Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Review ArticleReview
Open Access

Mechanism of CDK4/6 Inhibitor Resistance in Hormone Receptor-positive Breast Cancer and Alternative Treatment Strategies

CHUN-MING CHANG and HO YIN PEKKLE LAM
Anticancer Research December 2023, 43 (12) 5283-5298; DOI: https://doi.org/10.21873/anticanres.16732
CHUN-MING CHANG
1Department of General Surgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, R.O.C.;
2Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan, R.O.C.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
HO YIN PEKKLE LAM
2Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan, R.O.C.;
3Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan, R.O.C.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: pekklelavabo{at}mail.tcu.edu.tw
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Breast cancer (BC) is a common malignancy in women, with hormone receptor (HR)-positive subtype responsible for approximately 70% of cases. Currently, patients with metastatic HR-positive BC rely on endocrine therapy and cyclin-dependent kinase (CDK)-4/6 inhibitors for treatment. Currently, approved CDK4/6 inhibitors include palbociclib, ribociclib, and abemaciclib. However, clinical evidence of CDK-4/6 inhibitor resistance is emerging, suggesting that the gap in the knowledge of its resistance mechanism requires further investigation. This review discusses the mechanisms of CDK4/6 inhibitor resistance in BC, including both intrinsic and extrinsic mechanisms. We also discuss possible alternative strategies to overcome CDK4/6 inhibitor resistance in future clinical applications.

Key Words:
  • CDK4/6 inhibitors-resistance
  • breast cancer
  • palbociclib
  • ribociclib
  • abemaciclib
  • review

Breast cancer (BC) is a commonly diagnosed malignancy accounting for 25% of cancer cases among women worldwide (1). Currently, BC can be divided into different subtypes based on the presence of three hormone receptors: estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Different combinations of these hormone receptors characterize three main subtypes of breast cancer: luminal type, also known as hormone receptor (HR)-positive subtype, HER2-positive subtype, and triple-negative subtype. Among these, HR-positive BC is the most common subtype, accounting for 70% of all BC malignancies (2).

Clinically, HR-positive BC relies on endocrine therapy as its first-line treatment (3). These include tamoxifen, used in premenopausal women, and aromatase inhibitors, used in postmenopausal women (4). However, many patients develop resistance to hormonal blockade, resulting in distant recurrence (5). Because cyclin-dependent kinases (CDKs) play an essential role in initiating the cell cycle in cancers (6), CDKs inhibitors have been developed over the past two decades for the treatment of BC. CDK4/6 inhibitors combined with endocrine therapy for HR-positive BC have recently become a new treatment standard (7) (Figure 1). However, clinical evidence of CDKs inhibitor resistance (8, 9) suggests the need for alternative approaches for HR-positive BC patients. In this review, we will discuss the mechanism of CDK inhibitor resistance in HR-positive BC. We will also discuss possible alternative strategies to overcome CDKs inhibitor resistance for future clinical application.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Regulation of cell cycle in hormone receptor-positive breast cancer. In breast cancer cells, mechanisms involved in cell cycle regulation can be broadly classified as cell cycle-specific mechanisms and cell cycle non-specific mechanisms. In cell cycle-specific mechanisms, cyclin-dependent kinases (CDKs) and cyclins remain the major regulatory factors. Several CDKs including CDK1, CDK2, CDK4, CDK6, and CDK7 bind with different cyclins (cyclin-A, -B, -D, and -E). CDKs-cyclin complexes then work together to facilitate cell cycle progression, but these complexes can also be inhibited by cyclin inhibitors, such as p16 and p21. Other cell cycle non-specific mechanisms, such as PI3K signaling, MAPK signaling, or DNA repair systems may also indirectly affect cell cycle progression. Many current drugs target these pathways to combat hormone receptor-positive breast cancer. Arrows suggest positive regulation; blunt arrows indicate inhibitory regulation. The red box shows current therapeutic targets and corresponding drugs.

CDK4/6 Inhibitors

Cyclins, including cyclin G1, cyclin G1/S, cyclin S, and cyclin M, are the most critical cell cycle regulators that drive cells in a particular cell cycle phase, which helps to regulate their growth and metabolism (10). Cyclins are activated or inactivated by cyclin-dependent kinases (CDKs) (11). However, aberrant CDK activation is usually observed in cancers. Overactivation of CDKs, such as CDK4 and CDK6 has been shown to positively contribute to tumor progression (12). The CDK4/6 complex, after interacting with cyclin D1, a direct transcriptional target of estrogen-receptor signaling, can facilitate the inhibition of retinoblastoma (Rb) protein, which leads cells to enter the G1 checkpoint to the S phase of the cell cycle (13). In BC, the dysregulated activity of cyclin D1 and CDK4/6 is considered to play a significant role in estrogen-driven tumor proliferation (14). CDK4/6 up-regulation and cyclin D1 amplification have also been found in HR-positive BC (15).

Due to the importance of CDK4/6 activity in BC progression, CDK4/6 inhibitors have been considered a promising target (13, 16, 17). Three U.S. Food and Drug Administration (FDA)-approved CDK4/6 inhibitors, palbociclib, ribociclib, and abemaciclib are currently used for BC treatment (18). Combining these CDK4/6 inhibitors with endocrine therapy has been shown to prolong the progression-free survival of patients with advanced HR-positive BC (19-24) (Table I). In addition, newer CDK4/6 inhibitors, such as trilaciclib and dalpiciclib (also known as SHR6390) have recently been developed and have shown favorable pharmaceutical properties in in vivo models (25-27). These drugs have recently entered phase III clinical trials (ClinicalTrials.gov identifier: NCT04799249 and NCT03927456).

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table I.

Current usage of CDK4/6 inhibitors with hormone therapy.

Although CDK4/6 inhibitors hold great promise against BC, tumor resistance to CDK4/6 inhibitors has been reported worldwide (28, 29), and there is an urgent need to investigate its mechanism.

CDK4/6 Inhibitor Resistance

Approximately 20% of patients with BC are not responsive to CDK4/6 inhibitors (30). The tumors of these patients were shown to process intrinsic mutations that allowed them to avoid the action of CDK4/6 inhibitors and proliferate in the presence of the drug. These intrinsic mutations include but are not limited to CDK4/6 over-expression, loss of Rb function, and p16 amplification (Figure 2).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Mechanisms of CDK4/6 inhibitor resistance. Multiple mechanisms, directly or indirectly, involved in cell cycle regulation may be associated with CDK4/6 inhibitor resistance. Current mechanisms implicated in developing resistance to CDK4/6 inhibitors are highlighted in the figure. The red box shows intrinsic mechanisms, whereas the blue box shows extrinsic mechanisms. CDK4/6 over-expression remains a primary reason for intrinsic mechanisms. Intrinsic mechanisms that lead to CDK4/6 inhibitor resistance also include loss of Rb or ER and over-expression of p16 or CDK-2. Some of the miRNAs can also affect the sensitivity of tumor cells to CDK4/6 inhibitors. Resistance to CDK4/6 inhibitors can also occur through an acquired (extrinsic) mechanism. These mechanisms include over-activation of oncogenic signaling pathways, such as PI3K, MAPK, and TGFR signaling. PI3K mutation can also lead to overactivation of its signaling cascade. Defects in DNA damage repair mechanisms, such as ATR Chk1 and ATM Chk2 pathways, result in un-arrested cell cycles. Other mechanisms, such as alternation in the Hippo pathway, HDAC over-expression, and ER over-activation may also lead to CDK4/6 inhibitor resistance.

