibition led to changes in T-cell activity through modulation of NFAT (nuclear factor of activated T cells), which can drive a more potent antitumor response.68 These results have been extended by illustrating cooperation of CDK4/6 inhibition with the immune-checkpoint blockade in a variety of tumor cell types. In addition, correlative analysis from tumors treated with CDK4/6 inhibitors have indicated enhanced immunologic infiltrates.67 These findings, in concert with the overall excitement surrounding immunotherapy, have led to a proliferation of clinical studies combining CDK4/6 inhibitors, alone or in combination with other targeted agents and with immunotherapy (Table 2). Because many of these studies have only recently been initiated, the data on clinical efficacy and tolerability are not yet mature; however, current preliminary findings appear promising.72
TABLE 2.
Representative Clinical Trials Combining CDK4/6 Inhibition With Immune Checkpoint Blockade or With Inhibitors of Cellular Metabolism
| Combination | Representative Clinical Studies | NCT Identifier |
|---|---|---|
| + Immune-checkpoint inhibitor (anti-PD-1/anti-PD-L1) | Abemaciclib and Pembrolizumab in Locally Advanced Unresectable or Metastatic Gastroesophageal Adenocarcinoma: Big Ten Cancer Research Consortium BTCRC-GI18–149 | NCT03997448 |
| Clinical Trial of Abemaciclib in Combination With Pembrolizumab in Patients With Metastatic or Recurrent Head and Neck Cancer | NCT03938337 | |
| Phase Ib Study of TNO155 in Combination With Spartalizumab or Ribociclib in Selected Malignancies | NCT04000529 | |
| Avelumab, Cetuximab, and Palbociclib in Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma | NCT03498378 | |
| Pilot Study of Pembrolizumab Combined With Pemetrexed or Abemaciclib for High Grade Glioma | NCT04220892 | |
| Abemaciclib and Nivolumab for Subjects With Hepatocellular Carcinoma | NCT03781960 | |
| Ribociclib and Spartalizumab in R/M HNSCC (RISE-HN) | NCT04213404 | |
| + Metabolic inhibitors | Hydroxychloroquine, Palbociclib, and Letrozole Before Surgery in Treating Participants With Estrogen Receptor Positive, HER2 Negative Breast Cancer | NCT03774472 |
| A Study of Telaglenastat (CB-839) in Combination With Palbociclib in Patients With Solid Tumors | NCT03965845 |
The RB pathway, in addition to controlling cell cycle progression, also plays a role in coordinating different features of metabolism. Evolutionarily, this makes sense because cellular division is tightly linked to nutrient availability to support the biologic mass of two daughter cells. In the case of CDK4/6 inhibition, there is a shift toward oxidative73–75 or autophagic metabolism.53,76 These findings could be leveraged using selective inhibitors targeting these processes to essentially shift the cytostatic state induced by CDK4/6 inhibit into a selective vulnerability.53 Similarly, because RB activation can limit the expression of multiple DNA repair-associated genes, treatment with CDK4/6 inhibitors could yield a functional state similar to multiple forms of DNA repair deficiency, which ostensibly can be exploited by chemotherapy or radiation therapy. Recent studies have also suggested that CDK4/6 inhibitors could yield sufficient deficiency in homologous recombination-mediated repair to drive cooperative interactions with PARP inhibitors.77 Evaluation of these vulnerabilities are being explored clinically (Table 2).
PHARMACODYNAMIC MONITORING OF CDK4/6 INHIBITION IN CLINICAL TRIALS
As clinical trials of CDK4/6 inhibitors study multiple tumor types in combination with other agents, the assessment of pharmacodynamic markers capable of monitoring target engagement and/or pathway modulation will be critical. Several candidates have been identified for this purpose, including expression of phosphorylated RB (phospho-RB) protein, 3′-deoxy-3′ [18F]-fluorothymidine (FLT)-positron emission tomography (PET) and serum thymidine kinase-1 (TK1) activity.
