Abstract
Given the pivotal role of CDK4/6 deregulation in the pathogenesis of various malignancies, CDK4/6 inhibitors have been incorporated into cancer treatment protocols. Beyond their antiproliferative effects, these inhibitors exhibit prominent immunomodulatory properties. Preclinical data indicated that CDK4/6 inhibitors enhance the tumor cell antigen presentation capacity and restrict the proliferation of immunosuppressive regulatory T-cells. Furthermore, studies showed that CDK4/6 inhibitors could enhance the development of memory T-cell in vivo in melanoma mouse models and in the clinical setting in breast cancer (BC) patients. In silico analysis identified 225 protein targets for CDK4/6 inhibitors involved in 57 immune-related pathways which further supported the immune-modulatory potential of these inhibitors in innate immunity, cytokine signaling, and adaptive immunity. Furthermore, 24 targets exhibited significantly higher protein expression in immune cell types compared to non-immune cell types, as indicated by data from the Human Protein Atlas. The favorable modulation of T-cell immunity by CDK4/6 inhibitors could complement the antitumor effect of immunotherapies, such as immune checkpoint inhibitors (ICIs), which hamper PD-1 /PD-L1 signaling and result in the reactivation of the antitumor T-cell response. Herein, we explore the clinical portfolio of CDK4/6 inhibitors across various solid malignancies, with emphasis on emerging combinations with ICIs.
Supplementary Information
The online version contains supplementary material available at 10.1007/s12032-025-02996-8.
Keywords: Immunotherapy, Immune checkpoint inhibitors, CDK4/6 inhibitors, Clinical trials, Solid tumors
Introduction
Cyclin-dependent kinases (CDKs) are essential for controlling the progression of the cell cycle, and their activities are modulated by cyclins (which act as activators) or cyclin-dependent kinase inhibitors (CKIs) [1–3]. CDK4 and CDK6 (CDK4/6) are particularly critical due to their fundamental role in the G1/S transition as depicted in Fig. 1 [1]. CDKs are a family of serine/threonine protein kinases, which form heterodimers with their respective regulatory cyclin subunits and phosphorylate specific substrates. In the G1 phase, the substrate for CDK4/6 is retinoblastoma susceptibility protein (Rb), which interacts with the E2F transcriptional family in its hypophosphorylated state, whereby it suppresses the transcription of target genes, and when a cell senses mitogenic signals, CDK4/6 are activated by cyclin D and then phosphorylate Rb, thus relieving the inhibition [2, 3]. Sequential phosphorylation of Rb by CDK4/6 and CDK2 results in E2F activation and transcription initiation of genes required for S-phase progression [1]. Deregulation of cell cycle is a hallmark of cancer. Aberrant activity of cyclin D–CDK4/6 pathway has been described in many types of cancer and it invariably leads to uncontrolled cell proliferation with rapid progression through the cell cycle [4–6]. Because of their key role in the cell cycle regulation and implication in carcinogenesis, CDK4/6 inhibition represents an attractive intervention for cancer management as they prevent the phosphorylation of Rb leading to halting the cell cycle progression [7, 8]. Cyclin-D 1 protein is often overexpressed in 50% of patients with breast cancer [9]. Since CDK4/6 are crucial checkpoints for G1/S transition, specific CDK4/6 inhibitors were approved to tackle these kinases [10]. Abemaciclib, palbociclib, and ribociclib are three oral CDK4/6 inhibitors that successfully secured FDA approval for advanced breast cancer (BC) patients with hormone receptor-positive (HR +) and human epidermal growth factor receptor 2-negative (HER2-) tumor status[9]. Palbociclib and ribociclib were both developed from pyrido [2,3-d] pyrimidin-7-one scaffold. However, abemaciclib was developed from 2-anilino-2,4-pyrimidine-[5-benzimidazole] scaffold [11]. The three inhibitors act on CDK4 and 6. However, abemeciclib inhibits a wider scope of kinases including CDK1,2, 5, 9, 14, 16–18 along with GSK3α/β and PIM1 kinases [11]. The development of CDK4/6 inhibitors represents a key milestone in cancer therapeutics, demonstrating improved overall survival with manageable toxicities [9].
Fig. 1.
Mechanism of cell cycle regulation by Cyclin D–CDK4/6-Rb-E2F axis
It is noteworthy that CDK4/6 inhibitors have the capacity to act beyond the cell cycle through the modulation of cellular energetics and metabolic reprogramming [12]. Studies also reported that CDK4/6 inhibitors can induce senescence, autophagy [13], as well as epigenetic modifications [14]. Interestingly CDK4/6 inhibitors were also reported to exhibit a positive impact within the tumor immune microenvironment by augmenting the antitumor immunity [8].
Studies supported that CDK4/6 inhibitors enhance tumor antigen presentation by macrophages and dendritic cells [8]. Furthermore, they promote inflammatory signaling in tumor cells, stimulate immunogenic cell death, reduce immunosuppressive cell populations (Tregs and myeloid-derived suppressor cells MDSCs). Additionally, they are capable of activating effector T-cells and boost the infiltration of B-lymphocytes and natural killer (NK) cells [15]. The immune-modulatory effects of CDK4/6 inhibitors are depicted in Fig. 2. In this review, we highlight the ongoing clinical studies that explore the development of combinations of CDK4/6 inhibitors with PD-1/PD-L1 immune checkpoint immunotherapies in solid malignancies (Table 1).
Fig. 2.
CDK4/6 inhibitors possess favorable immunomodulatory effects within the tumor microenvironment (TME)
Table 1.
The current clinical trials investigating CDK4/6 inhibitor/Immunotherapy combinations in solid tumors
| Cancer Type | Disease subtype/stage | Drug Combination | Trial Phase | Main Outcomes | Key Toxicities | Ref |
|---|---|---|---|---|---|---|
| Breast Cancer | HR + /HER2- metastatic breast cancer | Abemaciclib + Pembrolizumab ± Anastrozole | Phase Ib | ORR 23.1%-28.6%; DCR 82.1–84.6% | Grade ≥ 3 neutropenia, diarrhea, hepatotoxicity; 2 deaths | [30], NCT02779751 |
| HER2 − MBC and metastatic ovarian cancer | Ribociclib + Spartalizumab + Fulvestrant | Phase Ib | CBR 12.5%; PFS 3.1 mo; OS 6.0 mo in HER2-MBC | Grade ≥ 3 neutropenia (50%), liver enzyme elevation | [37], NCT03294694 | |
| HR + , HER2- MBC | Abemaciclib + Nivolumab + Endocrine Therapy | Phase II | ORR 54.5% (fulv), 20% (letro); DCR 90.9% (fulv), 80% (letro) | Severe immune-related AEs; trial terminated | [31], JapicCTI-194782 | |
| HR+ HER2− MBC | Palbociclib + Avelumab + Fulvestrant | Phase II | Median PFS 8.1 mo vs 4.6 mo without avelumab | Neutropenia (grade 3 to 4), fatigue, anemia, and thrombocytopenia | [32, 33] | |
| Stage IV HR+ HER2− MBC | Palbociclib + Pembrolizumab + Letrozole | Phase I/II | Median PFS 25.2 mo; Median OS 36.9 mo; well tolerated | Grade ≥ 3 neutropenia (83%) and leucopenia (65%) | [34] | |
| HR+ HER2− 1ry BC | Palbociclib + Nivolumab + Anastrozole | Phase Ib/II | Trial terminated early | High prevalence of Grade 3/4 hepatotoxicity | [35], NCT04075604 | |
| HNSCC | Recurrent/ metastatic HNSCC | Ribociclib + Spartalizumab | Phase I | ORR 23%; PFS 2.1 mo | Tolerable | [50], NCT04213404 |
| Palbociclib + Avelumab + Cetuximab | Phase I | ORR by RECIST v1.1 was 42% | Well-tolerated with grade 1 to 2 treatment-related adverse events as rash and fatigue | NCT03498378 | ||
| NSCLC | Stage IV NSCLC | Abemaciclib + Pembrolizumab | Phase Ib | ORR 24% (non-sq), 8% (sq); OS 6–27.8 mo | Grade ≥ 3 toxicities; trial terminated | [62], NCT02779751 |
| Liver cancer | Hepatocellular carcinoma (HCC) | Abemaciclib + Nivolumab | Phase II | Trial terminated prematurely | Safety concerns like sepsis (14.29%) | NCT03781960 |
| Ovarian Cancer | Metastatic ovarian cancer | Ribociclib + Spartalizumab + Fulvestrant | Phase Ib | PR (13.3%) | Hypertransaminasemia,neutropenia, fatigue, and anemia | [37] |
| Melanoma | Metastatic Cutaneous Melanoma | Abemaciclib + Anti-PD-L1 (LY3300054) | Phase Ia/Ib | 1 PR (5%), 10 SD (50%) in concurrent arm | Diarrhea, fatigue; autoimmune hepatitis in lead-in arm | [73] |
| Prostate Cancer | Metastatic castration-resistant prostate cancer | Abemaciclib + Atezolizumab | Phase II | Ongoing | Awaiting results | [79], NCT04751929 |
| Glioblastoma | Recurrent glioblastoma | Abemaciclib + Pembrolizumab | Phase II | Trial halted | Safety concerns | NCT04118036 |
| CRC | Advanced CRC | SHR6390 + Camrelizumab | Phase Ib/II | Ongoing | Awaiting results | NCT03601598 |
| Sarcomas | Soft tissue sarcoma | Palbociclib and pembrolizumab | Phase Ib | Ongoing | Awaiting results | NCT06113809 |
| advanced dedifferentiated liposarcoma | Palbociclib and retifanlimab | Phase II | Ongoing | Awaiting results | NCT04438824 |
Furthermore, we performed an in silico analysis that offers insights into the targets and pathways affected by the specific drug classes to provide more insights on their potential molecular targets. Herein, the protein targets of the four reviewed CDK4/6 inhibitors (abemaciclib, dalpiciclib, palbociclib, and ribociclib) were predicted, along with the biological pathways in which they are involved, as well as their cell-type-specific protein expression. The primary objective was to investigate the role of CDK4/6 inhibitors in immunological pathways for the purpose of gaining insights into their immune-modulatory role when administered in conjunction with immune checkpoint inhibitors. Overall, the data from this analysis provided further support for the favorable immune-modulatory role for CDK4/6 inhibitors, which could be exploited to boost the efficacy of ICIs and improve response rates in cancer patients.