The phase III clinical trial, PALOMA-2 (ClinicalTrials.gov identifier: NCT01740427) (20) showed that more than 30% of BC patients experienced a recurrence within two years of palbociclib treatment, and over 70% of patients developed resistance to palbociclib and letrozole treatment 40 months later, suggesting that even the combined use of CDK4/6 inhibitor with other therapeutic drugs may lead to insensitivity to CDK inhibitors and allow tumor cells to return to a proliferative phenotype. Several underlying mechanisms have been proposed to explain this acquired resistance, such as epigenetic alternations, overactivation of mitogenic signaling, such as PI3K-AKT-mTOR signaling, or mutations in the component of the signaling pathway (Figure 2).

Intrinsic Resistance Mechanism

CDK4/6 over-expression. Over-expression of CDK4 or CDK6 has been suggested as the primary resistance mechanism to CDK4/6 inhibitors. Higher CDK4 expression has been detected in CDK4/6 inhibitor-resistant cancer cells (31), indicating that CDK4 over-expression may be correlated with resistance to CDK4/6 inhibitors. In addition, over-expression of CDK6 suppressed the sensitivity of CDK4/6 inhibitors in luminal BC cells (12). Although there are reports that amplification of CDK6 promotes BC resistance to CDK4/6 inhibitors, and CDK6 knockdown rescued therapy sensitivity, this phenomenon may be independent of CDK4 (12). In support of this, CDK4 amplification has not been detected in a CDK inhibitor-resistant BC model, although amplification of CDK4 has been reported in other resistant cancers (32). Cyclin D1, D2, and D3 up-regulation has also been observed in CDK4/6 inhibitors-resistant BCs (33-35). These results suggested that resistance could occur by either non-canonical activation of CDKs or by forming cyclin D-CDK4/6 complexes.

Although no reports have so far described mutations in CDK4 or CDK6 that could possibly reduce the affinity of CDK4/6 inhibitors, this remains a possible mechanism which cannot be overlooked.

Loss of Rb Function. Rb is a tumor suppressor that prevents G1/S transition during cell cycle progression (36). One of the major mechanisms of resistance to CDK4/6 inhibitors is the loss of functional Rb, which has been observed in many tumor types. In a case report in which HR-positive BC patients were treated with a combination of palbociclib and fulvestrant or ribociclib and letrozole, acquired Rb mutations were detected (28). In the phase III PALOMA-3 trial (ClinicalTrials.gov identifier: NCT0192135), whole-exome sequencing of circulating tumor DNA (ctDNA) confirmed CDK4/6-induced, acquired Rb mutations in patients receiving palbociclib and fulvestrant (29). Loss of functional Rb was subsequently identified in other studies where patients resisted CDK4/6 inhibitors (37-39). Notably, the loss of Rb is correlated with the increased expression of E2F (40), resulting in the constitutive activation of its downstream target proteins. However, the presence of Rb does not usually guarantee resistance to CDK4/6 inhibitors, as seen in Rb-mutated bladder cancers where palbociclib was effective (41). Furthermore, putative attenuation of Rb functions through Rb mutations, identified in ctDNA, has been observed in CDK4/6 inhibitor-resistant patients (28, 42).

p16 over-expression. p16 is a CDK4 inhibitor that prevents Rb phosphorylation and promotes G1 cell cycle arrest, leading to cell senescence (10, 43). Therefore, p16 generally serves as a tumor suppressor. However, p16 over-expression may suppress the activity of CDK4 and expression of cyclin D1 (44), which are the main targets of CDK4/6 inhibitors, therefore reducing the effects of CDK4/6 inhibitors (45). Furthermore, resistance to CDK4/6 inhibitors is related more to the over-expression of p16 than Rb expression, as the expression level of p16 largely correlates to the effectiveness of CDK4/6 inhibition (44). However, low expression of p16 did not improve the therapeutic benefit of palbociclib in Rb-positive or HR-positive BC patients (46). However, p16 over-expression in the absence of Rb in CDK4/6 inhibitor-resistant patients (47) suggests that p16 over-expression may mediate CDK4/6 inhibitor resistance through other mechanisms, such as Rb loss, to introduce.

Overactivation of cyclin E-CDK2 signaling. In normal cells, phosphorylated cyclin D-CDK4/6 and cyclin E1/2-CDK2 complexes work together to inactivate Rb through phosphorylation, which releases E2F transcription factors. Therefore, without cyclin D1-CDK4/6, endogenous cyclin E1/2-CDK2 cannot effectively phosphorylate Rb to release E2F. Although cyclin E2 is also a transcriptional target of E2F, cyclin E2-CDK2 complexes are thought to be suppressed by CDK4/6 inhibition (48). However, over-expression of cyclins E or CDK2 may allow cells to overturn CDK4/6 inhibition.

Expression of cyclin E1, cyclin E2, and CDK2 has been shown to be up-regulated in several CDK4/6 inhibitor resistance models (12, 34, 49). The up-regulation can occur through the amplification of CCNE1 (which encodes cyclin E1) (49) or CCNE2 (34). Cyclin E1 or CDK2 ablation also re-sensitized palbociclib-resistant cells to cell cycle arrest (49). In the PALOMA-3 (ClinicalTrials.gov identifier: NCT01942135) trial, higher CCNE1 gene expression was associated with poor response to palbociclib (50). Similar unsatisfactory responses to palbociclib were also observed in patients with higher CCNE2 gene expression in preoperative-palbociclib (POP) trial (ClinicalTrials.gov identifier: NCT02008734) (51). In this context, CDK2 inhibitors may benefit CDK4/6 inhibitor-resistant tumors (52).

Regulation of microRNAs. MicroRNAs (miRNAs) regulate approximately 30% of human genes. More than 50% of these miRNAs are tumor-associated or located in fragile sites of chromosomes (53). Changes in miRNAs in tumor cells revealed an essential role in tumor development (53). While miRNAs could have been seen as a hallmark in BC (54), they are also related to resistance to chemotherapy and endocrine therapy (53).

Currently, six miRNAs, miR-126, miR-326, miR3613-3p, miR29b-3p, miR-497, and miR-17-92, have been suggested to be associated with sensitivity to CDK4/6 inhibitors (55-57). miR-126 was also found to be a modulator of CDK4/6 inhibitors, as its transfection increased anti-tumor activity of CDK 4/6 inhibitor (58). Conversely, six miRNAs, miR-193b, miR-432-5p, miR-200a, miR-223, Let-7a and miR-21, are associated with resistance to CDK4/6 inhibitors (55, 59). CDK4/6 inhibitor resistance was mediated by miR-432-5p through the suppression of the TGF-β pathway and induction of CDK6 expression (60). Down-regulation of miR-223 contributed to HR-positive BC resistance to CDK4/6 inhibitors (59). Other miRNAs, such as miR-124a, miR9, miR200b, and miR-106b mediate cellular response to CDK4/6 inhibitors without affecting sensitivity to treatment (55).