Clinical studies support the monitoring of phospho-RB for evidence of CDK4/6 inhibition. This was initially performed in a pharmacodynamic study of palbociclib in MCL, which demonstrated an 89% reduction in phospho-RB in on-treatment biopsies taken after 3 weeks of palbociclib treatment (125 mg daily for 3 of 4 weeks), compared with pretreatment, with no corresponding changes in total RB.15 There was a concurrent reduction in the proliferative marker Ki-67 that correlated with the degree of phospho-RB reduction and that confirmed phospho-RB as a potential marker for monitoring CDK4/6 inhibitor-mediated antiproliferative responses. However, further correlative analyses indicated that reductions in Ki-67 and phospho-RB were necessary but not sufficient to predict long-term clinical benefit in heavily pretreated patients with MCL, likely because of subsequent development of resistance in a subset of patients that limited the use of phospho-RB early after treatment initiation as a predictive biomarker. However, the absence of reduced RB phosphorylation early after the start of treatment may correlate with ultimate lack of response. For example, in a short-term preoperative breast cancer trial with palbociclib as a single agent, nonresponders (measured by change in Ki67) were characterized by no change in levels of phospho-RB.78 Similar results were seen in the initial trial of abemaciclib monotherapy, in which there was a significant correlation between clinical efficacy and modulation of phospho-RB in proliferating keratinocytes and tumor biopsies. Here, a 60% phospho-RB reduction threshold was able to separate most patients with stable disease or response from those who progressed.16
The role of phospho-RB as a marker for CDK4/6 inhibitor activity was also assessed in a randomized phase II study of palbociclib (125 or 100 mg) in combination with either fulvestrant or tamoxifen in metastatic hormone receptor-positive breast cancer (TBCRC 035).79 Findings were in line with those observed in patients with MCL; there was a considerable decrease in phospho-RB and Ki-67 staining observed in both skin and tumor biopsies. The degree of change was comparable between the 125- and 100-mg palbociclib dose levels, as was clinical outcome, which indicated that dose reduction of palbociclib, if required for hematologic toxicity, was unlikely to compromise pharmacodynamic effects and efficacy signals. In another phase I clinical trial of palbociclib and the MEK inhibitor PD-0325901 in RAS-mutant solid tumors that compared continuous and intermittent (3 weeks of every 4 weeks [3/1 dosing]) palbociclib dosing, serial analysis of skin keratinocytes during the first treatment cycle demonstrated that phospho-RB and Ki-67 staining mirrored palbociclib exposure. There was a sustained decrease in patients who underwent continuous dosing, whereas intermittent dosing resulted in a recovery of phospho-RB staining during the off week of palbociclib.80
The ability to use noninvasive or minimally invasive biomarkers is preferential to biopsy-based biomarkers, and [18F] FLT identifies cells undergoing active DNA synthesis, which allows for its potential use as a surrogate marker for S-phase and the quantification of cellular proliferation.81 The MCL study also examined the role of pre- and on-treatment [18F] FLT-PET as an imaging biomarker to demonstrate the biologic effect of G1 arrest mediated by palbociclib.15 Of 16 evaluable patients, there were significant reductions in the summed [18F] FLT-PET maximal standard uptake value in most patients, and in patients with PFS longer than 1 year (n = 5, including 1 patient with complete response and 2 patients with partial responses), there was a more than 70% reduction in summed [18F] FLT maximal standard uptake value. However, correlative analysis with clinical response indicated that as with phospho-RB and Ki-67 reductions, the reduction in [18F] FLT uptake was necessary but not sufficient for predicting long-term survival benefit.
TK1 is an enzyme critical for DNA synthesis and is expressed in all human cells during normal cell division through E2F-dependent transcription, with small amounts found in serum. Tumors, because of their higher replication rates compared with normal tissues, are capable of secreting pathologic levels of TK1 detectable in serum. Serum TK1 activity can be measured in patients with cancer using the DiviTum Assay (Biovica, Uppsala, Sweden). This is an enzyme-linked immunosorbent- based assay in which BrdU (5-bromo-2′-deoxyuridine) is incorporated into a synthetic DNA strand fixed to the assay plate, with the extent of BrdU incorporation reflecting TK1 activity present in the serum (Fig. 3). In breast cancer, serum TK1 activity was associated with advanced stage, higher grade, tumor necrosis, vascularity, and estrogen receptor and progesterone receptor negativity.82 TK1 activity was also elevated in patients with BRCA1/2 mutations compared with wild-type and was an independent predictor of disease recurrence.82 In addition, TK1 activity levels were determined in patients with advanced breast cancer before they underwent combination chemotherapy with either epirubicin/paclitaxel or epirubicin/paclitaxel/capecitabine using the DiviTum assay. Analysis then identified TK1 activity to be predictive for PFS and overall survival, as well as response to treatment.83 Similarly, patients with hormone receptor-positive breast cancer with low baseline levels of serum TK1 activity or an early drop in TK1 activity had improved outcomes in response to endocrine therapy.84
FIGURE 3. Liquid Biopsy for G1 arrest (Biovica DiviTum Assay).