The clinically investigated ICI combinations with CDK4/6 inhibitors in solid tumors
Breast cancer
CDK4/6 kinases are integral to the orchestration of cellular replication in the breast tissue [16, 17]. The upregulated activity of cyclin D–CDK4/6 axis in breast cancer (BC) highlights it as a legitimate therapeutic target [16–18]. Hormone receptor-positive (HR +) BC is more sensitive to CDK4/6 inhibitors given that estrogen boosts CCND1 gene expression which encodes cyclin, contributing to aberrant cell cycle [19–21]. CDK4/6 inhibitors combined with anti-estrogens were approved by the FDA for BC management especially in advanced-stage HR + /HER2- subtype [22–27]. Certain subsets of BC proved to be more enriched with tumor-infiltrating lymphocytes (TILs) [28]. The greater abundance of TILs was linked to better prognosis. [29]. The immune checkpoint PD-L1 is overexpressed in about 20% of BC patients. TNBC and HER2-positive subtypes are known to possess higher PD-L1 expression (33% and 56%) compared to ER + BC (11%) [29].
The combination of pembrolizumab (anti-PD-1 mAb) with chemotherapy has acquired FDA approval for managing inoperable or locally advanced/metastatic PD-L1-positive TNBC. Additionally, pembrolizumab is approved as a monotherapy for early-stage TNBC with high risk of recurrence and for treating metastatic or inoperable breast malignancies that are mismatch repair deficient (dMMR) or have high microsatellite instability (MSI-H) [30]. However, more efforts are needed to amplify the benefit of immunotherapy to a broad range of BC patients.
One of the promising avenues is the integration of CDK4/6 inhibitors with immune checkpoint blockade. The combination of abemaciclib/pembrolizumab ± anastrozole was tested in HR + /HER2- metastatic BC patients in a phase Ib trial (NCT02779751). The 1st cohort consisted of patients who were treatment-naïve to systemic anticancer therapy and received a triple combination of abemaciclib/pembrolizumab/ anastrozole. The 2nd cohort included patients previously treated with one or two lines of chemotherapy. They received abemaciclib/pembrolizumab without anastrazole. Despite that the disease control rate was a bit higher in cohort 1 (84.6 versus 82.1% in cohort 2). The overall response rate (ORR) was much higher in cohort 2 (28.6% vs 23.1% in cohort 1). [30]. Assessment of the safety data showed that cohort 1 experienced an increased prevalence of grade ≥ 3 adverse events (AEs) including neutropenia, diarrhea, and severe alterations in liver function tests. It is noteworthy that treatment-induced AEs resulted in 2 deaths in cohort 1. Patients in both cohorts showed higher incidence of pneumonitis and interstitial lung disease. Given the high incidence of toxicity, the benefit/risk analysis did not advocate further testing of the abemaciclib/pembrolizumab combination for HR + /HER2 − metastatic BC.
Abemaciclib was tested in combination with a different checkpoint inhibitor (nivolumab) and endocrine therapy (fulvestrant or letrozole) through a phase II trial in the same setting of HR + /HER2- metastatic BC [31]. The triple combination showed clinical activity with an ORR of 54.5% with fulvestrant vs. 20% letrozole endocrine therapy. Similarly, the disease control rate (DCR) was higher with fulvestrant vs. letrozole (90.9% vs 80.0%). Despite the clinical activity of the tested combination, the development of severe and sustained immune-related AEs led to study termination. Findings from the two clinical studies imply that combining abemaciclib with immunotherapy may amplify treatment-related toxicities.
Various clinical trials tested the therapeutic benefit of palbociclib combinations with immunotherapy (avelumab/ nivolumab/ pembrolizumab) for breast cancer. Two palbociclib combinations were promising in HR + /HER2- metastatic BC. The randomized phase II trial PACE tested palbociclib/avelumab/fulvestrant combination for HR + /HER2- metastatic BC previously progressed on CDK4/6 inhibitor + aromatase dual therapy. Adding avelumab extended the median PFS to 8.1 vs. 4.6 months without avelumab which encourages further investigation of this triple combination in this population [32, 33]. Palbociclib was investigated in combination with pembrolizumab/letrozole as a front-line therapy in a phase I/II clinical trial for patients with stage-IV HR + /HER2- metastatic BC [34]. The data generated from the trial indicated significant clinical benefits, with 31% of patients achieving a complete response, 25% experiencing a partial recovery, and 31% maintaining a stable disease. The median PFS was 25.2 months, and the median OS was 36.9 months. The combination is tolerable, and grade ≥ 3 AEs included neutropenia (83%) and leucopenia (65%). Additional research is warranted to elucidate the immune-modulatory mechanisms of palbociclib in this therapeutic context [34]. On the other hand, testing palbociclib in combination with nivolumab/ anastrozole in ER + /HER2- primary BC showed disappointing outcomes in the CheckMate 7A8 trial. The data of the early run-in-phase showed a high prevalence of grade 3/4 hepatotoxicity. Therefore, the study was terminated [35].
PAveMenT phase Ib trial (NCT04360941) investigated palbociclib/ avelumab combination in patients with androgen receptor-positive (AR +) TNBC(n = 12) and ER + /HER2- advanced BC(n = 2) [36]. The dose-finding phase investigated two dosing schedules for palbociclib including intermittent dosing (75–125 mg) versus continuous dosing (100 mg). Four out of twelve TNBC patients were AR + , and five out of ten TNBC patients with an adequate amount of tissue samples positive for PD-L1 expression. Notably, one of the PD-L1 + ve patients and, separately, two of the five PD-L1 − ve TNBC cases were AR + . The outcomes showed that all the tested schedules were tolerable. However, the continuous dosing (100 mg) was recommended for the anticipated expansion phase in AR + TNBC because of the rapid proliferation characteristics of TNBC and the greater potential for tumor re-growth with intermittent dosing (NCT04360941).
A different triple combination (Ribociclib/spartalizumab ± fulvestrant) was assessed clinically for HR + /metastatic BC and metastatic ovarian cancer in a phase Ib trial [37]. The clinical outcomes were variable. One breast cancer patient accomplished a partial response (ORR = 6.25%), while another had stable disease lasting over 24 weeks. The clinical benefit rate (CBR) for the group was 12.5% through a median follow-up period of 3.5 months. Notably, both patients who experienced clinical benefit received the triple combination therapy. The median PFS across all patients was 3.1 months, with a median overall survival (OS) of 6.0 months. The trial also highlighted that grade ≥ 3 AEs included neutropenia (50%) and elevated liver transaminases (25%). Despite that the combination of ribociclib/spartalizumab was deemed tolerable with AEs comparable to those observed with monotherapy. Further dose expansion cohorts are currently recruiting patients with HR + metastatic BC and metastatic ovarian cancer [37].
Although trials involving abemaciclib in combination with immunotherapy were discontinued due to heightened toxicity concerns, investigations into palbociclib-based immunotherapy regimens have yielded encouraging outcomes. Similarly, early data from studies evaluating ribociclib with spartalizumab suggest potential therapeutic promise.
Head and neck squamous cell carcinoma (HNSCC)
HNSCC is well suited for ICIs due to abundant evidence demonstrating immune evasion mechanisms that contribute to disease progression. These mechanisms often involve the PD-1/PD-L1 axis. Clinical trials showed the benefit of ICIs (nivolumab and pembrolizumab) in recurrent/metastatic HNSCC patients with better PFS and OS compared to standard therapy (NCT01848834; NCT02358031; NCT02252042; NCT02105636) [38–41]. Therefore, ICIs received FDA approval as a first-line treatment for metastatic and recurrent HNSCC [42]. CCND1 amplification and CDKN2A mutations, and Rb presence are considered as biomarkers to predict the activity of CDK4/6 inhibitors in HPV-negative HNSCC [43–45]. Preclinical studies proved the positive effects of CDK4/6 inhibitors in HNSCC. Abemaciclib suppressed the growth of HNSCC cell lines (OSC-19, FaDu, and YD-10B) and led to cell cycle arrest at G0/G1. The same study also indicated that abemaciclib significantly suppressed the growth of OSC-19 xenografts in mice [46]. Similarly, ribociclib exhibited cytostatic effect in HPV-negative cell lines (Detroit562, Cal27, FaDu, SCC9, SCC15, and SCC25) [47]. In vivo data indicated that ribociclib reduced tumor growth in HNSCC xenograft in mice [48].
Clinical studies specifically focus on HPV-negative patients due to the presence of overactive CDK4/6-Rb pathway; in contrast, HPV-positive HNSCC is characterized by the viral oncoprotein E7, which degrades Rb protein, so the upstream CDK4/6 pathway rendered redundant, making HPV-positive patients resistant to CDK4/6 inhibitors[49]. From an immunotherapy perspective, HPV-positive tumors are typically more immunogenic and demonstrate higher response rates to ICIs immunotherapy. Therefore, clinical studies regarding combination therapy of CDK4/6 inhibitors and ICIs target the HPV-negative patient because they respond well to CDK4/6 inhibitors. Additionally, they can augment their modest response to ICIs [50].
The combination of ribociclib/spartalizumab was examined in a phase I trial in patients with recurrent/metastatic HPV-negative HNSCC [51]. The efficacy was modest with ORR 23% and median PFS of 2.1 months. The safety data showed that the combination is tolerable. On the other hand, a phase I study investigated abemaciclib/ nivolumab combination in patients with recurrent/metastatic HNSCC patients who recurred or progressed within 6 months after platinum-based chemotherapy (NCT03655444). The study was terminated due to the development of severe treatment-related toxicity [52]. Other ongoing clinical trials for ICI and CDK4/6 inhibitors in patients with recurrent/metastatic HNSCC include the combination abemaciclib/pembrolizumab (NCT03938337) and palbociclib/avelumab/cetuximab (NCT03498378).