The long noncoding RNA TROJAN has been found to promote HR-positive BC proliferation by regulating the G1/S transition. An antisense oligonucleotide of TROJAN has been shown to impair tumor cell proliferation. Moreover, combining an anti-TROJAN antisense oligonucleotide with palbociclib significantly enhanced the efficacy of palbociclib in HR-positive BC (61).

Loss of ER expression. In HR-positive BC, ER signaling predominantly induces the activation of cyclin D1 and CDK4/6 (16). Therefore, CDK4/6 inhibitors have been widely used with ER-related endocrine drugs, such as fulvestrant, tamoxifen, and aromatase inhibitors. The current understanding is that resistance to CDK4/6 inhibitors may be related to the down-regulation of cyclin D1 due to the loss of ER (62). Resistance to abemaciclib has been reported to be associated with the loss of cyclin D1 and ER/PR expression (12). Additionally, CDK6 over-expression may reduce the effectiveness of ER antagonists and lead to CDK4/6 inhibitor resistance by reducing ER expression (12, 63).

Acquired Resistance Mechanism

Overactivation of PI3K–AKT–mTOR signaling. The phosphatidylinositol 3-kinase (PI3K; also known as PIK3CA)–AKT–mammalian target of rapamycin (mTOR) signaling pathway is involved in tumor cell growth and progression and is activated in almost 40% of BC cases (64, 65). In HR-positive BC, ER expression can be induced by activating PI3K–AKT–mTOR signaling, which drives endocrine therapy resistance (66). Furthermore, activation of the PI3K–AKT–mTOR pathway can stabilize CDK4/6, leading to resistance to CD4/6 inhibitors (33, 49, 66). The loss of phosphatase and tensin homolog (PTEN) expression can increase AKT activation and decrease p27 expression, leading to excessive activation of CDK4 and inducing CDK4/6 inhibitor resistance (38). However, CDK4 can phosphorylate the tumor suppressor folliculin (FLCN) and activate the mammalian target of rapamycin complex 1 (mTORC1), which is associated with cancer progression (67). Furthermore, CDK4/6 inhibitors tend to activate PI3K–AKT–mTOR pathway by phosphoinositide-dependent kinase-1 (PDK-1) (33, 68), further leading to drug resistance.

PI3K mutation. Because overactivation of the PI3K–AKT–mTOR pathway has been linked to BC oncogenesis, pharmacological targeting of PI3K by PI3K inhibitors, such as alpelisib, has shown significant benefits in HR-positive, endocrine therapy-resistant BC patients (64). Treatment with alpelisib-fulvestrant has also prolonged progression-free survival among HR-positive BC patients with PI3K mutation and prior endocrine therapy failure (69). While inhibition of PI3K and mTOR has been shown to restore treatment sensitivity of CDK4/6 inhibitor-resistant BC cells (70-72), PI3K inhibitors have been implicated in the prevention of early CDK4/6 inhibitor resistance by down-regulating cyclin D1 expression (49). However, an assessment of the genetic aberrations of CDK4/6 inhibitor-resistant BC showed that PI3K mutations are more frequent than Rb mutations (29). Therefore, the combination of PI3K inhibitors or mTOR inhibitors with CDK4/6 inhibitors may require more clinical trials before it can be used as a standard strategy in the future. Currently, triple combination therapy with CDK4/6 inhibitors, PI3K inhibitors, and endocrine therapy is being investigated in patients whose tumors progress after CDK4/6 inhibitor treatment. Results from this trial suggested that such a treatment strategy is a promising strategy for patients with pre-treated, PI3K-mutated, HR-positive BC (73).

Defects of the ATR-Chk1 and ATM–Chk2 pathway. The ataxia telangiectasia mutated (ATM) and checkpoint kinase-2 (Chk2) pathways are critical in DNA damage repair (74). The ATM recognizes double-strand breaks, whereas the ataxia telangiectasia and Rad3-related protein (ATR) recognize single-strand breaks. Upon recruitment to DNA damage sites, ATM and ATR activate the checkpoint kinase 2 (Chk2) and Chk1. Both pathways lead to the inhibition of CDC25A, a phosphatase that hampers the inhibition of CDK4/6 and CDK2, leading to the blockage of cell entry into cell cycle checkpoints (74). A recent study showed that defects in the ATM-Chk2 pathway and single-strand break repair in HR-positive BC can drive endocrine therapy resistance (75). Moreover, the efficacy of endocrine agents in HR-positive BC depends on both the ATM and Chk2 expression; inactivation of either ATM or Chk2 prevents ER inhibition-mediated cell cycle arrest (76). Because the ATM-Chk2 pathway is highly associated with CDK4/6 (74), a defect of the ATM-Chk2 pathway may also be associated with CDK4/6 inhibitor resistance. In addition, Chk1 also stimulates WEE1 G2 checkpoint kinase (WEE1), a serine/threonine protein kinase that suppresses the function of CDK1; therefore, defects in the ATR-Chk1 pathway or over-expression of WEE1 may allow the cells to undergo cell cycle, lowering the efficiency of CDK4/6 inhibitors (77).

Alterations of Hippo pathway. In human breast cells, the Hippo pathway has been linked with ER to regulate breast cell development (78, 79). In the Hippo pathway, mammalian sterile 20-like kinase (MST)-1/2 and large tumor suppressor kinase (LATS)-1/2, phosphorylate the yes-associated protein 1 (YAP1) and WW domain containing transcription regulator 1 (WWTR1; also known as TAZ), resulting in the inhibition of cell growth (80). Similarly, tumors that lack MST-1/2 and LATS-1/2 fail to inhibit YAP1 and TAZ (79, 81). The Hippo pathway is associated with the development and progression of BC and has emerged as a keystone in therapy resistance (82, 83). It has been suggested, directly or indirectly, that alternations in the Hippo pathway are associated with CDK4/6 inhibitor resistance. Transcriptional enhanced associate domain (TEAD), a regulatory transcription factor in the Hippo pathway, has been shown to induce CDK6 expression (84). The loss of the FAT atypical cadherin 1 (FAT1) gene, which encodes a proto-cadherin, is associated with CDK4/6 inhibitor resistance mediated by YAP/TAZ nuclear localization and CDK6 over-expression in ER-positive breast cancer (37).

FGFR1 activation. The fibroblast growth factor receptor (FGFR) activation is involved in the proliferation and survival of HR-positive BC (85). Similar to other mitogenic signalings, FGFR links cyclin D and CDK4/6. Of the five FGFRs, FGFR1 is associated with resistance to CDK4/6 inhibitor. FGFR1 activates the PI3K–AKT–mTOR and RAS–MEK–ERK signaling pathways (86). FGFR1 over-expression was shown to mediate resistance to palbociclib or ribociclib (87, 88), which can be reversed by the FGFR tyrosine kinase inhibitor (TKI), lucitanib (89). In addition, treatment of tumors with increased FGFR2 expression with the combination of palbociclib and letrozole has been associated with higher progression-free survival (90). FGFR1/2 amplification or activating mutations were also identified in ctDNA in patients with progression after CDK4/6 inhibitors treatment (67).