TK phosphorylates the nucleoside analog BrdU (5-bromo-2′-deoxyuridine) to BrdUMP (5-bromo-2′-deoxyuridine 5′-monophosphate), which is further phosphorylated to BrdUTP (5-bromo-2′-deoxyuridine 5′-triphosphate). BrdUTP is then incorporated into a solid-phase DNA-strand and incorporated BrdU detected using an anti-BrdU monoclonal antibody conjugated to the signal generating enzyme alkaline phosphatase. The level of BrdU incorporated over time is proportional to the level of TK activity in the sample. Reproduced with permission from Biovica.com/Divitum.
Abbreviation: TK, thymidine kinase.
Because TK1 expression is E2F-dependent, CDK4/6 inhibitor treatment should result in reduced serum TK1 activity levels. Coupled with its demonstrated prognostic value in early-stage breast cancer, serum TK1 activity may act as a potential biomarker for target engagement and clinical benefit following CDK4/6 inhibitor treatment. This was examined as part of the NeoPalAna trial14 that examined neoadjuvant palbociclib and anastrazole in early-stage hormone receptor-positive breast cancer, in which serum TK1 activity was serially monitored until the time of surgery using the DiviTum assay.85 Patients received an initial 4 weeks of anastrozole, followed by palbociclib on cycle 1, day 1 (C1D1) for four 28-day cycles. Surgery occurred following 3–5 weeks of washout from the last dose of palbociclib, except in a small subset of patients who received palbociclib (cycle 5) continuously until surgery. Serum TK1 activity was determined at baseline, on C1D1, on C1D15, and at time of surgery. Despite a considerable drop in tumor Ki-67 with anastrozole monotherapy, there was no statistically significant change in TK1 activity. However, a striking reduction in TK1 activity was observed 2 weeks after initiation of palbociclib (C1D15), which then rose significantly with palbociclib washout. At C1D15, TK1 activity was below the detection limit in more than 90% of patients, indicating a profound effect of CDK4/6 inhibition. There was high concordance between changes in serum TK1 and tumor Ki-67 and TK1 mRNA levels in the same direction from C1D1 to C1D15 and from C1D15 to surgery time points. Similar findings were demonstrated in the phase I study of palbociclib and the MEK inhibitor PD-0325901, which compared two different palbociclib dosing schedules—continuous and 3 weeks of every 4 weeks (3/1 dosing).80 Serial measurements of TK activity during the first treatment cycle demonstrated that serum TK activity mirrored palbociclib administration; in patients who received 3/1 palbociclib dosing serum, TK1 activity decreased for the 3 weeks of palbociclib exposure, with subsequent increases during the off week. In patients treated with continuous palbociclib, TK activity remained low throughout the cycle. In addition, TK activity profiles also reflected those seen for phospho-RB and Ki-67 staining in keratinocytes.
These preliminary results highlight the potential for serum TK1 activity to act as a noninvasive biomarker for CDK4/6 inhibitor target engagement. This was further examined in 46 patients enrolled in the TREnd study, a phase II trial that tested the activity and safety of single-agent palbociclib against palbociclib combined with the endocrine therapy on which patients had progressed most recently before enrollment. Although baseline TK1 activity was not prognostic, after one cycle of treatment, patients who demonstrated an increase in TK1 activity had a worse outcome compared with those in which activity was decreased or stable, with a median PFS of 3.0 months versus 9.0 months, respectively. TK1 activity was also assayed at the time of resistance; those with activity higher than the median fared worse on poststudy treatment compared with those with lower activity.86 This work suggested that an initial rise in TK1 activity correlated with intrinsic drug resistance and that a rise after an initial reduction correlated with acquired resistance that could be stratified prognostically.