Lung cancer (non-small cell lung cancer NSCLC)
Cell cycle deregulation and disruption of the CDK/cyclin/Rb pathway are adversely implicated in the pathology of NSCLC [53]. Several clinical trials tested CDK4/6 inhibitors in patients with advanced-stage NSCLC (NCT02154490; NCT01291017; NCT02152631; NCT02079636; NCT01394016; and NCT02411591). Abemaciclib demonstrated better PFS and ORR in a phase III trial concerning patients with stage-IV NSCLC [54]. Additionally, palbociclib exhibited antitumor activity in a phase II study targeting CDKN2A-mutant NSCLC, yielding a median PFS of 8.1 weeks and a median OS of 21.6 weeks [55].
PD-L1 is expressed in 35–95% of NSCLC patients [56], and ICIs are currently approved by the FDA for advanced pretreated NSCLC [57, 58]. Nivolumab showed longer PFS, and OS versus docetaxel in CheckMate 017 and CheckMate 057 clinical trials [59, 60]. A pooled analysis of CheckMate 017 and CheckMate 057 studies revealed that the two-year OS rates with nivolumab compared to docetaxel were 23% versus 8% in squamous NSCLC and 29% versus 16% in non-squamous (NS)-NSCLC [61]. The combination of Abemaciclib/pembrolizumab was evaluated in a phase Ib trial in stage-IV NSCLC patients (NCT02779751) [62]. The study enrolled two cohorts: cohort 1 comprising patients with non-squamous non-small cell lung cancer (NS-NSCLC), and cohort 2 consisting of squamous NSCLC cases. Partial responses were observed in 24% of cohort 1 and 8% of cohort 2. Disease control rates were 56% and 64%, respectively, with median PFS of 7.6 months in cohort 1 and 3.3 months in cohort 2, and median OS of 27.8 and 6.0 months. Despite these promising clinical outcomes, both cohorts experienced a high incidence of grade ≥ 3 adverse events, ultimately leading to trial discontinuation. Interestingly, the same therapeutic combination demonstrated a more favorable safety profile in patients with HR + /HER2 − BC (NCT02079636)[63].
Hepatocellular carcinoma (HCC)
Perturbations of cyclin D-CDK4/6/Rb signaling are frequently reported in HCC [64]. CDK4/6 inhibitors proved to be efficacious for combination with tyrosine kinase inhibitors in the management of HCC [65, 66]. Clinically, palbociclib was tested in a phase II trial in adult patients with advanced HCC who have proved resistant/intolerant of first-line sorafenib. Results showed that palbociclib was well tolerated and has activity against advanced HCC with median OS of 19 weeks, and median time to progression (TTP) of 24 weeks. Safety reports indicated that grade ≥ 3 AEs included neutropenia and thrombocytopenia (NCT01356628) [67].
ICIs demonstrated clinical benefits in patients with advanced HCC. The phase I/II trial CheckMate 040 investigated nivolumab in patients with advanced HCC and Child–Pugh B cirrhosis. Data generated from this study demonstrate clinical efficacy of nivolumab (55% disease control rate) with an acceptable safety profile in patients with HCC with Child–Pugh B status who have mild to moderate impairment of liver function or liver decompensation that limits other treatment options. Furthermore, the efficacy of nivolumab was compared to sorafenib in the multicenter phase III trial CheckMate 459 [68]. Efficacy data indicated that nivolumab was superior to sorafenib in ORR (15% vs 7%) and the median OS (16.4 vs 14.7 months). Nivolumab was well tolerated in patients with advanced HCC. The efficacy of pembrolizumab in advanced-stage pretreated HCC patients was assessed in a phase III trial (KEYNOTE-240) [69]. The results showed that pembrolizumab mitigated the risk of death by 22% and improved PFS compared to placebo in patients with advanced HCC. The ORR was 16.9% in pembrolizumab arm versus 2.2% in placebo arm. The combination of abemaciclib/nivolumab was examined in a phase II clinical trial. However, the study was concluded prematurely due to safety concerns (NCT03781960).
Ovarian cancer
A few preclinical studies investigated co-treatment with ICIs and CDK4/6 inhibitors in ovarian cancer. Zhang et al. studied the combination of abemaciclib/anti-PD-1 antibody in a murine syngeneic ovarian cancer model [70]. Abemaciclib-alone augmented a pro-inflammatory tumor response by increasing the tumor infiltration of immune cells, particularly B-cells and cytotoxic T-cells. While the combination therapy significantly increased the activity of B-cells and cytotoxic T-cells leading to a more profound pro-inflammatory anti-tumor response and consequently a synergistic reduction in tumor volume [70]. Overall, this study proved that the combined treatment strategy of CDK4/6 and PD-1 inhibitors holds great promise and offers potential for treating poorly immune-infiltrated ovarian cancers. There is an ongoing phase Ib clinical trial investigating the combination of ribociclib/spartalizumab ± fulvestrant for patients with HR + metastatic BC or metastatic ovarian cancer [37]. The efficacy data for the ovarian cancer cohort are still anticipated.
Melanoma
Immunotherapies have become central for the treatment of advanced melanoma [71]. Nonetheless, a modest set of studies has combined ICIs with CDK4/6 inhibitors especially after the emergence of refractory cases despite the initial success of immunotherapies in advanced melanoma [72]. Abemaciclib was investigated in combination with anti-PD-L1 antibody (LY3300054) in 24 patients as part of the PACT phase Ia/Ib clinical trial in patients with different types of solid malignancies including 1 melanoma patient [73]. Abemaciclib was given either first (lead-in) followed by LY3300054 in 4 patients, which triggered autoimmune hepatitis, or concurrently with LY3300054 (n = 20) which was better tolerated by patients. The most observed AEs in patients receiving the concurrent therapy were diarrhea (55%) and fatigue (35%). In the lead-in arm, 2 patients experienced stable disease, and one showed disease progression. However, in the concurrent arm, 1 patient had partial response, 10 patients had stable disease, and 6 patients had progressive disease [73]. The findings of this trial suggest that combination dosing strategies require better optimization to improve tolerability. This is illustrated by the adjustment from a lead-in to a concurrent dosing sequence in the combination of abemaciclib with LY3300054. Furthermore, the study provided preliminary evidence of the combination therapy’s durable clinical benefit, with some patients achieving either a partial response or stable disease [73]. However, more clinical studies are needed to test the combination in larger number of melanoma patients to validate its benefit.
Prostate cancer
Similar to estrogen receptors, the androgen receptors regulate the transition from G1 phase to S-phase contributing to cell cycle control. Therefore, CDK4/6 inhibitors are capable of disrupting androgen receptors signaling [74, 75]. Abemaciclib showed cytotoxic and apoptotic activity in vitro against metastatic castration-resistant prostate cancer (mCRPC) androgen receptor (AR) negative PC-3 and AR mutant LNCaP PC cells [76]. Clinical testing of ribociclib in combination with docetaxel in a phase Ib/II clinical study in mCRPC showed clinical benefits observed by radiographic PFS (8.1 months) with acceptable toxicity (NCT02494921) [77]. Reports indicated the upregulated expression of PDL1 by dendritic cells of mCRPC patients progressing on AR antagonists. A phase Ia clinical study was initiated to investigate atezolizumab (anti-PDL1), in patients with mCRPC (NCT01375842) [78]. The outcomes showed that atezolizumab exhibited favorable safety with long-term disease control in heavily pretreated mCRPC patients. The combination of abemaciclib with atezolizumab is being tested in an ongoing clinical study in mCRPC patients with CDK12 loss of function mutation [79]. The outcomes are still anticipated.
Glioblastoma (GBM)
The hallmark of genomic instability in GBM is the perturbation in the cell cycle [80]. Genomic profiling of GBM has characterized alterations in core signaling pathways connected with CDK, such as inactivation of the CDKN2 locus (50%) encoding the CDK4/6 inhibitor p16INK4 and the amplification of CDK4 (13%) and CDK6 (1.5%) [81]. Furthermore, molecular studies have indicated that the CDK4/6-Rb axis is dysregulated in roughly 80% of GBM cases [82]. Therefore, CDK4 and CDK6 are valid targets to be tackled for GBM treatment.
CDK4/6 inhibitors have generally demonstrated positive outcomes in preclinical studies of glioblastoma. However, clinical testing of CDK4/6 inhibitors in glioma patients did not produce promising outcomes [83, 84]. Studies showed that abemaciclib has the capacity to cross the blood–brain barrier achieving cerebrospinal fluid (CSF) concentrations comparable to plasma levels which implies its potential merit for glioma [85]. At present, there are four ongoing clinical trials investigating abemaciclib in patients with GBM (NCT02644460, NCT04074785, NCT04391595, and NCT02981940). INSIGhT phase II trial assessed the efficacy of abemaciclib combination with the standard of care in patients with newly diagnosed GB (NCT02977780). The outcomes showed that abemaciclib enhanced the PFS but not the OS versus the standard of care (radio-chemotherapy). Safety reports indicated that abemaciclib was well tolerated [86]. A phase II clinical trial was initiated to investigate abemaciclib in combination with pembrolizumab in patients with recurrent glioblastoma (NCT04118036). Unfortunately, the trial was halted due to safety issues.
Colorectal cancer (CRC)
CDK4/6 as well as cyclin D1 are typically overexpressed in CRC and are linked to poor prognosis [87]. The expression of cyclin D1 in patients with CRC helps to predict responses to future CDK4/6 inhibitors [88]. Clinical testing of CDK4/6 inhibitors as monotherapy did not produce any positive responses. Dysregulated expression of CCNE, RB1, AKT1, AURKA, and RAS confers resistance against CDK4/6 inhibitors [89]. As a result, the therapeutic potential of CDK4/6 inhibitors has been assessed in CRC in the form of combination with other agents including Raf, and mitogen-activated protein kinase (MAPK) inhibitors and ICIs [90]. Interestingly, KRAS-mutant CRCs were especially responsive to a combination of MAPK and CDK4/6 inhibitors [91].