Overactivation of MAPK signaling pathway. The mitogen-activated protein kinase (MAPK) pathway is one of the signaling pathways downstream of FGFR activation. The MAPK is composed of six distinct kinase groups: extracellular signal-regulated kinase (ERK)-1/2, ERK-3/4, ERK-5, ERK-7/8, Jun N-terminal kinase (JNK)-1/2/3 and the p38; therefore, the MAPK pathway is also known as the RAS/RAF/MEK/ERK pathway (91). These pathways interfere with the cell cycle machinery at many stages. Among these, ERK signaling is the best-studied MAPK pathway. ERK signaling is activated by extracellular signals, such as growth factors and mitogens, which are particularly relevant to cancer. These signals phosphorylate RAS and subsequently lead to RAF phosphorylation. Activated RAF, in turn, phosphorylates the kinases MEK-1/2, which then phosphorylates ERK-1/2 (91).

Transcriptional regulation of cell cycle components by ERK links CDKs to the ERK signaling. Whereas ERK signaling stimulates cyclin D1 expression upon growth factor exposure, p38 signaling suppresses cyclin D1 expression (91). Therefore, the alternation of cyclin D levels largely affects their cell cycle partners, CDK4 and CDK6, leading to acquired resistance to CDK4/6 inhibitor (92).

Immune evasion. CDK4/6 inhibitors promote anti-tumor immunity, while stimulating the interferon-lambda (IFN-λ) and increasing the presentation of tumor antigens (93, 94). CDK4/6 inhibitors also inhibit the proliferation of regulatory T cells, which hinders immune surveillance against tumor cells (94). In addition, CDK4/6 inhibitors promote cytotoxic T-cell-mediated clearance of tumor cells (94).

However, immune evasion or alterations in the tumor microenvironment eventually leads to resistance to CDK4/6 inhibitor (94). Cytokine array analysis suggested that chemokine (C-C motif) ligand 5 (CCL-5) was up-regulated in palbociclib-treated cancer cells (95), which may be related to immune invasion (96). Moreover, RNA sequencing analysis suggested enriched IFN-α and IFN-β expression in CDK4/6 inhibitor-resistant BC cells (97). Recently, transforming growth factor-beta (TGF-β) has also been suggested to induce the expression of epithelial-mesenchymal transition (EMT)-related genes via the suppressor of mothers against decapentaplegic (SMAD) and the PI3K/AKT/mTOR pathway (98, 99), which may therefore lead to the insensitivity to CDK4/6 inhibitors.

Other Possible Mechanisms

A study has revealed that the loss of CDK inhibitors p21 and p27, which targeted CDK2, could lead to palbociclib insensitivity (100). Furthermore, p21 can be inhibited by histone deacetylase (HDAC), leading to cell cycle progression; therefore, over-expression of HDAC could also lead to CDK4/6 inhibitor resistance (101). The NeoPalAna trial (ClinicalTrials.gov Identifier: NCT01723774) has identified that p19, which is transcribed by E2F after phosphorylation of Rb, was over-expressed in patients with palbociclib resistance (35, 102). In addition to CDK4/6 over-expression, as mentioned earlier, CDK7 over-expression may also contribute to drug resistance, as CDK7 may activate CDK4/6, promoting G1 progression (103). This has also been observed in a mouse model, showing up-regulation of CDK7 after palbociclib treatment (104). Thymidine kinase and CDK9 levels in plasma-derived exosomes have been shown to play a role in reducing effectiveness of palbociclib (105). Overactivation of androgen receptor (AR) has been suggested in palbociclib-resistant BC cells (106), where dual inhibition of AR could reverse palbociclib resistance.

Potential Strategies to Overcome CDK4/6 Inhibitor Resistance in the Future

Use of CDK4/6 inhibitor with other clinically available drugs or other target therapeutic drugs. Although CDK4/6 inhibitors are currently used with endocrine therapy for HR-positive BC (107), their persistent use may induce acquired-tumor drug resistance. Yet, interestingly, combination therapies with other drugs have been shown to inhibit this adaptive response.

Currently, several clinical trials have investigated combination treatment in BC patients with CDK4/6 inhibitor resistance, and most of them have resulted in a promising result (Table II). For example, the MAINTAIN trial (ClinicalTrials.gov identifier: NCT02632045) has suggested a prolonged progression-free survival in CDK4/6 inhibitor-resistance patients receiving ribociclib combined with endocrine therapy, compared to ribociclib monotherapy, therefore offering a potential therapeutic benefit of continuing on CDK 4/6 inhibitors with endocrine therapy (108). Although the elderly population with BC remains excluded from most clinical trials, the beneficial effects of combined therapy can still be observed in these patients in some studies (109). The EMERALD trial (ClinicalTrials.gov identifier: NCT03778931) showed that elacestrant was more effective than aromatase inhibitor in BC patients who had advanced following prior co-therapy with CDK4/6 inhibitors and endocrine therapy (110, 111). The TRINITI-1 trial (ClinicalTrials.gov identifier: NCT02732119) with aromatase inhibitors-resistant patients following progression after treatment with CDK4/6 inhibitors showed clinical benefits of triplet treatment combination of ribociclib, everolimus, and exemestane; further supporting the idea that therapy targeting both CDK4/6, PI3K, and mTOR may show therapeutic benefits to CDK4/6 inhibitor resistant patients (112).

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table II.

On-going clinical trials on patients with CDK4/6 inhibitors resistance.

Current target therapeutic drugs for BC focus mainly on the PI3K–AKT–mTOR signaling. In cell models, the use of PI3K inhibitor with palbociclib delayed the recommencement of S phase entry and abolished the accumulation of cyclin D1, consistent with the role of PI3K signaling in promoting cyclin D1 expression (49). Another study showed that the mTOR inhibitors synergized with CDK4/6 inhibitors to prevent the resumption of breast cancer proliferation, and this regimen induced a significant suppression of E2F genes (71). In addition, activated AKT recruits and activates more PI3K on the cell membrane. In turn, activated PI3K phosphorylates AKT, leading to a feedback amplification loop (113). Therefore, AKT inhibitors may become one of the treatment choices, especially when patients are PI3K-mutated or unresponsive to PI3K inhibitors. In the ongoing TAKTIC trial (ClinicalTrials.gov Identifier: NCT03959891), the efficacy of ipatasertib, an AKT-1 inhibitor, was evaluated in combination with endocrine therapy and palbociclib in HR-positive patients who had failed prior CDK4/6 inhibitor treatment. The results so far have suggested a possible intervention for CDK4/6 inhibitor-resistant patients (114).

Histone deacetylase (HDAC) can increase the activation of CDK4/6 and inhibit cell cycle arrest by suppressing p21 expression in CDK4/6 inhibitor-resistant tumors (101). In terms of this, HDAC inhibitors can inhibit BC proliferation (115), induce apoptosis (116), and suppress DNA repair (117), producing an anti-cancer effect. In the ACE trial (ClinicalTrials.gov Identifier: NCT02482753), the use of chidamide, an HDAC inhibitor, with exemestane significantly improved progression-free survival in CDK4/6 inhibitors-resistant patients (118).

While the anti-apoptotic protein B-cell lymphoma (BCL)-2 is highly responsive to estrogen (119), targeting BCL-2 may be a potential option for CDK4/6 inhibitor resistance. The ongoing VERONICA trial (ClinicalTrials.gov Identifier: NCT03584009) has indicated that BCL-2 inhibition, although not statically significant, may be expected to be a treatment option for CDK4/6 inhibitor-resistant patients (120).