Further work will be required to fully define the degree of reduction in TK1 activity necessary to predict response or long-term clinical benefit. Identification of this threshold is particularly important in combination therapy studies in which attenuation of CDK4/6 inhibitor dosing may be required. In addition, further studies with more intensive serial sampling will be required to determine whether a rise in TK1 activity that indicates acquired resistance predates resistance defined radiographically. Finally, to date, most studies using serum TK1 activity have focused on palbociclib, and additional studies of the effects of abemaciclib and ribociclib on this marker are warranted.
FUTURE PROSPECTS
With intense ongoing research, it is expected that more therapeutic combinatorial strategies will emerge from the preclinical arena. Rationally developed clinical trials that incorporate biomarkers for patient selection, assess tumors at the time of resistance, and incorporate pharmacodynamic endpoints will be crucial to fully leverage the promise of targeting the RB pathway in cancers.
PRACTICAL APPLICATIONS.
Selective CDK4/6 inhibitors typically lead to G1 arrest and/or senescence against solid tumors and have demonstrated meaningful clinical and survival benefit in combination with hormonal treatment in estrogen receptor-positive breast cancer.
Multiple intrinsic and acquired resistance mechanisms have been described, including loss of RB, elevated CDK6 activity, FGFR pathway activation, and cyclin E-CDK2 activation.
Analysis of matched pre- and post-progression biopsies among early and late progressors will help define biomarkers of intrinsic and acquired resistance that will ultimately guide development of rational combinations relevant to breast cancer and other tumor types.
Pharmacodynamic markers of selective CDK4/6 inhibition include reduced phosphorylation of RB, as well as reduced fluorothymidine-positron emission tomography uptake and reduced serum thymidine kinase 1 activity as noninvasive markers of G1 arrest that can confirm target engagement in clinical trials.
Footnotes
AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST AND DATA AVAILABILITY STATEMENT
Disclosures provided by the authors and data availability statement (if applicable) are available with this article at DOI https://doi.org/10.1200/EDBK_281085.
REFERENCES
- 1.Sherr CJ. Cancer cell cycles. Science. 1996;274:1672–1677. [DOI] [PubMed] [Google Scholar]
- 2.Shapiro GI. Cyclin-dependent kinase pathways as targets for cancer treatment. J Clin Oncol. 2006;24:1770–1783. [DOI] [PubMed] [Google Scholar]
- 3.Asghar U, Witkiewicz AK, Turner NC, et al. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat Rev Drug Discov. 2015; 14:130–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Sherr CJ, Beach D, Shapiro GI. Targeting CDK4 and CDK6: from discovery to therapy. Cancer Discov. 2016;6:353–367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Knudsen ES, Pruitt SC, Hershberger PA, et al. Cell cycle and beyond: exploiting new RB1 controlled mechanisms for cancer therapy. Trends Cancer. 2019; 5:308–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Diehl JA. Cycling to cancer with cyclin D1. Cancer Biol Ther. 2002;1:226–231. [DOI] [PubMed] [Google Scholar]
- 7.Matsushime H, Ewen ME, Strom DK, et al. Identification and properties of an atypical catalytic subunit (p34PSK-J3/cdk4) for mammalian D type G1 cyclins. Cell. 1992;71:323–334. [DOI] [PubMed] [Google Scholar]
- 8.Palmero I, Peters G. Perturbation of cell cycle regulators in human cancer. Cancer Surv. 1996;27:351–367. [PubMed] [Google Scholar]
- 9.Cam H, Dynlacht BD. Emerging roles for E2F: beyond the G1/S transition and DNA replication. Cancer Cell. 2003;3:311–316. [DOI] [PubMed] [Google Scholar]
- 10.Toogood PL, Harvey PJ, Repine JT, et al. Discovery of a potent and selective inhibitor of cyclin-dependent kinase 4/6. J Med Chem. 2005; 48:2388–2406. [DOI] [PubMed] [Google Scholar]
- 11.Fry DW, Harvey PJ, Keller PR, 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;3:1427–1438. [PubMed] [Google Scholar]