There is an ongoing Phase Ib/II clinical trial investigating the safety and efficacy of CDK4/6 inhibitor (SHR6390) combined with the anti-PD-1 antibody camrelizumab (SHR-1210) in patients with advanced CRC, HCC or NSCLC(NCT03601598). The 1st phase of the trial is the dose-finding phase. Then, the trial will proceed for the expansion phase. The outcomes of this study are still awaited to evaluate the safety and efficacy of this combination in CRC.
Sarcomas
Sarcomas are rare poorly controlled malignant heterogeneous tumors originating from the connective tissues of bone or soft tissues including fat, muscle, blood vessels, or nerves with altered cell cycle and cyclin D-CDK4/6- Rb pathway. Clinical and preclinical studies showed efficacy of CDK4/6 inhibitors in different types of sarcoma[7]. Advanced soft tissue sarcoma overexpressing CDK4 was targeted by palbociclib in a phase II clinical trial, and the results showed median PFS of 4.2 months, and OS of 12 months[92].
Many clinical studies of different phases targeting different types of sarcoma showed that ICIs like pembrolizumab, Ipilimumab, and others showed wide range of PFS and OS due to the heterogeneity of sarcomas. So combination with CD4/6 inhibitors is a good strategy to improve the response[93]. The combination of CDK4/6 inhibitors and immunotherapy against sarcoma is being studied in two ongoing clinical trials. Palbociclib is combined with retifanlimab (anti-PD1) in a phase II study clinical trial in patients with advanced dedifferentiated liposarcoma in which CDK4 gene is highly amplified (NCT04438824)[94]. Palbociclib and pembrolizumab are being investigated in an ongoing phase Ib clinical trial for patients with sarcoma (NCT06113809).
In silico investigation of CDK4/6 inhibitors, drug targets, and pathways
In silico methods
The SwissTargetPrediction (http://www.swisstargetprediction.ch/) was used to identify the protein targets of four CDK4/6 inhibitors: abemaciclib, dalpiciclib, palbociclib, and ribociclib. SwissTargetPrediction is a web-based tool that predicts human protein targets for small molecules by analyzing their 2D and 3D structural similarities to known ligands [95]. It requires a SMILES (Simplified Molecular Input Line Entry System) input and provides a ranked list of protein targets with associated probability scores. Predictions with a confidence score of zero were excluded. The predicted targets for all four inhibitors were merged, which served as the basis for subsequent pathway enrichment analysis.
Cytoscape (version 3.9.1) [96], a platform for network visualization and functional analysis, was utilized to explore the biological pathways enriched among the CDK4/6 inhibitor targets and to gain insight into their potential biological regulations. The ReactomeFIViz plugin [97] was used to perform Voronoi tessellation-based FoamTree clustering, which enables system-level interpretation of functionally grouped biological pathways. In addition, Gene Ontology (GO) Biological Processes (BPs) were identified using the g:Profiler web tool (https://biit.cs.ut.ee/gprofiler/gost), which performs functional enrichment analysis by mapping input genes to GO terms and other biological annotations.
To study the protein expression of the identified CDK4/6 inhibitor targets across different tissues and cell types, the Immunohistochemistry (IHC) data from the Human Protein Atlas (HPA, version 24.0) [98] were retrieved. The HPA IHC dataset includes protein profiles from 45 healthy human tissues, determined by immunohistochemistry using tissue microarrays. It contains Ensembl gene IDs, tissue and cell type annotations, expression levels (categorized as “Not detected,” “Low,” “Medium,” or “High”), and measurement reliability (classified as “Approved,” “Enhanced,” “Supported,” or “Uncertain”).
Data labeled "Uncertain" were excluded from analysis. For quantitative comparison, expression levels were converted to ordinal values: Not detected = 0, Low = 1, Medium = 2, High = 3. A Mann–Whitney U test was conducted to assess the differences in protein expression between immune and non-immune cell types for each target. Cell type annotation included 127 cell types, including immune cell types such as lymphoid tissue, macrophages, hematopoietic cells, germinal center cells, non-germinal center cells, Hofbauer cells, Langerhans cells, lymphocytes, mucosal lymphoid cells, Paneth cells, medullary cells, and cells in the red and white pulp. To correct for multiple comparisons, the Benjamini–Hochberg correction was applied to p values. A schematic summary of the analysis pipeline is provided in Fig. 3.
Fig. 3.
Schematic overview of the methodology for the in silico analysis
In silico results
Protein target prediction resulted in a total of 225 protein targets (Supplementary Table S1), of which the top 3 categories are kinases (45.8%), family A G protein-coupled receptors (14.2%), and proteases (11.6%), as shown in Fig. 4. Functional enrichment analysis using the ReactomeFIViz plugin in Cytoscape identified 310 significantly enriched Reactome pathways (FDR < 0.05, Supplementary Table S2), the majority of which were related to signal transduction, cell cycle regulation, and the immune system, as visualized through a Voronoi FoamTree clustering (Fig. 5). These findings highlight the central involvement of CDK4/6 inhibitor targets in core cellular processes as well as immunological functions.
Fig. 4.
Distribution of protein targets of the four CDK4/6 inhibitors. This diagram demonstrates the categorization of the 225 predicted targets, showing kinases (45.8%), family A G protein-coupled receptors (14.2%), and proteases (11.6%) as the top three categories
Fig. 5.
The Voronoi treemap of the significantly enriched Reactome pathways influenced by 225 CDK4/6 inhibitor protein targets
Given the growing interest in combining CDK4/6 inhibitors with immune checkpoint blockades, we focused on exploring the immune-modulatory potential of these targets. Fifty-seven immune-related enriched pathways were identified and categorized into three categories—innate immunity (e.g., TRIF(TICAM1)-mediated TLR4 signaling and MAP kinase activation), cytokine signaling (e.g., Interleukin-17 signaling and FLT3 Signaling), and adaptive immunity (e.g., CD28 co-stimulation and CTLA4 inhibitory signaling). A summary of these pathways is shown in Table 2.
Table 2.
Summary of Reactome’s immune-related enriched pathways, categorized into three categories: innate immunity, cytokine signaling, and adaptive immunity
| Cytokine Signaling | Adaptive Immunity | |||
|---|---|---|---|---|
| Pathway | FDR | Pathway | FDR | |
| Cytokine Signaling in Immune system | 5.83E-09 | CD28 co-stimulation | 0.012 | |
| Signaling by Interleukins | 6.96E-09 | CTLA4 inhibitory signaling | 0.02 | |
| Interleukin-17 signaling | 3.20E-07 | Activation of NF-kappaB in B-cells | 0.023 | |
| Regulation of signaling by CBL | 1.75E-04 | CD28 dependent PI3K/Akt signaling | 0.029 | |
| Signaling by CSF1 (M-CSF) in myeloid cells | 1.97E-04 | Downstream TCR signaling | 0.034 | |
| FLT3 Signaling | 5.28E-04 | Costimulation by the CD28 family | 0.034 | |
| Interleukin-3, Interleukin-5 and GM-CSF signaling | 9.97E-04 | MHC class II antigen presentation | 0.034 | |
| FLT3 signaling through SRC family kinases | 1.15E-03 | Antigen processing-Cross presentation | 0.034 | |
| Interleukin-4 and Interleukin-13 signaling | 1.36E-03 | Downstream signaling events of B Cell Receptor (BCR) | 0.034 | |
| Interleukin-1 signaling | 5.07E-03 | Endosomal/Vacuolar pathway | 0.04 | |
| Fcgamma receptor (FCGR) dependent phagocytosis | 0.011 | |||
| Interleukin-1 family signaling | 0.021 | |||
| TNFR2 non-canonical NF-kB pathway | 0.032 | |||
| Interleukin receptor SHC signaling | 0.034 | |||
| Innate Immunity | ||||
| Pathway | FDR | Pathway | FDR | |
| TRIF(TICAM1)-mediated TLR4 signaling | 8.80E-09 | CREB phosphorylation | 8.80E-05 | |
| MAP kinase activation | 7.94E-08 | MAPK targets/ Nuclear events mediated by MAP kinases | 1.97E-04 | |
| Toll Like Receptor 4 (TLR4) Cascade | 3.23E-07 | activated TAK1 mediates p38 MAPK activation | 2.26E-04 | |
| TRAF6 mediated induction of NFkB and MAP kinases upon TLR7/8 or 9 activation | 1.32E-06 | NOD1/2 Signaling Pathway | 2.64E-04 | |
| MyD88 dependent cascade initiated on endosome | 1.41E-06 | ERK/MAPK targets | 3.94E-04 | |
| Toll Like Receptor 7/8 (TLR7/8) Cascade | 1.50E-06 | TICAM1, RIP1-mediated IKK complex recruitment | 2.13E-03 | |
| Toll Like Receptor 9 (TLR9) Cascade | 2.07E-06 | Nucleotide-binding domain, leucine rich repeat containing receptor (NLR) signaling pathways | 3.24E-03 | |
| Innate Immune System | 3.79E-06 | IKK complex recruitment mediated by RIP1 | 3.46E-03 | |
| MyD88 cascade initiated on plasma membrane | 4.46E-06 | MAP3K8 (TPL2)-dependent MAPK1/3 activation | 0.011 | |
| Toll Like Receptor 10 (TLR10) Cascade | 4.46E-06 | TRAF6 mediated IRF7 activation in TLR7/8 or 9 signaling | 0.034 | |
| Toll Like Receptor 5 (TLR5) Cascade | 4.46E-06 | Neutrophil degranulation | 0.034 | |
| Toll Like Receptor 2 (TLR2) Cascade | 1.87E-05 | IRAK1 recruits IKK complex | 0.034 | |
| MyD88:MAL(TIRAP) cascade initiated on plasma membrane | 1.87E-05 | IRAK1 recruits IKK complex upon TLR7/8 or 9 stimulation | 0.034 | |
| Toll Like Receptor TLR6:TLR2 Cascade | 1.87E-05 | Activation of the AP-1 family of transcription factors | 0.034 | |
| Trafficking and processing of endosomal TLR | 5.09E-05 | |||
Several enriched immune pathways have been previously identified through experimental studies. For example, signaling by colony-stimulating factor (CSF1) in myeloid cells (FDR = 1.97E-04) and granulocyte–macrophage colony-stimulating factor (GM-CSF) (FDR = 9.97E-04) were enriched, consistent with the findings of Kumar et al. [99], who showed that CDK4/6 inhibition enhances CSF2 expression during abemaciclib treatment, promoting anti-tumorigenic M1 macrophage polarization and enhancing antigen presentation. In addition, multiple MyD88-related pathways were enriched, supporting prior evidence that CDK4/6 inhibitors can reprogram the tumor microenvironment by modulating the IL-33–MyD88 axis, therefore inhibiting suppressive immune cells [100]. Furthermore, Gene Ontology (GO) Biological Process (BP) enrichment analysis further supported these findings, yielding 1,613 enriched terms (FDR < 0.05, Supplementary Table S3). Among these were several strongly immune-related processes, including positive regulation of the immune system process (GO:0002684 with FDR of 8.24E-15), immune effector process (GO:0002252 with FDR of 4.97E-06), and immune response-regulating cell surface receptor signaling pathway (GO:0002768 with FDR of 4.00E-08).