Last but not least, CDK4/6 inhibitors increase the expression of PD-L1 raising the possibility of using CDK4/6 inhibitors as a priming approach to attract T cells into immune-cold, T cell-excluded tumors because pre-existing tumor-infiltrating CD8+ T cells are more likely to respond to anti-PD-1 (121). Studies using a MMTV-rtTA/tetO-HER2 transgenic mouse model have shown that CDK4/6 inhibitors may enhance the susceptibility of tumors to PD-1/PD-L1 blockade, through the promotion of the CD8+ T cell-mediated clearance of cancer cells (94). Further investigation has suggested that the addition of PD-L1 inhibitors to CDK4/6 inhibitors suppresses cell-cycle gene-mediated proliferation and increases MHC expression in BC cells. Such combination immunotherapy also increases the activation of T-cells, macrophages, and dendritic cells, leading to higher efficiency of antigen presentation (121). Several ongoing clinical trials using CDK4/6 inhibitors in combination with PD-L1 inhibitors, aromatase inhibitors, or selective estrogen-receptor degraders in hormone-positive BC patients (ClinicalTrials.gov Identifier: NCT02778685; NCT02778685; NCT03294694) may provide us with a better understanding of this immune-priming effects in the future. However, since cytotoxic T cells depend on CDKs [6], the long-term effects of CDK4/6 inhibitors on T cell proliferation and activation should be carefully monitored.

Traditional Chinese herbs. Because of the different mechanisms in BC resistance to CDK4/6 inhibitors, finding a new drug may be a more realistic approach to deal with this clinically significant malignancy. Traditional Chinese medicine (TCM) has been widely studied for cancer treatment in recent years. These include crude extracts or active compounds derived from plants (122).

For example, extracts of rhizome bollbostemmatis could induce DNA fragmentation in a three-dimensional BC culture (123). Germacrone extracted from Rhizma curcuma can induce apoptosis of doxorubicin-resistant MCF-7 cells (124). In addition, active compounds, such as ganoderic acid, emodin azide methyl-anthraquinone derivative, and quercetin can suppress BC cell proliferation and induce apoptosis (125, 126). Because many TCMs can modulate the host’s immunity, they may complement immunotherapy.

In addition, several reports have suggested that complementary use of TCMs during cytotoxic chemotherapy may reduce adverse effects, prolong patients’ survival, and prevent recurrence (127). However, despite no evidence regarding TCM treatment in CDK4/6 inhibitor-resistant BC, this has put forward a future potential therapeutic approach.

Other alternative drugs. Triazoles and their derivatives have also been used extensively as anti-cancer, anti-viral, anti-microbial, anti-inflammatory, and anti-oxidant agents (128). 1,2,3-triazole derivatives have displayed high cytotoxicity against doxorubicin-resistant MCF-7 cells (129). Because of the usefulness of triazoles and the flavonoid compound isolated from TCM, several triazole-bridged flavonoid dimers [such as Ac22(Az8)2 and Ac15(Az8)2] have recently been synthesized as a highly selective breast cancer resistance protein (BCRP) inhibitors (130, 131), revealing that triazole-based compounds might also be promising candidates for future BC treatment.

Conclusion

CDK4/6 inhibitors are currently the effectual standard therapy in combination with endocrine therapy in HR-positive BC. Despite that, issues regarding resistance to CDK4/6 inhibitors in a clinical setting is an inevitable issue. Current knowledge of the molecular mechanisms of CDK4/6 inhibitor resistance is far from complete, as it is based mainly on single-agent studies or cell line models. As such a resistance problem may arise from different intrinsic or acquired mechanisms, future studies on the current topic are therefore recommended to elucidate the mechanism. At the same time, newer therapeutic options or alternative drugs may require continuous development to combat resistance.

Footnotes

  • Authors’ Contributions

    CMC and HYPL made substantial contributions to the concept, literature research and article writing. HYPL was involved in the production of figures and revising the article. All Authors gave their final approval of the version to be published.

  • Conflicts of Interest

    The Authors declare that there are no conflicts of interest related to the manuscript.

  • Funding

    This work was financially supported by Buddhist Tzu Chi Medical Foundation (Grant number: TCMMP110-01-03(112)). The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

  • Received September 15, 2023.
  • Revision received October 17, 2023.
  • Accepted October 18, 2023.
  • Copyright © 2023 The Author(s). Published by the International Institute of Anticancer Research.

This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY-NC-ND) 4.0 international license (https://creativecommons.org/licenses/by-nc-nd/4.0).