- 12.Gelbert LM, Cai S, Lin X, et al. Preclinical characterization of the CDK4/6 inhibitor LY2835219: in-vivo cell cycle-dependent/independent anti-tumor activities alone/in combination with gemcitabine. Invest New Drugs. 2014;32:825–837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Finn RS, Dering J, Conklin D, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Ma CX, Gao F, Luo J, et al. NeoPalAna: neoadjuvant palbociclib, a cyclin-dependent kinase 4/6 inhibitor, and anastrozole for clinical stage 2 or 3 estrogen receptor-positive breast cancer. Clin Cancer Res. 2017;23:4055–4065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Leonard JP, LaCasce AS, Smith MR, et al. Selective CDK4/6 inhibition with tumor responses by PD0332991 in patients with mantle cell lymphoma. Blood. 2012; 119:4597–4607. [DOI] [PubMed] [Google Scholar]
- 16.Patnaik A, Rosen LS, Tolaney SM, et al. Efficacy and safety of abemaciclib, an inhibitor of CDK4 and CDK6, for patients with breast cancer, non-small cell lung cancer, and other solid tumors. Cancer Discov. 2016;6:740–753. [DOI] [PubMed] [Google Scholar]
- 17.Infante JR, Cassier PA, Gerecitano JF, 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;22:5696–5705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Konecny GE, Winterhoff B, Kolarova T, et al. Expression of p16 and retinoblastoma determines response to CDK4/6 inhibition in ovarian cancer. Clin Cancer Res. 2011;17:1591–1602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Dean JL, Thangavel C, McClendon AK, et al. Therapeutic CDK4/6 inhibition in breast cancer: key mechanisms of response and failure. Oncogene. 2010; 29:4018–4032. [DOI] [PubMed] [Google Scholar]
- 20.O’Leary B, Finn RS, Turner NC. Treating cancer with selective CDK4/6 inhibitors. Nat Rev Clin Oncol. 2016;13:417–430. [DOI] [PubMed] [Google Scholar]
- 21.Klein ME, Kovatcheva M, Davis LE, et al. CDK4/6 inhibitors: the mechanism of action may not be as simple as once thought. Cancer Cell. 2018;34:9–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kovatcheva M, Liao W, Klein ME, et al. ATRX is a regulator of therapy induced senescence in human cells. Nat Commun. 2017;8:386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kovatcheva M, Liu DD, Dickson MA, et al. MDM2 turnover and expression of ATRX determine the choice between quiescence and senescence in response to CDK4 inhibition. Oncotarget. 2015;6:8226–8243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Thangavel C, Dean JL, Ertel A, et al. Therapeutically activating RB: reestablishing cell cycle control in endocrine therapy-resistant breast cancer. Endocr Relat Cancer. 2011;18:333–345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Miller TW, Balko JM, Fox EM, et al. ERα-dependent E2F transcription can mediate resistance to estrogen deprivation in human breast cancer. Cancer Discov. 2011;1:338–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Turner NC, Slamon DJ, Ro J, et al. Overall survival with palbociclib and fulvestrant in advanced breast cancer. N Engl J Med. 2018;379:1926–1936. [DOI] [PubMed] [Google Scholar]
- 27.Sledge GW Jr., Toi M, Neven P, et al. The effect of abemaciclib plus fulvestrant on overall survival in hormone receptor-positive, ERBB2-negative breast cancer that progressed on endocrine therapy-MONARCH 2: a randomized clinical trial. JAMA Oncol. 2019;6:116–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Im SA, Lu YS, Bardia A, et al. Overall survival with ribociclib plus endocrine therapy in breast cancer. N Engl J Med. 2019;381:307–316. [DOI] [PubMed] [Google Scholar]
- 29.Slamon DJ, Neven P, Chia S, et al. LBA7_PR - Overall survival results of the Phase III MONALEESA-3 trial of postmenopausal patients with hormone receptor-positive human epidermal growth factor 2-negative advanced breast cancer treated with fulvestrant 6 ribociclib. Ann Oncol. 2019;30 (Suppl_5):856–857. [Google Scholar]
- 30.Dickler MN, Tolaney SM, Rugo HS, 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 [published correction appears in Clin Cancer Res. 2018;24:5485]. Clin Cancer Res. 2017;23:5218–5224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Finn RS, Crown JP, Lang I, 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. [DOI] [PubMed] [Google Scholar]