To further investigate the relevance of these targets in different cell types, we analyzed the protein expression profiles of the 225 predicted targets using the HPA IHC dataset. This analysis showed that 24 protein targets exhibited a statistically significant higher expression in immune cell types (adjusted p < 0.05). In comparison, four targets showed higher expression in non-immune cell types. These include targets such as HLA-DRB1 (adj p-val 2.00E-13), JAK3 (adj p-val 3.46E-09), and TLR7 (adj p-val 1.90E-08). Identified proteins may serve as context-specific targets through which CDK4/6 inhibitors exert immunomodulatory effects. Their immune-preferential expression profiles may help guide further mechanistic or biomarker-driven studies (Supplementary Table S4).
Together, these results suggest that the targets of CDK4/6 inhibitors are not only involved in classical cell cycle pathways but also participate in key immune system processes. The functional enrichment and cell type-specific expression analyses together support a model in which CDK4/6 inhibition could synergize with immune checkpoint blockade by affecting immune signaling networks.
Conclusion and future perspectives
The studies depicted in this review highlight the valuable potential of CDK4/6 inhibitors to sensitize various solid malignancies to immune checkpoint inhibitor (ICI) immunotherapy. The review encompasses numerous solid tumor types for which clinical data have been published or ongoing clinical studies are underway. Early clinical data suggest that combining CDK4/6 inhibitors with immune checkpoint inhibitors (ICIs) holds therapeutic promise across multiple breast cancer subtypes, including hormone receptor-positive/HER2-negative (HR + /HER2 −) and androgen receptor-positive triple-negative breast cancer (AR + TNBC). Despite that, a significant challenge observed across several clinical trials is the heightened incidence of severe adverse events (AEs) when CDK4/6 inhibitors are combined with ICIs, particularly abemaciclib. Combinations like abemaciclib/pembrolizumab ± anastrozole in HR + /HER2- metastatic breast cancer led to two deaths and a high incidence of grade ≥ 3 AEs, including neutropenia, diarrhea, and liver function alterations, leading to trial discontinuation. Similar safety concerns prompted the termination of abemaciclib/nivolumab studies in HNSCC, NSCLC, HCC, and recurrent glioblastoma. High-grade hepatotoxicity was also reported with palbociclib/nivolumab/ anastrozole in primary breast cancer, causing study termination. These findings imply that such combinations may augment treatment-related toxicities. The high incidence of toxicity in some combination regimens highlights the critical need for better dosing optimization to improve tolerability and maximize the benefit/risk ratio. This includes exploring different schedules and drug sequences. The data from clinical trials in ovarian cancer, prostate cancer, and CRC are still awaited to judge if these novel approaches will allow the broader utilization of checkpoint inhibition in such settings. Efforts are needed to identify reliable biomarkers that can predict which patients or tumor types will best respond to CDK4/6 inhibitor combinations, especially with ICIs, and to help anticipate and manage toxicities. The identification of immune-preferential protein targets (e.g., HLA-DRB1, JAK3, TLR7) through in silico analysis offers a promising avenue for guiding further mechanistic or biomarker-driven studies.
Supplementary Information
Below is the link to the electronic supplementary material.
Acknowledgements
This work is partially supported by RESPECT grant (ID:10073) from the Egyptian Academy of Scientific Research and Technology (ASRT) to MFT.
Author contribution
MFT conceived the review idea and was responsible for project administration and funding acquisition; SA conducted the literature review, summarized and drafted several sections of the article; NW contributed tables and reference formatting; MR, AA, RA, and A-MR contributed to specific sections of the article; EB and AS performed the in silico analysis; A-MR, EB, and AS contributed to figures; MFT revised, edited and finalized the manuscript. All authors revised and approved the manuscript.
Funding
Open access funding provided by The Science, Technology & Innovation Funding Authority (STDF) in cooperation with The Egyptian Knowledge Bank (EKB). This work is partially supported by RESPECT grant (ID:10073) from the Egyptian Academy of Scientific Research and Technology (ASRT) to MFT.
Data availability
No datasets were generated or analyzed during the current study.
Declarations
Conflict of interest
The authors declare no competing interests.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- 1.Qi J, Ouyang Z. Targeting CDK4/6 for anticancer therapy. Biomedicines. 2022. 10.3390/biomedicines10030685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Shapiro GI. Cyclin-dependent kinase pathways as targets for cancer treatment. J Clin Oncol. 2006;24:1770–83. [DOI] [PubMed] [Google Scholar]
- 3.Witkiewicz AK, Knudsen ES. Retinoblastoma tumor suppressor pathway in breast cancer: prognosis, precision medicine, and therapeutic interventions. Breast Cancer Res. 2014;16:207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Rampioni Vinciguerra GL, Sonego M, et al. CDK4/6 inhibitors in combination therapies: better in company than alone: A mini review. Front Oncol. 2022;12: 891580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Rugo HS, Kabos P, et al. Abemaciclib in combination with pembrolizumab for HR+, HER2- metastatic breast cancer: phase 1b study. NPJ Breast Cancer. 2022;8:118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer. 2009;9:153–66. [DOI] [PubMed] [Google Scholar]
- 7.Matthews HK, Bertoli C, de Bruin RAM. Cell cycle control in cancer. Nat Rev Mol Cell Biol. 2022;23(1):74–88. [DOI] [PubMed] [Google Scholar]
- 8.Zhang S, Xu Q, Sun W, Zhou J, Zhou J. Immunomodulatory effects of CDK4/6 inhibitors. Biochim Biophys Acta Rev Cancer. 2023;1878(4): 188912. [DOI] [PubMed] [Google Scholar]
- 9.Spring LM, Wander SA, Andre F, Moy B, Turner NC, Bardia A. Cyclin-dependent kinase 4 and 6 inhibitors for hormone receptor-positive breast cancer: past, present, and future. Lancet. 2020;395(10226):817–27. [DOI] [PubMed] [Google Scholar]
- 10.Klein ME, Kovatcheva M, Davis LE, Tap WD, Koff A. CDK4/6 inhibitors: the mechanism of action may not be as simple as once thought. Cancer Cell. 2018;34(1):9–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.George MA, Qureshi S, Omene C, Toppmeyer DL, Ganesan S. Clinical and pharmacologic differences of CDK4/6 inhibitors in breast cancer. Front Oncol. 2021;12(11): 693104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Franco J, Balaji U, Freinkman E, Witkiewicz AK, Knudsen ES. Metabolic reprogramming of pancreatic cancer mediated by CDK4/6 inhibition elicits unique vulnerabilities. Cell Rep. 2016;14(5):979–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Bourdeau V, Ferbeyre G. CDK4-CDK6 inhibitors induce autophagy-mediated degradation of DNMT1 and facilitate the senescence antitumor response. Autophagy. 2016;12(10):1965–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zhou Y, Jin X, Ma J, Ding D, Huang Z, Sheng H, Yan Y, Pan Y, Wei T, Wang L, Wu H, Huang H. HDAC5 loss impairs RB repression of pro-oncogenic genes and confers CDK4/6 inhibitor resistance in cancer. Cancer Res. 2021;81(6):1486–99. [DOI] [PubMed] [Google Scholar]
- 15.Deng J, Wang ES, Jenkins RW, Li S, Dries R, Yates K, Chhabra S, Huang W, Liu H, Aref AR, Ivanova E, Paweletz CP, Bowden M, Zhou CW, Herter-Sprie GS, Sorrentino JA, Bisi JE, Lizotte PH, Merlino AA, Quinn MM, Bufe LE, Yang A, Zhang Y, Zhang H, Gao P, Chen T, Cavanaugh ME, Rode AJ, Haines E, Roberts PJ, Strum JC, Richards WG, Lorch JH, Parangi S, Gunda V, Boland GM, Bueno R, Palakurthi S, Freeman GJ, Ritz J, Haining WN, Sharpless NE, Arthanari H, Shapiro GI, Barbie DA, Gray NS, Wong KK. CDK4/6 inhibition augments antitumor immunity by enhancing T-cell activation. Cancer Discov. 2018;8(2):216–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Yu Q, Sicinska E, Geng Y, Ahnström M, Zagozdzon A, Kong Y, Gardner H, Kiyokawa H, Harris LN, Stål O, Sicinski P. Requirement for CDK4 kinase function in breast cancer. Cancer Cell. 2006;9(1):23–32. [DOI] [PubMed] [Google Scholar]