References

  1. ↵
    1. Bray F,
    2. Ferlay J,
    3. Soerjomataram I,
    4. Siegel RL,
    5. Torre LA,
    6. Jemal A
    : Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68(6): 394-424, 2018. DOI: 10.3322/caac.21492
    OpenUrlCrossRefPubMed
  2. ↵
    1. Anderson WF,
    2. Chatterjee N,
    3. Ershler WB,
    4. Brawley OW
    : Estrogen receptor breast cancer phenotypes in the surveillance, epidemiology, and end results database. Breast Cancer Res Treat 76(1): 27-36, 2002. DOI: 10.1023/a:1020299707510
    OpenUrlCrossRefPubMed
  3. ↵
    1. Turner NC,
    2. Neven P,
    3. Loibl S,
    4. Andre F
    : Advances in the treatment of advanced oestrogen-receptor-positive breast cancer. Lancet 389(10087): 2403-2414, 2017. DOI: 10.1016/S0140-6736(16)32419-9
    OpenUrlCrossRefPubMed
  4. ↵
    1. Lin H,
    2. Yang G,
    3. Yu J,
    4. Wang J,
    5. Li Q,
    6. Guo S,
    7. Cao B
    : KDM5c inhibits multidrug resistance of colon cancer cell line by down-regulating ABCC1. Biomed Pharmacother 107: 1205-1209, 2018. DOI: 10.1016/j.biopha.2018.08.041
    OpenUrlCrossRefPubMed
  5. ↵
    1. Osborne CK,
    2. Schiff R
    : Mechanisms of endocrine resistance in breast cancer. Annu Rev Med 62: 233-247, 2011. DOI: 10.1146/annurev-med-070909-182917
    OpenUrlCrossRefPubMed
  6. ↵
    1. Ghafouri-Fard S,
    2. Khoshbakht T,
    3. Hussen BM,
    4. Dong P,
    5. Gassler N,
    6. Taheri M,
    7. Baniahmad A,
    8. Dilmaghani NA
    : A review on the role of cyclin dependent kinases in cancers. Cancer Cell Int 22(1): 325, 2022. DOI: 10.1186/s12935-022-02747-z
    OpenUrlCrossRefPubMed
  7. ↵
    1. Hui R,
    2. de Boer R,
    3. Lim E,
    4. Yeo B,
    5. Lynch J
    : CDK4/6 inhibitor plus endocrine therapy for hormone receptor-positive, HER2-negative metastatic breast cancer: The new standard of care. Asia Pac J Clin Oncol 17(Suppl 1): 3-14, 2021. DOI: 10.1111/ajco.13555
    OpenUrlCrossRef
  8. ↵
    1. Li Z,
    2. Zou W,
    3. Zhang J,
    4. Zhang Y,
    5. Xu Q,
    6. Li S,
    7. Chen C
    : Mechanisms of CDK4/6 inhibitor resistance in luminal breast cancer. Front Pharmacol 11: 580251, 2020. DOI: 10.3389/fphar.2020.580251
    OpenUrlCrossRefPubMed
  9. ↵
    1. Ogata R,
    2. Kishino E,
    3. Saitoh W,
    4. Koike Y,
    5. Kurebayashi J
    : Resistance to cyclin-dependent kinase (CDK) 4/6 inhibitors confers cross-resistance to other CDK inhibitors but not to chemotherapeutic agents in breast cancer cells. Breast Cancer 28(1): 206-215, 2021. DOI: 10.1007/s12282-020-01150-8
    OpenUrlCrossRefPubMed
  10. ↵
    1. Schafer KA
    : The cell cycle: a review. Vet Pathol 35(6): 461-478, 1998. DOI: 10.1177/030098589803500601
    OpenUrlCrossRefPubMed
  11. ↵
    1. Romero-Pozuelo J,
    2. Figlia G,
    3. Kaya O,
    4. Martin-Villalba A,
    5. Teleman AA
    : Cdk4 and Cdk6 couple the cell-cycle machinery to cell growth via mTORC1. Cell Rep 31(2): 107504, 2020. DOI: 10.1016/j.celrep.2020.03.068
    OpenUrlCrossRefPubMed
  12. ↵
    1. Yang C,
    2. Li Z,
    3. Bhatt T,
    4. Dickler M,
    5. Giri D,
    6. Scaltriti M,
    7. Baselga J,
    8. Rosen N,
    9. Chandarlapaty S
    : Acquired CDK6 amplification promotes breast cancer resistance to CDK4/6 inhibitors and loss of ER signaling and dependence. Oncogene 36(16): 2255-2264, 2017. DOI: 10.1038/onc.2016.379
    OpenUrlCrossRefPubMed
  13. ↵
    1. Hamilton E,
    2. Infante JR
    : Targeting CDK4/6 in patients with cancer. Cancer Treat Rev 45: 129-138, 2016. DOI: 10.1016/j.ctrv.2016.03.002
    OpenUrlCrossRefPubMed
  14. ↵
    1. Dickson C,
    2. Fantl V,
    3. Gillett C,
    4. Brookes S,
    5. Bartek J,
    6. Smith R,
    7. Fisher C,
    8. Barnes D,
    9. Peters G
    : Amplification of chromosome band 11q13 and a role for cyclin D1 in human breast cancer. Cancer Lett 90(1): 43-50, 1995. DOI: 10.1016/0304-3835(94)03676-a
    OpenUrlCrossRefPubMed
  15. ↵
    1. Cancer Genome Atlas Network
    : Comprehensive molecular portraits of human breast tumours. Nature 490(7418): 61-70, 2012. DOI: 10.1038/nature11412
    OpenUrlCrossRefPubMed
  16. ↵
    1. Finn RS,
    2. Aleshin A,
    3. Slamon DJ
    : Targeting the cyclin-dependent kinases (CDK) 4/6 in estrogen receptor-positive breast cancers. Breast Cancer Res 18(1): 17, 2016. DOI: 10.1186/s13058-015-0661-5
    OpenUrlCrossRefPubMed
  17. ↵
    1. Morita M,
    2. Ooe A,
    3. Ishii W,
    4. Watanabe A,
    5. Matsui C,
    6. Okuyama Y,
    7. Kitano S,
    8. Kato C,
    9. Onishi M,
    10. Sakaguchi K,
    11. Naoi Y
    : Optimal treatment of hormone receptor-positive advanced breast cancer patients with palbociclib. Anticancer Res 43(6): 2783-2789, 2023. DOI: 10.21873/anticanres.16447
    OpenUrlAbstract/FREE Full Text
  18. ↵
    1. Braal CL,
    2. Jongbloed EM,
    3. Wilting SM,
    4. Mathijssen RHJ,
    5. Koolen SLW,
    6. Jager A
    : Inhibiting CDK4/6 in breast cancer with palbociclib, ribociclib, and abemaciclib: Similarities and differences. Drugs 81(3): 317-331, 2021. DOI: 10.1007/s40265-020-01461-2
    OpenUrlCrossRefPubMed
  19. ↵
    1. Cristofanilli M,
    2. Turner NC,
    3. Bondarenko I,
    4. Ro J,
    5. Im SA,
    6. Masuda N,
    7. Colleoni M,
    8. Demichele A,
    9. Loi S,
    10. Verma S,
    11. Iwata H,
    12. Harbeck N,
    13. Zhang K,
    14. Theall KP,
    15. Jiang Y,
    16. Bartlett CH,
    17. Koehler M,
    18. Slamon D
    : Fulvestrant plus palbociclib versus fulvestrant plus placebo for treatment of hormone-receptor-positive, HER2-negative metastatic breast cancer that progressed on previous endocrine therapy (PALOMA-3): final analysis of the multicentre, double-blind, phase 3 randomised controlled trial. Lancet Oncol 17(4): 425-439, 2016. DOI: 10.1016/s1470-2045(15)00613-0
    OpenUrlCrossRefPubMed
  20. ↵
    1. Finn RS,
    2. Martin M,
    3. Rugo HS,
    4. Jones S,
    5. Im SA,
    6. Gelmon K,
    7. Harbeck N,
    8. Lipatov ON,