- 32.Shapiro GI. Genomic biomarkers predicting response to selective CDK4/6 inhibition: progress in an elusive search. Cancer Cell. 2017;32:721–723. [DOI] [PubMed] [Google Scholar]
- 33.Gong X, Litchfield LM, Webster Y, et al. Genomic aberrations that activate D-type cyclins are associated with enhanced sensitivity to the CDK4 and CDK6 inhibitor abemaciclib. Cancer Cell. 2017;32:761–776.e6. [DOI] [PubMed] [Google Scholar]
- 34.Li Z, Razavi P, Li Q, et al. Loss of the FAT1 tumor suppressor promotes resistance to CDK4/6 inhibitors via the Hippo pathway. Cancer Cell. 2018;34:893–905.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Condorelli R, Spring L, O’Shaughnessy J, et al. Polyclonal RB1 mutations and acquired resistance to CDK 4/6 inhibitors in patients with metastatic breast cancer. Ann Oncol. 2018;29:640–645. [DOI] [PubMed] [Google Scholar]
- 36.Knudsen ES, Witkiewicz AK. The strange case of CDK4/6 inhibitors: mechanisms, resistance, and combination strategies. Trends Cancer. 2017;3:39–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Malumbres M, Sotillo R, Santamaría D, et al. Mammalian cells cycle without the D-type cyclin-dependent kinases Cdk4 and Cdk6. Cell. 2004;118:493–504. [DOI] [PubMed] [Google Scholar]
- 38.Kozar K, Sicinski P. Cell cycle progression without cyclin D-CDK4 and cyclin D-CDK6 complexes. Cell Cycle. 2005;4:388–391. [DOI] [PubMed] [Google Scholar]
- 39.Yang C, Li Z, Bhatt T, et al. Acquired CDK6 amplification promotes breast cancer resistance to CDK4/6 inhibitors and loss of ER signaling and dependence. Oncogene. 2017;36:2255–2264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Cornell L, Wander SA, Visal T, et al. MicroRNA-mediated suppression of the TGF-beta pathway confers transmissible and reversible CDK4/6 inhibitor resistance. Cell Rep. 2019;26:2667–2680.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Knudsen ES, Kumarasamy V, Ruiz A, et al. Cell cycle plasticity driven by MTOR signaling: integral resistance to CDK4/6 inhibition in patient-derived models of pancreatic cancer. Oncogene. 2019;38:3355–3370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Haines E, Chen T, Kommajosyula N, et al. Palbociclib resistance confers dependence on an FGFR-MAP kinase-mTOR-driven pathway in KRAS-mutant non-small cell lung cancer. Oncotarget. 2018;9:31572–31589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Formisano L, Lu Y, Servetto A, et al. Aberrant FGFR signaling mediates resistance to CDK4/6 inhibitors in ER+ breast cancer. Nat Commun. 2019;10:1373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Cristofanilli M, Turner NC, Bondarenko I, 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 [published correction appears in Lancet Oncol. 2016;e136; e270]. Lancet Oncol. 2016;17:425–439. [DOI] [PubMed] [Google Scholar]
- 45.Fribbens C, O’Leary B, Kilburn L, et al. Plasma ESR1 mutations and the treatment of estrogen receptor-positive advanced breast cancer. J Clin Oncol. 2016; 34:2961–2968. [DOI] [PubMed] [Google Scholar]
- 46.Hortobagyi GN, Stemmer SM, Burris HA, et al. Ribociclib as first-line therapy for HR-positive, advanced breast cancer. N Engl J Med. 2016;375:1738–1748. [DOI] [PubMed] [Google Scholar]
- 47.Herrera-Abreu MT, Palafox M, Asghar U, et al. Early adaptation and acquired resistance to CDK4/6 inhibition in estrogen receptor-positive breast cancer. Cancer Res. 2016;76:2301–2313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Turner NC, Liu Y, Zhu Z, et al. Cyclin E1 expression and palbociclib efficacy in previously treated hormone receptor-positive metastatic breast cancer. J Clin Oncol. 2019;37:1169–1178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Caruso JA, Duong MT, Carey JPW, et al. Low-molecular-weight cyclin E in human cancer: cellular consequences and opportunities for targeted therapies. Cancer Res. 2018;78:5481–5491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Hunt KK, Karakas C, Ha MJ, et al. Cytoplasmic cyclin E predicts recurrence in patients with breast cancer. Clin Cancer Res. 2017;23:2991–3002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Karakas C, Biernacka A, Bui T, et al. Cytoplasmic cyclin E and phospho-cyclin-dependent kinase 2 are biomarkers of aggressive breast cancer. Am J Pathol. 