- 17.Sicinski P, Donaher JL, Parker SB, Li T, Fazeli A, Gardner H, Haslam SZ, Bronson RT, Elledge SJ, Weinberg RA. Cyclin D1 provides a link between development and oncogenesis in the retina and breast. Cell. 1995;82(4):621–30. [DOI] [PubMed] [Google Scholar]
- 18.Arnold A, Papanikolaou A. Cyclin D1 in breast cancer pathogenesis. J Clin Oncol. 2005;23(18):4215–24. [DOI] [PubMed] [Google Scholar]
- 19.Altucci L, Addeo R, Cicatiello L, Dauvois S, Parker MG, Truss M, Beato M, Sica V, Bresciani F, Weisz A. 17beta-estradiol induces cyclin D1 gene transcription, p36D1-p34cdk4 complex activation and p105Rb phosphorylation during mitogenic stimulation of G(1)-arrested human breast cancer cells. Oncogene. 1996;12(11):2315–24. [PubMed] [Google Scholar]
- 20.Watts CK, Sweeney KJ, Warlters A, Musgrove EA, Sutherland RL. Antiestrogen regulation of cell cycle progression and cyclin D1 gene expression in MCF-7 human breast cancer cells. Breast Cancer Res Treat. 1994;31(1):95–105. [DOI] [PubMed] [Google Scholar]
- 21.Musgrove EA, Caldon CE, Barraclough J, Stone A, Sutherland RL. Cyclin D as a therapeutic target in cancer. Nat Rev Cancer. 2011;11(8):558–72. [DOI] [PubMed] [Google Scholar]
- 22.Ali LR, Garrido-Castro AC, Lenehan PJ, Bollenrucher N, Stump CT, Dougan M, Goel S, Shapiro GI, Tolaney SM, Dougan SK. PD-1 blockade and CDK4/6 inhibition augment nonoverlapping features of T cell activation in cancer. J Exp Med. 2023;220(4): e20220729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Watanabe K, Niikura N, Kikawa Y, Oba M, Kobayashi K, Tada H, Ozaki S, Toh U, Yamamoto Y, Tsuneizumi M, Okuno T, Iwakuma N, Takeshita T, Iwamoto T, Ishiguro H, Masuda N, Saji S. Fulvestrant plus palbociclib in advanced or metastatic hormone receptor-positive/human epidermal growth factor receptor 2-negative breast cancer after fulvestrant monotherapy: Japan Breast Cancer Research Group-M07 (FUTURE trial). Breast Cancer Res Treat. 2023;199(2):253–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Finn RS, Martin M, Rugo HS, Jones S, Im SA, Gelmon K, Harbeck N, Lipatov ON, Walshe JM, Moulder S, Gauthier E, Lu DR, Randolph S, Diéras V, Slamon DJ. Palbociclib and letrozole in advanced breast cancer. N Engl J Med. 2016;375(20):1925–36. [DOI] [PubMed] [Google Scholar]
- 25.Hortobagyi GN, Stemmer SM, Burris HA, Yap YS, Sonke GS, Paluch-Shimon S, Campone M, Blackwell KL, André F, Winer EP, Janni W, Verma S, Conte P, Arteaga CL, Cameron DA, Petrakova K, Hart LL, Villanueva C, Chan A, Jakobsen E, Nusch A, Burdaeva O, Grischke EM, Alba E, Wist E, Marschner N, Favret AM, Yardley D, Bachelot T, Tseng LM, Blau S, Xuan F, Souami F, Miller M, Germa C, Hirawat S, O’Shaughnessy J. Ribociclib as first-line therapy for HR-positive, advanced breast cancer. N Engl J Med. 2016;375(18):1738–48. [DOI] [PubMed] [Google Scholar]
- 26.Sledge GW Jr, Toi M, Neven P, Sohn J, Inoue K, Pivot X, Burdaeva O, Okera M, Masuda N, Kaufman PA, Koh H, Grischke EM, Frenzel M, Lin Y, Barriga S, Smith IC, Bourayou N, Llombart-Cussac A. MONARCH 2: abemaciclib in combination with fulvestrant in women with HR+/HER2- advanced breast cancer who had progressed while receiving endocrine therapy. J Clin Oncol. 2017;35(25):2875–84. [DOI] [PubMed] [Google Scholar]
- 27.Sledge GW Jr, Toi M, Neven P, Sohn J, Inoue K, Pivot X, Burdaeva O, Okera M, Masuda N, Kaufman PA, Koh H, Grischke EM, Conte P, Lu Y, Barriga S, Hurt K, Frenzel M, Johnston S, Llombart-Cussac A. 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. 2020;6(1):116–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Loi S, Sirtaine N, Piette F, Salgado R, Viale G, Van Eenoo F, Rouas G, Francis P, Crown JP, Hitre E, de Azambuja E, Quinaux E, Di Leo A, Michiels S, Piccart MJ, Sotiriou C. Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG 02–98. J Clin Oncol. 2013;31(7):860–7. [DOI] [PubMed] [Google Scholar]
- 29.Swoboda A, Nanda R. Immune checkpoint blockade for breast cancer. Cancer Treat Res. 2018;173:155–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Rugo HS, Kabos P, Beck JT, Jerusalem G, Wildiers H, Sevillano E, Paz-Ares L, Chisamore MJ, Chapman SC, Hossain MA, Chen Y, Tolaney SM. Abemaciclib in combination with pembrolizumab for HR+, HER2- metastatic breast cancer: phase 1b study. NPJ Breast Cancer. 2022;8(1): 118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Masuda J, Tsurutani J, Masuda N, Tanabe Y, Iwasa T, Takahashi M, Futamura M, Matsumoto K, Aogi K, Iwata H, Hosonaga M, Mukohara T, Yoshimura K, Takano T. Abstract PS12–10: Phase II study of nivolumab in combination with abemaciclib plus endocrine therapy in patients with HR+, HER2- metastatic breast cancer: WJOG11418B NEWFLAME trial. Cancer Res. 2021;81(4):PS12–PS10. [Google Scholar]
- 32.Mayer EL, Ren Y, Wagle N, Mahtani R, Ma C, DeMichele A, Cristofanilli M, Meisel J, Miller KD, Abdou Y, Riley EC, Qamar R, Sharma P, Reid S, Sinclair N, Faggen M, Block CC, Ko N, Partridge AH, Chen WY, DeMeo M, Attaya V, Okpoebo A, Alberti J, Liu Y, Gauthier E, Burstein HJ, Regan MM, Tolaney SM. PACE: A randomized phase II study of fulvestrant, palbociclib, and avelumab after progression on cyclin-dependent kinase 4/6 inhibitor and aromatase inhibitor for hormone receptor–positive/human epidermal growth factor receptor–negative metastatic breast cancer. J Clin Oncol. 2024;42(17):2050–60. [DOI] [PubMed] [Google Scholar]
- 33.Mayer E, Ren Y, Wagle N, Mahtani R, Ma C, DeMichele A, Cristofanilli M, Meisel J, Miller K, Jolly T, Riley E, Qamar R, Sharma P, Reid S, Sinclair N, Faggen M, Block C, Ko N, Partridge A, Tolaney S. Abstract GS3–06: GS3–06 palbociclib After CDK4/6i and endocrine therapy (PACE): a randomized phase II study of fulvestrant, palbociclib, and avelumab for endocrine pre-treated ER+/HER2- metastatic breast cancer. Cancer Res. 2023;83:GS3-06. [Google Scholar]
- 34.Yuan Y, Lee JS, Yost SE, Frankel PH, Ruel C, Egelston CA, Guo W, Padam S, Tang A, Martinez N, Schmolze D, Presant C, Ebrahimi B, Yeon C, Sedrak M, Patel N, Portnow J, Lee P, Mortimer J. Phase I/II trial of palbociclib, pembrolizumab and letrozole in patients with hormone receptor-positive metastatic breast cancer. Eur J Cancer. 2021;154:11–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Jerusalem G, Prat A, Salgado R, Reinisch M, Saura C, Ruiz-Borrego M, Nikolinakos P, Ades F, Filian J, Huang N, Mazzei-Abba A, Tolane SM. Neoadjuvant nivolumab + palbociclib + anastrozole for oestrogen receptor-positive/human epidermal growth factor receptor 2-negative primary breast cancer: results from CheckMate 7A8. Breast. 2023;72:103580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Okines A, Moghadam H, Sparks L, Mohammed K, Dunne K, Nerurkar A, Osin P, Swift C, Sardinha R, Turner N. Abstract P4–01-02: Results from a dose escalation phase 1b study of palbociclib and avelumab in advanced breast cancer in the PAveMenT Trial. Cancer Res. 2023;83(5):P4-01. [Google Scholar]
- 37.Herold CI, Trippa L, Li T, Do K, Bardia A, Anderson L, Montazeri K, Pittenger J, Andrews C, Mittendorf EA, Goel S, Winer EP, Shapiro GI, Tolaney SM. Abstract P3–14-03: A phase 1b study of the CDK4/6 inhibitor ribociclib in combination with the PD-1 inhibitor spartalizumab in patients with hormone receptor-positive metastatic breast cancer (HR+ MBC) and metastatic ovarian cancer (MOC). Cancer Res. 2020;80(4):P3-14. [Google Scholar]
- 38.Ferris RL, Blumenschein G Jr, Fayette J, Guigay J, Colevas AD, Licitra L, Harrington K, Kasper S, Vokes EE, Even C, Worden F, Saba NF, Iglesias Docampo LC, Haddad R, Rordorf T, Kiyota N, Tahara M, Monga M, Lynch M, Geese WJ, Kopit J, Shaw JW, Gillison ML. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N Engl J Med. 2016;375(19):1856–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Chow LQM, Haddad R, Gupta S, Mahipal A, Mehra R, Tahara M, Berger R, Eder JP, Burtness B, Lee SH, Keam B, Kang H, Muro K, Weiss J, Geva R, Lin CC, Chung HC, Meister A, Dolled-Filhart M, Pathiraja K, Cheng JD, Seiwert TY. Antitumor Activity of Pembrolizumab in Biomarker-Unselected Patients With Recurrent and/or Metastatic Head and Neck Squamous Cell Carcinoma: Results From the Phase Ib KEYNOTE-012 Expansion Cohort. J Clin Oncol. 2016;34(32):3838–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Wise-Draper TM, Gulati S, Palackdharry S, Hinrichs BH, Worden FP, Old M, Dunlap NE, Kaczmar JM, Patil Y, Riaz MK, Tang A, Mark J, Zender C, Gillenwater AM, Bell D, Kurtzweil N, Mathews M, Allen CL, Mierzwa ML, Casper K, Jandarov R, Medvedovic M, Lee JJ, Harun N, Takiar V, Gillison M. Phase II Clinical Trial of Neoadjuvant and Adjuvant Pembrolizumab in Resectable Local-Regionally Advanced Head and Neck Squamous Cell Carcinoma. Clin Cancer Res. 2022;28(7):1345–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Yen CJ, Kiyota N, Hanai N, Takahashi S, Yokota T, Iwae S, Shimizu Y, Hong RL, Goto M, Kang JH, Li WSK, Ferris RL, Gillison M, Endo T, Jayaprakash V, Tahara M. Two-year follow-up of a randomized phase III clinical trial of nivolumab vs. the investigator’s choice of therapy in the Asian population for recurrent or metastatic squamous cell carcinoma of the head and neck (CheckMate 141). Head Neck. 2020;42(10):2852–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Zolkind P, Uppaluri R. Checkpoint immunotherapy in head and neck cancers. Cancer Metastasis Rev. 2017;36(3):475–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Taberna M, Mena M, Pavón MA, Alemany L, Gillison ML, Mesía R. Human papillomavirus-related oropharyngeal cancer. Ann Oncol. 2017;28(10):2386–98. [DOI] [PubMed] [Google Scholar]