    9. Walshe JM,
    10. Moulder S,
    11. Gauthier E,
    12. Lu DR,
    13. Randolph S,
    14. Diéras V,
    15. Slamon DJ
    : Palbociclib and letrozole in advanced breast cancer. N Engl J Med 375(20): 1925-1936, 2016. DOI: 10.1056/NEJMoa1607303
    OpenUrlCrossRefPubMed
    1. Sledge GW Jr.,
    2. Toi M,
    3. Neven P,
    4. Sohn J,
    5. Inoue K,
    6. Pivot X,
    7. Burdaeva O,
    8. Okera M,
    9. Masuda N,
    10. Kaufman PA,
    11. Koh H,
    12. Grischke EM,
    13. Frenzel M,
    14. Lin Y,
    15. Barriga S,
    16. Smith IC,
    17. Bourayou N,
    18. 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 35(25): 2875-2884, 2017. DOI: 10.1200/jco.2017.73.7585
    OpenUrlCrossRefPubMed
    1. Park YH,
    2. Kim TY,
    3. Kim GM,
    4. Kang SY,
    5. Park IH,
    6. Kim JH,
    7. Lee KE,
    8. Ahn HK,
    9. Lee MH,
    10. Kim HJ,
    11. Kim HJ,
    12. Lee JI,
    13. Koh SJ,
    14. Kim JY,
    15. Lee KH,
    16. Sohn J,
    17. Kim SB,
    18. Ahn JS,
    19. Im YH,
    20. Jung KH,
    21. Im SA, Korean Cancer Study Group (KCSG)
    : Palbociclib plus exemestane with gonadotropin-releasing hormone agonist versus capecitabine in premenopausal women with hormone receptor-positive, her2-negative metastatic breast cancer (kcsg-br15-10): A multicentre, open-label, randomised, phase 2 trial. Lancet Oncol 20(12): 1750-1759, 2019. DOI: 10.1016/s1470-2045(19)30565-0
    OpenUrlCrossRefPubMed
    1. Tripathy D,
    2. Im SA,
    3. Colleoni M,
    4. Franke F,
    5. Bardia A,
    6. Harbeck N,
    7. Hurvitz SA,
    8. Chow L,
    9. Sohn J,
    10. Lee KS,
    11. Campos-Gomez S,
    12. Villanueva Vazquez R,
    13. Jung KH,
    14. Babu KG,
    15. Wheatley-Price P,
    16. De Laurentiis M,
    17. Im YH,
    18. Kuemmel S,
    19. El-Saghir N,
    20. Liu MC,
    21. Carlson G,
    22. Hughes G,
    23. Diaz-Padilla I,
    24. Germa C,
    25. Hirawat S,
    26. Lu YS
    : Ribociclib plus endocrine therapy for premenopausal women with hormone-receptor-positive, advanced breast cancer (MONALEESA-7): a randomised phase 3 trial. Lancet Oncol 19(7): 904-915, 2018. DOI: 10.1016/s1470-2045(18)30292-4
    OpenUrlCrossRefPubMed
  21. ↵
    1. Sledge GW Jr.,
    2. Toi M,
    3. Neven P,
    4. Sohn J,
    5. Inoue K,
    6. Pivot X,
    7. Burdaeva O,
    8. Okera M,
    9. Masuda N,
    10. Kaufman PA,
    11. Koh H,
    12. Grischke EM,
    13. Conte P,
    14. Lu Y,
    15. Barriga S,
    16. Hurt K,
    17. Frenzel M,
    18. Johnston S,
    19. 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 6(1): 116-124, 2020. DOI: 10.1001/jamaoncol.2019.4782
    OpenUrlCrossRefPubMed
  22. ↵
    1. Long F,
    2. He Y,
    3. Fu H,
    4. Li Y,
    5. Bao X,
    6. Wang Q,
    7. Wang Y,
    8. Xie C,
    9. Lou L
    : Preclinical characterization of SHR6390, a novel CDK 4/6 inhibitor, in vitro and in human tumor xenograft models. Cancer Sci 110(4): 1420-1430, 2019. DOI: 10.1111/cas.13957
    OpenUrlCrossRefPubMed
    1. Xu B,
    2. Zhang Q,
    3. Zhang P,
    4. Hu X,
    5. Li W,
    6. Tong Z,
    7. Sun T,
    8. Teng Y,
    9. Wu X,
    10. Ouyang Q,
    11. Yan X,
    12. Cheng J,
    13. Liu Q,
    14. Feng J,
    15. Wang X,
    16. Yin Y,
    17. Shi Y,
    18. Pan Y,
    19. Wang Y,
    20. Xie W,
    21. Yan M,
    22. Liu Y,
    23. Yan P,
    24. Wu F,
    25. Zhu X,
    26. Zou J, DAWNA-1 Study Consortium
    : Dalpiciclib or placebo plus fulvestrant in hormone receptor-positive and HER2-negative advanced breast cancer: a randomized, phase 3 trial. Nat Med 27(11): 1904-1909, 2021. DOI: 10.1038/s41591-021-01562-9
    OpenUrlCrossRefPubMed
  23. ↵
    1. Tan AR,
    2. Wright GS,
    3. Thummala AR,
    4. Danso MA,
    5. Popovic L,
    6. Pluard TJ,
    7. Han HS,
    8. Vojnović Ž,
    9. Vasev N,
    10. Ma L,
    11. Richards DA,
    12. Wilks ST,
    13. Milenković D,
    14. Xiao J,
    15. Sorrentino J,
    16. Horton J,
    17. O’Shaughnessy J
    : Trilaciclib prior to chemotherapy in patients with metastatic triple-negative breast cancer: Final efficacy and subgroup analysis from a randomized phase II study. Clin Cancer Res 28(4): 629-636, 2022. DOI: 10.1158/1078-0432.CCR-21-2272
    OpenUrlCrossRefPubMed
  24. ↵
    1. Condorelli R,
    2. Spring L,
    3. O’Shaughnessy J,
    4. Lacroix L,
    5. Bailleux C,
    6. Scott V,
    7. Dubois J,
    8. Nagy RJ,
    9. Lanman RB,
    10. Iafrate AJ,
    11. Andre F,
    12. Bardia A
    : Polyclonal RB1 mutations and acquired resistance to CDK 4/6 inhibitors in patients with metastatic breast cancer. Ann Oncol 29(3): 640-645, 2018. DOI: 10.1093/annonc/mdx784
    OpenUrlCrossRef
  25. ↵
    1. O’Leary B,
    2. Cutts RJ,
    3. Liu Y,
    4. Hrebien S,
    5. Huang X,
    6. Fenwick K,
    7. André F,
    8. Loibl S,
    9. Loi S,
    10. Garcia-Murillas I,
    11. Cristofanilli M,
    12. Huang Bartlett C,
    13. Turner NC
    : The genetic landscape and clonal evolution of breast cancer resistance to palbociclib plus fulvestrant in the PALOMA-3 trial. Cancer Discov 8(11): 1390-1403, 2018. DOI: 10.1158/2159-8290.CD-18-0264
    OpenUrlAbstract/FREE Full Text
  26. ↵
    1. Kong T,
    2. Xue Y,
    3. Cencic R,
    4. Zhu X,
    5. Monast A,
    6. Fu Z,
    7. Pilon V,
    8. Sangwan V,
    9. Guiot MC,
    10. Foulkes WD,
    11. Porco JA Jr.,
    12. Park M,
    13. Pelletier J,
    14. Huang S
    : eIF4A inhibitors suppress cell-cycle feedback response and acquired resistance to CDK4/6 inhibition in cancer. Mol Cancer Ther 18(11): 2158-2170, 2019. DOI: 10.1158/1535-7163.MCT-19-0162
    OpenUrlAbstract/FREE Full Text
  27. ↵
    1. Bollard J,
    2. Miguela V,
    3. Ruiz de Galarreta M,
    4. Venkatesh A,
    5. Bian CB,
    6. Roberto MP,
    7. Tovar V,
    8. Sia D,
    9. Molina-Sánchez P,
    10. Nguyen CB,
    11. Nakagawa S,
    12. Llovet JM,
    13. Hoshida Y,
    14. Lujambio A