2016;186:1900–1912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Karakas C, Francis AM, Ha MJ, et al. Cytoplasmic cyclin E expression predicts for response to neoadjuvant chemotherapy in breast cancer. Ann Surg. Epub 2019. August 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Vijayaraghavan S, Karakas C, Doostan I, et al. CDK4/6 and autophagy inhibitors synergistically induce senescence in Rb positive cytoplasmic cyclin E negative cancers. Nat Commun. 2017;8:15916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kettner NM, Vijayaraghavan S, Durak MG, et al. Combined inhibition of STAT3 and DNA repair in palbociclib-resistant ER-positive breast cancer. Clin Cancer Res. 2019;25:3996–4013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Comstock CE, Augello MA, Goodwin JF, et al. Targeting cell cycle and hormone receptor pathways in cancer. Oncogene. 2013;32:5481–5491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Franco J, Witkiewicz AK, Knudsen ES. CDK4/6 inhibitors have potent activity in combination with pathway selective therapeutic agents in models of pancreatic cancer. Oncotarget. 2014;5:6512–6525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Teh JLF, Cheng PF, Purwin TJ, et al. In Vivo E2F reporting reveals efficacious schedules of MEK1/2-CDK4/6 targeting and mTOR-S6 resistance mechanisms [published correction appears in Cancer Discov. 2018;8:1654]. Cancer Discov. 2018;8:568–581.29496664 [Google Scholar]
- 58.Vora SR, Juric D, Kim N, et al. CDK 4/6 inhibitors sensitize PIK3CA mutant breast cancer to PI3K inhibitors. Cancer Cell. 2014;26:136–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Roberts PJ, Bisi JE, Strum JC, et al. Multiple roles of cyclin-dependent kinase 4/6 inhibitors in cancer therapy. J Natl Cancer Inst. 2012;104:476–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.McClendon AK, Dean JL, Rivadeneira DB, et al. CDK4/6 inhibition antagonizes the cytotoxic response to anthracycline therapy. Cell Cycle. 2012;11:2747–2755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Chou A, Froio D, Nagrial AM, et al. ; Australian Pancreatic Cancer Genome Initiative (APGI). Tailored first-line and second-line CDK4-targeting treatment combinations in mouse models of pancreatic cancer. Gut. 2018;67:2142–2155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Kumarasamy V, Ruiz A, Nambiar R, et al. Chemotherapy impacts on the cellular response to CDK4/6 inhibition: distinct mechanisms of interaction and efficacy in models of pancreatic cancer. Oncogene. 2019;39:1831–1845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Witkiewicz AK, Cox D, Knudsen ES. CDK4/6 inhibition provides a potent adjunct to Her2-targeted therapies in preclinical breast cancer models. Genes Cancer. 2014;5:261–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Cao J, Zhu Z, Wang H, et al. Combining CDK4/6 inhibition with taxanes enhances anti-tumor efficacy by sustained impairment of pRB-E2F pathways in squamous cell lung cancer. Oncogene. 2019;38:4125–4141. [DOI] [PubMed] [Google Scholar]
- 65.Clark AS, McAndrew NP, Troxel A, et al. Combination paclitaxel and palbociclib: results of a phase I trial in advanced breast cancer. Clin Cancer Res. 2019; 25:2072–2079. [DOI] [PubMed] [Google Scholar]
- 66.Chaikovsky AC, Sage J. Beyond the cell cycle: enhancing the immune surveillance of tumors via CDK4/6 inhibition. Mol Cancer Res. 2018;16:1454–1457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Goel S, DeCristo MJ, Watt AC, et al. CDK4/6 inhibition triggers anti-tumour immunity. Nature. 2017;548:471–475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Deng J, Wang ES, Jenkins RW, et al. CDK4/6 inhibition augments antitumor immunity by enhancing T-cell activation. Cancer Discov. 2018;8:216–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Schaer DA, Beckmann RP, Dempsey JA, 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;22:2978–2994. [DOI] [PubMed] [Google Scholar]