- 44.van Caloen G, Machiels JP. Potential role of cyclin-dependent kinase 4/6 inhibitors in the treatment of squamous cell carcinoma of the head and neck. Curr Opin Oncol. 2019;31(3):122–30. [DOI] [PubMed] [Google Scholar]
- 45.Riess C, Irmscher N, Salewski I, Strüder D, Classen CF, Große-Thie C, Junghanss C, Maletzki C. Cyclin-dependent kinase inhibitors in head and neck cancer and glioblastoma—backbone or add-on in immune-oncology? Cancer Metastasis Rev. 2021;40(1):153–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Sridaran D, Ramamoorthi G, MahaboobKhan R, Kumpati P. Oxystressed tumor microenvironment potentiates epithelial to mesenchymal transition and alters cellular bioenergetics towards cancer progression. Tumour Biol. 2016;37(10):13307–22. [DOI] [PubMed] [Google Scholar]
- 47.Van Caloen G. Preclinical activity of cyclin-dependent kinase 4/6 inhibitor in squamous cell carcinoma of the head and neck. European J Cancer. 2016;69:S128. [Google Scholar]
- 48.van Caloen G, Schmitz S, El Baroudi M, Caignet X, Pyr Dit Ruys S, Roger PP, Vertommen D, Machiels JP. Preclinical activity of ribociclib in squamous cell carcinoma of the head and neck. Mol Cancer Ther. 2020;19(3):777–89. [DOI] [PubMed] [Google Scholar]
- 49.Billard-Sandu C, Tao YG, Sablin MP, Dumitrescu G, Billard D, Deutsch E. CDK4/6 inhibitors in P16/HPV16-negative squamous cell carcinoma of the head and neck. Eur Arch Otorhinolaryngol. 2020;277(5):1273–80. [DOI] [PubMed] [Google Scholar]
- 50.Gatfield ER, Tadross J, Ince W. Immune checkpoint inhibitor use in head and neck squamous cell carcinoma: the current landscape and future perspectives. Future Oncol. 2024;20(23):1695–711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kao H, Huang H, Liao B, Hsu M, Hong R. 907P ribociclib and spartalizumab for head and neck squamous cell carcinoma: a phase I study with expansion cohort. Ann Oncol. 2021;32:S805–6. [Google Scholar]
- 52.Ettl T, Schulz D, Bauer RJ. The renaissance of cyclin dependent kinase inhibitors. Cancers (Basel). 2022;14(2): 293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Zhang J, Xu D, Zhou Y, Zhu Z, Yang X. Mechanisms and implications of CDK4/6 inhibitors for the treatment of NSCLC. Front Oncol. 2021;11: 676041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Goldman JW, Mazieres J, Barlesi F, Dragnev KH, Koczywas M, Göskel T, Cortot AB, Girard N, Wesseler C, Bischoff H, Nadal E, Park K, Lu S, Taus A, Cobo M, Estrem ST, Wijayawardana SR, Turner K, Oakley GJ3rd, Hurt KC, Chiang AY, Hossain AM, John WJ, Paz-Ares L. A randomized phase III study of abemaciclib versus erlotinib in patients with stage IV non-small cell lung cancer with a detectable KRAS mutation who failed prior platinum-based therapy: JUNIPER. Front Oncol. 2020;10: 578756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Ahn ER, Mangat PK, Garrett-Mayer E, Halabi S, Dib EG, Haggstrom DE, Alguire KB, Calfa CJ, Cannon TL, Crilley PA, Gaba AG, Marr AS, Sangal A, Thota R, Antonelli KR, Islam S, Rygiel AL, Bruinooge SS, Schilsky RL. Palbociclib in patients with non-small-cell lung cancer with CDKN2A alterations: results from the targeted agent and profiling utilization registry study. JCO Precis Oncol. 2020;4:757–66. [DOI] [PubMed] [Google Scholar]
- 56.Chen DS, Irving BA, Hodi FS. Molecular pathways: Next-Generation immunotherapy—inhibiting programmed death-ligand 1 and programmed death-1. Clin Cancer Res. 2012;18(24):6580–7. [DOI] [PubMed] [Google Scholar]
- 57.Zimmermann S, Peters S, Owinokoko T, Gadgeel SM. Immune checkpoint inhibitors in the management of lung cancer. Am Soc Clin Oncol Educ Book. 2018;23(38):682–95. [DOI] [PubMed] [Google Scholar]
- 58.Fan Y, Mao WJCCO. Immune checkpoint inhibitors in lung cancer: current status and future directions. Chin Clin Oncol. 2017;6(2):17. [DOI] [PubMed] [Google Scholar]
- 59.Reck M, Taylor F, Penrod JR, DeRosa M, Morrissey L, Dastani H, Orsini L, Gralla RJ. Impact of nivolumab versus docetaxel on health-related quality of life and symptoms in patients with advanced squamous non-small cell lung cancer: results from the CheckMate 017 study. J Thorac Oncol. 2018;13:194–204. [DOI] [PubMed] [Google Scholar]
- 60.Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, Chow LQ, Vokes EE, Felip E, Holgado E, Barlesi F, Kohlhäufl M, Arrieta O, Burgio MA, Fayette J, Lena H, Poddubskaya E, Gerber DE, Gettinger SN, Rudin CM, Rizvi N, Crinò L, Blumenschein GR Jr., Antonia SJ, Dorange C, Harbison CT, Graf Finckenstein F, Brahmer JR. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373(17):1627–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Borghaei H, Gettinger S, Vokes EE, Chow LQM, Burgio MA, de Castro Carpeno J, Pluzanski A, Arrieta O, Frontera OA, Chiari R, Butts C, Wójcik-Tomaszewska J, Coudert B, Garassino MC, Ready N, Felip E, García MA, Waterhouse D, Domine M, Barlesi F, Antonia S, Wohlleber M, Gerber DE, Czyzewicz G, Spigel DR, Crinò L, Eberhardt WEE, Li A, Marimuthu S, Brahmer J. Five-year outcomes from the randomized, phase III trials CheckMate 017 and 057: nivolumab versus docetaxel in previously treated non-small-cell lung cancer. J Clin Oncol. 2021;39(7):723–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Pujol JL, Vansteenkiste J, Paz-Ares Rodríguez L, Gregorc V, Mazieres J, Awad M, Jänne PA, Chisamore M, Hossain AM, Chen Y, Beck JT. Abemaciclib in combination with pembrolizumab for stage IV KRAS-mutant or squamous NSCLC: a phase 1b study. JTO Clin Res Rep. 2021;2(11): 100234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Lopez PG, Goldman J, Kelly K, Kim ES, Beck TJ, Garcia MA, Yang Z, Ingram KJ, Amstutz L, John WJ, Pulla MP. Efficacy and safety of abemaciclib combined with either LY3023414 or pembrolizumab in stage IV NSCLC. Ann Oncol. 2017;28: v487. [Google Scholar]
- 64.Lu JW, Lin YM, Chang JG, Yeh KT, Chen RM, Tsai JJ, Su WW, Hu RM. Clinical implications of deregulated CDK4 and cyclin D1 expression in patients with human hepatocellular carcinoma. Med Oncol. 2013;30(1): 379. [DOI] [PubMed] [Google Scholar]
- 65.Digiacomo G, Fumarola C, La Monica S, Bonelli MA, Cretella D, Alfieri R, Cavazzoni A, Galetti M, Bertolini P, Missale G, Petronini PG. Simultaneous combination of the CDK4/6 inhibitor palbociclib with regorafenib induces enhanced anti-tumor effects in hepatocarcinoma cell lines. Front Oncol. 2020;10: 563249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Sheng J, Kohno S, Okada N, Okahashi N, Teranishi K, Matsuda F, Shimizu H, Linn P, Nagatani N, Yamamura M, Harada K, Horike SI, Inoue H, Yano S, Kumar S, Kitajima S, Ajioka I, Takahashi C. Treatment of RB1-intact hepatocellular carcinoma with CDK4/6 inhibitor combination therapy. Hepatology (Baltimore, MD). 2021;74(4):1971–93. [DOI] [PubMed] [Google Scholar]
- 67.Littman S, Brus C, Burkart A. A phase II study of palbociclib (PD-0332991) in adult patients with advanced hepatocellular carcinoma. J Clin Oncol. 2015;33:277–277. [Google Scholar]
- 68.Yau T, Park JW, Finn RS, Cheng AL, Mathurin P, Edeline J, Kudo M, Harding JJ, Merle P, Rosmorduc O, Wyrwicz L, Schott E, Choo SP, Kelley RK, Sieghart W, Assenat E, Zaucha R, Furuse J, Abou-Alfa GK, El-Khoueiry AB, Melero I, Begic D, Chen G, Neely J, Wisniewski T, Tschaika M, Sangro B. Nivolumab versus sorafenib in advanced hepatocellular carcinoma (CheckMate 459): a randomised, multicentre, open-label, phase 3 trial. Lancet Oncol. 2022;23:77–90. [DOI] [PubMed] [Google Scholar]
- 69.Finn R, Ryoo BY, Merle P, Kudo M, Bouattour M, Lim HY, Breder V, Edeline J, Chao Y, Ogasawara S, Yau T, Garrido M, Chan S, Knox J, Daniele B, Ebbinghaus S, Chen E, Siegel A, Zhu A, Cheng AL. Results of KEYNOTE-240: phase 3 study of pembrolizumab (Pembro) vs best supportive care (BSC) for second line therapy in advanced hepatocellular carcinoma (HCC). J Clin Oncol. 2019;37:4004–4004. [Google Scholar]
- 70.Zhang QF, Li J, Jiang K, Wang R, Ge JL, Yang H, Liu SJ, Jia LT, Wang L, Chen BL. CDK4/6 inhibition promotes immune infiltration in ovarian cancer and synergizes with PD-1 blockade in a B cell-dependent manner. Theranostics. 2020;10(23):10619–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Vázquez-Montero L, de la Gala M, de la Cruz-Merino L. Nivolumab plus ipilimumab in metastatic melanoma: a critical appraisal focused on specific subpopulations. Front Oncol. 2023;13:1187840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Lau PKH, Cullinane C, Jackson S, Walker R, Smith LK, Slater A, Kirby L, Patel RP, von Scheidt B, Slaney CY, McArthur GA, Sheppard KE. Enhancing adoptive cell transfer with combination BRAF-MEK and CDK4/6 inhibitors in melanoma. Cancers (Basel). 2021;13(24): 6342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Patnaik A, Yap TA, Chung HC, de Miguel MJ, Bang YJ, Lin CC, Su WC, Italiano A, Chow KH, Szpurka AM, Yu D, Zhao Y, Carlsen M, Schmidt S, Vangerow B, Gandhi L, Xu X, Bendell J. Safety and clinical activity of a new anti-PD-L1 antibody as monotherapy or combined with targeted therapy in advanced solid tumors: the PACT phase Ia/Ib trial. Clin Cancer Res. 2021;27(5):1267–77. [DOI] [PubMed] [Google Scholar]
- 74.Balk SP, Knudsen KE. AR, the cell cycle, and prostate cancer. Nucl Recept Signal. 2008;6: e001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kase AM, Copland Iii JA, Tan W. Novel therapeutic strategies for cdk4/6 inhibitors in metastatic castrate-resistant prostate cancer. Onco Targets Ther. 2020;13:10499–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Guney Eskiler G, Deveci Ozkan A, Haciefendi A, Bilir C. Mechanisms of abemaciclib, a CDK4/6 inhibitor, induced apoptotic cell death in prostate cancer cells in vitro. Transl Oncol. 2022;15(1): 101243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.de Kouchkovsky I, Rao A, Carneiro BA, Zhang L, Lewis C, Phone A, Small EJ, Friedlander T, Fong L, Paris PL, Ryan CJ, Szmulewitz RZ, Aggarwal RA. A Phase Ib/II Study of the CDK4/6 inhibitor ribociclib in combination with docetaxel plus prednisone in metastatic castration-resistant prostate cancer. Clin Cancer Res. 2022;28(8):1531–9. [DOI] [PubMed] [Google Scholar]
- 78.Kim J, Shaffer D, Massard C, Powles T, Harshman L, Braiteh F, Conkling P, Sarkar I, Kadel E, Mariathasan S, O’Hear C, Schiff C, Fasso M, Carroll S, Petrylak D. A phase Ia study of safety and clinical activity of atezolizumab (atezo) in patients (pts) with metastatic castration-resistant prostate cancer (mCRPC). JClin Oncol. 2018;36(6):187–187. [Google Scholar]
- 79.Rao A, Kwak L, Reimers M, Reichert Z, Thyagarajan B, Fernandez K, Bretta K, Pfaff K, Rodig S, Alva A, Shapiro G, Ryan C, Choudhury A. A phase II trial of abemaciclib (abema) and atezolizumab (atezo) in unselected and CDK12-loss metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol. 2022;40: TPS213-TPS213. [Google Scholar]
- 80.Wang Y, Li Y, Liu D, Zheng D, Li X, Li C, Huang C, Wang Y, Wang X, Li Q, Xu J. A potential anti-glioblastoma compound LH20 induces apoptosis and arrest of human glioblastoma cells via CDK4/6 inhibition. Molecules. 2023;28:5047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Xiao J, Liang J, Fan J, Hou P, Li X, Zhang H, Li K, Bu L, Li P, He M, Zhong Y, Guo L, Jia P, Xiao Q, Wu J, Peng H, Li C, Xing F, Guo D. CDK4/6 inhibition enhances oncolytic virus efficacy by potentiating tumor-selective cell killing and T-cell activation in refractory glioblastoma. Cancer Res. 2022;82(18):3359–74. [DOI] [PubMed] [Google Scholar]
- 82.Juric V, Murphy B. Cyclin-dependent kinase inhibitors in brain cancer: current state and future directions. Cancer Drug Resist. 2020;3(1):48–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Lubanska D, Porter L. Revisiting CDK inhibitors for treatment of glioblastoma multiforme. Drugs R D. 2017;17(2):255–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Riess C, Irmscher N, Salewski I, Strüder D, Classen CF, Große-Thie C, Junghanss C, Maletzki C. Cyclin-dependent kinase inhibitors in head and neck cancer and glioblastoma-backbone or add-on in immune-oncology? Cancer Metastasis Rev. 2021;40(1):153–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Olmez I, Zhang Y, Manigat L, Benamar M, Brenneman B, Nakano I, Godlewski J, Bronisz A, Lee J, Abbas T, Abounader R, Purow B. Combined c-Met/Trk inhibition overcomes resistance to CDK4/6 inhibitors in glioblastoma. Cancer Res. 2018;78(15):4360–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Khasraw M, Fujita Y, Lee-Chang C, Balyasnikova IV, Najem H, Heimberger AB. New approaches to glioblastoma. Annu Rev Med. 2022;73:279–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Jardim DL, Millis SZ, Ross JS, Woo MS, Ali SM, Kurzrock R. Cyclin pathway genomic alterations across 190,247 solid tumors: leveraging large-scale data to inform therapeutic directions. Oncologist. 2021;26(1):e78–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Jun SY, Kim J, Yoon N, Maeng LS, Byun JH. Prognostic potential of cyclin D1 expression in colorectal cancer. J Clin Med. 2023;12(2): 572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Yu M, Wu W, Sun Y, Yan H, Zhang L, Wang Z, Gong Y, Wang T, Li Q, Song J, Wang M, Zhang J, Tang Y, Zhan J, Zhang H. FRMD8 targets both CDK4 activation and RB degradation to suppress colon cancer growth. Cell Rep. 2023;42(8): 112886. [DOI] [PubMed] [Google Scholar]
- 90.Thoma OM, Neurath MF, Waldner MJ. Cyclin-dependent kinase inhibitors and their therapeutic potential in colorectal cancer treatment. Front Pharmacol. 2021;12: 757120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Ziemke EK, Dosch JS, Maust JD, Shettigar A, Sen A, Welling TH, Hardiman KM, Sebolt-Leopold JS. Sensitivity of KRAS-mutant colorectal cancers to combination therapy that cotargets MEK and CDK4/6. Clin Cancer Res. 2016;22(2):405–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Martin-Broto J, Martinez-Garcia J, et al. Phase II trial of CDK4/6 inhibitor palbociclib in advanced sarcoma based on mRNA expression of CDK4/CDKN2A. Signal Transduct Target Ther. 2023;8:405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Anastasiou M, Kyriazoglou A, et al. Immune checkpoint inhibitors in sarcomas: a systematic review. Immunooncol Technol. 2023;20: 100407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.D’Angelo SP, Rosenbaum E, et al. A phase II study of palbociclib combined with retifanlimab in patients with advanced dedifferentiated liposarcoma. J Clin Oncol. 2023;41: TPS11590-TPS11590. [Google Scholar]
- 95.Daina A, Michielin O, Zoete V. SwissTargetPrediction: updated data and new features for efficient prediction of protein targets of small molecules. Nucleic Acids Res. 2019;47(W1):W357–64. 10.1093/nar/gkz382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T. Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Blucher AS, McWeeney SK, Stein L, Wu G. Visualization of drug target interactions in the contexts of pathways and networks with ReactomeFIViz. F1000Research. 2019;8:908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Thul PJ, Lindskog C. The human protein atlas: a spatial map of the human proteome. Protein Sci. 2018;27(1):233–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Kumar A, Ramani V, Bharti V, de Lima Bellan D, Saleh N, Uzhachenko R, Shen C, Arteaga C, Richmond A, Reddy SM, Vilgelm A. Dendritic cell therapy augments antitumor immunity triggered by CDK4/6 inhibition and immune checkpoint blockade by unleashing systemic CD4 T-cell responses. J Immunother Cancer. 2023;11(5): e006019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Sun M, Dong L, Wang Y, Liu C, Du J, Wang B, Xing B, Yao X, Ren Y, Zhou X. The role of targeting CDK4/6 in cancer immunotherapy. Holistic Integ Oncol. 2024;3(1):32. [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
No datasets were generated or analyzed during the current study.