- 70.Jerby-Arnon L, Shah P, Cuoco MS, et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell. 2018;175:984–997.e24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Zhang J, Bu X, Wang H, et al. Cyclin D-CDK4 kinase destabilizes PD-L1 via cullin 3-SPOP to control cancer immune surveillance [published correction appears in Nature 2019;571:E10]. Nature. 2018;553:91–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Rugo HS, Kabos P, Dickler MN, et al. Abstract P1-09-01: A phase 1b study of abemaciclib plus pembrolizumab for patients with hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2−) metastatic breast cancer (MBC). Cancer Res. 2018;78:P1-09-01. [Google Scholar]
- 73.Franco J, Balaji U, Freinkman E, et al. Metabolic reprogramming of pancreatic cancer mediated by CDK4/6 inhibition elicits unique vulnerabilities. Cell Rep. 2016;14:979–990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Wang H, Nicolay BN, Chick JM, et al. The metabolic function of cyclin D3-CDK6 kinase in cancer cell survival. Nature. 2017;546:426–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Tarrado-Castellarnau M, de Atauri P, Tarragó-Celada J, et al. De novo MYC addiction as an adaptive response of cancer cells to CDK4/6 inhibition. Mol Syst Biol. 2017;13:940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Booth L, Roberts JL, Rais R, et al. Palbociclib augments neratinib killing of tumor cells that is further enhanced by HDAC inhibition. Cancer Biol Ther. 2019;20:157–168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Yi J, Liu C, Tao Z, et al. MYC status as a determinant of synergistic response to olaparib and palbociclib in ovarian cancer. EBioMedicine. 2019;43:225–237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Arnedos M, Bayar MA, Cheaib B, et al. Modulation of Rb phosphorylation and antiproliferative response to palbociclib: the Preoperative-Palbociclib (POP) randomized clinical trial. Ann Oncol. 2018;29:1755–1762. [DOI] [PubMed] [Google Scholar]
- 79.Rugo HS, Mayer EL, Storniolo AM, et al. Abstract PD2–12: palbociclib in combination with fulvestrant or tamoxifen as treatment for hormone receptor positive (HR+) metastatic breast cancer (MBC) with prior chemotherapy for advanced disease (TBCRC 035) A phase II study with pharmacodynamics markers. Cancer Res. 2019;79:PD2-PD12.31641034 [Google Scholar]
- 80.Shapiro GI, Hilton J, Gandi L, et al. Abstract CT046: phase I dose escalation study of the CDK4/6 inhibitor palbociclib in combination with the MEK inhibitor PD-0325901 in patients with RAS mutant solid tumors. Cancer Res. 2017;77:CT046. [Google Scholar]
- 81.Grierson JR, Schwartz JL, Muzi M, et al. Metabolism of 3′-deoxy-3′-[F-18]fluorothymidine in proliferating A549 cells: validations for positron emission tomography. Nucl Med Biol. 2004;31:829–837. [DOI] [PubMed] [Google Scholar]
- 82.Nisman B, Allweis T, Kaduri L, et al. Serum thymidine kinase 1 activity in breast cancer. Cancer Biomark. 2010;7:65–72. [DOI] [PubMed] [Google Scholar]
- 83.Bjöhle J, Bergqvist J, Gronowitz JS, et al. Serum thymidine kinase activity compared with CA 15–3 in locally advanced and metastatic breast cancer within a randomized trial. Breast Cancer Res Treat. 2013;139:751–758. [DOI] [PubMed] [Google Scholar]
- 84.Bonechi M, Galardi F, Biagioni C, et al. Plasma thymidine kinase-1 activity predicts outcome in patients with hormone receptor positive and HER2 negative metastatic breast cancer treated with endocrine therapy. Oncotarget. 2018;9:16389–16399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Bagegni N, Thomas S, Liu N, et al. Serum thymidine kinase 1 activity as a pharmacodynamic marker of cyclin-dependent kinase 4/6 inhibition in patients with early-stage breast cancer receiving neoadjuvant palbociclib. Breast Cancer Res. 2017;19:123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.McCartney A, Bonechi M, De Luca F, et al. Plasma thymidine kinase activity as a biomarker in patients with luminal metastatic breast cancer treated with palbociclib within the TREnd trial. Clin Cancer Res. Epub 2020 Jan 14. [DOI] [PubMed] [Google Scholar]