    : Palbociclib (PD-0332991), a selective CDK4/6 inhibitor, restricts tumour growth in preclinical models of hepatocellular carcinoma. Gut 66(7): 1286-1296, 2017. DOI: 10.1136/gutjnl-2016-312268
    OpenUrlAbstract/FREE Full Text
  28. ↵
    1. Olanich ME,
    2. Sun W,
    3. Hewitt SM,
    4. Abdullaev Z,
    5. Pack SD,
    6. Barr FG
    : CDK4 amplification reduces sensitivity to CDK4/6 inhibition in fusion-positive rhabdomyosarcoma. Clin Cancer Res 21(21): 4947-4959, 2015. DOI: 10.1158/1078-0432.CCR-14-2955
    OpenUrlAbstract/FREE Full Text
  29. ↵
    1. Jansen VM,
    2. Bhola NE,
    3. Bauer JA,
    4. Formisano L,
    5. Lee KM,
    6. Hutchinson KE,
    7. Witkiewicz AK,
    8. Moore PD,
    9. Estrada MV,
    10. Sánchez V,
    11. Ericsson PG,
    12. Sanders ME,
    13. Pohlmann PR,
    14. Pishvaian MJ,
    15. Riddle DA,
    16. Dugger TC,
    17. Wei W,
    18. Knudsen ES,
    19. Arteaga CL
    : Kinome-wide RNA interference screen reveals a role for PDK1 in acquired resistance to CDK4/6 inhibition in ER-positive breast cancer. Cancer Res 77(9): 2488-2499, 2017. DOI: 10.1158/0008-5472.CAN-16-2653
    OpenUrlAbstract/FREE Full Text
  30. ↵
    1. Martin L-A,
    2. Pancholi S,
    3. Ribas R,
    4. Gao Q,
    5. Simigdala N,
    6. Nikitorowicz-Buniak J,
    7. Johnston S,
    8. Dowsett M
    : Resistance to palbociclib depends on multiple targetable mechanisms highlighting the potential of drug holidays and drug switching to improve therapeutic outcome. Cancer Res 77(4_Suppl): P3-03-09, 2017. DOI: 10.1158/1538-7445.SABCS16-P3-03-09
    OpenUrlCrossRef
  31. ↵
    1. Hunt KK,
    2. Karakas C,
    3. Ha MJ,
    4. Biernacka A,
    5. Yi M,
    6. Sahin AA,
    7. Adjapong O,
    8. Hortobagyi GN,
    9. Bondy M,
    10. Thompson P,
    11. Cheung KL,
    12. Ellis IO,
    13. Bacus S,
    14. Symmans WF,
    15. Do KA,
    16. Keyomarsi K
    : Cytoplasmic cyclin E predicts recurrence in patients with breast cancer. Clin Cancer Res 23(12): 2991-3002, 2017. DOI: 10.1158/1078-0432.CCR-16-2217
    OpenUrlAbstract/FREE Full Text
  32. ↵
    1. Kitajima S,
    2. Takahashi C
    : Intersection of retinoblastoma tumor suppressor function, stem cells, metabolism, and inflammation. Cancer Sci 108(9): 1726-1731, 2017. DOI: 10.1111/cas.13312
    OpenUrlCrossRefPubMed
  33. ↵
    1. Li Z,
    2. Razavi P,
    3. Li Q,
    4. Toy W,
    5. Liu B,
    6. Ping C,
    7. Hsieh W,
    8. Sanchez-Vega F,
    9. Brown DN,
    10. Da Cruz Paula AF,
    11. Morris L,
    12. Selenica P,
    13. Eichenberger E,
    14. Shen R,
    15. Schultz N,
    16. Rosen N,
    17. Scaltriti M,
    18. Brogi E,
    19. Baselga J,
    20. Reis-Filho JS,
    21. Chandarlapaty S
    : Loss of the FAT1 tumor suppressor promotes resistance to CDK4/6 inhibitors via the Hippo pathway. Cancer Cell 34(6): 893-905.e8, 2018. DOI: 10.1016/j.ccell.2018.11.006
    OpenUrlCrossRefPubMed
  34. ↵
    1. Costa C,
    2. Wang Y,
    3. Ly A,
    4. Hosono Y,
    5. Murchie E,
    6. Walmsley CS,
    7. Huynh T,
    8. Healy C,
    9. Peterson R,
    10. Yanase S,
    11. Jakubik CT,
    12. Henderson LE,
    13. Damon LJ,
    14. Timonina D,
    15. Sanidas I,
    16. Pinto CJ,
    17. Mino-Kenudson M,
    18. Stone JR,
    19. Dyson NJ,
    20. Ellisen LW,
    21. Bardia A,
    22. Ebi H,
    23. Benes CH,
    24. Engelman JA,
    25. Juric D
    : PTEN loss mediates clinical cross-resistance to CDK4/6 and PI3Kα inhibitors in breast cancer. Cancer Discov 10(1): 72-85, 2020. DOI: 10.1158/2159-8290.Cd-18-0830
    OpenUrlAbstract/FREE Full Text
  35. ↵
    1. Wander SA,
    2. Cohen O,
    3. Gong X,
    4. Johnson GN,
    5. Buendia-Buendia JE,
    6. Lloyd MR,
    7. Kim D,
    8. Luo F,
    9. Mao P,
    10. Helvie K,
    11. Kowalski KJ,
    12. Nayar U,
    13. Waks AG,
    14. Parsons SH,
    15. Martinez R,
    16. Litchfield LM,
    17. Ye XS,
    18. Yu C,
    19. Jansen VM,
    20. Stille JR,
    21. Smith PS,
    22. Oakley GJ,
    23. Chu QS,
    24. Batist G,
    25. Hughes ME,
    26. Kremer JD,
    27. Garraway LA,
    28. Winer EP,
    29. Tolaney SM,
    30. Lin NU,
    31. Buchanan SG,
    32. Wagle N
    : The genomic landscape of intrinsic and acquired resistance to cyclin-dependent kinase 4/6 inhibitors in patients with hormone receptor-positive metastatic breast cancer. Cancer Discov 10(8): 1174-1193, 2020. DOI: 10.1158/2159-8290.CD-19-1390
    OpenUrlAbstract/FREE Full Text
  36. ↵
    1. Wu T,
    2. Wu L
    : The role and clinical implications of the retinoblastoma (RB)-E2F pathway in gastric cancer. Front Oncol 11: 655630, 2021. DOI: 10.3389/fonc.2021.655630
    OpenUrlCrossRefPubMed
  37. ↵
    1. Castellano D,
    2. Rubio C,
    3. Lopez-Calderon F,
    4. Segovia C,
    5. Duenas M,
    6. Martinez-Fernandez M,
    7. Otero I,
    8. Manneh R,
    9. de Velasco G,
    10. Paramio J
    : 8P Cdk4/6 inhibitor activity in metastatic bladder cancer cell lines is independently of RB1 status. Ann Oncol 27: ix3, 2016. DOI: 10.1016/S0923-7534(21)00170-8
    OpenUrlCrossRef
  38. ↵
    1. Xu B,
    2. Krie A,
    3. De P,
    4. Williams C,
    5. Elsey R,
    6. Klein J,
    7. Leyland-Jones B
    : Utilizing tumor and plasma liquid biopsy in treatment decision making for an estrogen receptor-positive advanced breast cancer patient. Cureus 9(6): e1408, 2017. DOI: 10.7759/cureus.1408
    OpenUrlCrossRef
  39. ↵
    1. Muss HB,
    2. Smitherman A,
    3. Wood WA,
    4. Nyrop K,
    5. Tuchman S,
    6. Randhawa PK,
    7. Entwistle AR,
    8. Mitin N,
    9. Shachar SS
    : p16 a biomarker of aging and tolerance for cancer therapy. Transl Cancer Res 9(9): 5732-5742, 2020. DOI: 10.21037/tcr.2020.03.39
    OpenUrlCrossRef
  40. ↵
    1. Witkiewicz AK,
    2. Knudsen KE,
    3. Dicker AP,
    4. Knudsen ES
    : The meaning of p16(ink4a) expression in tumors: functional significance, clinical associations and future developments. Cell Cycle 10(15): 2497-2503, 2011. DOI: 10.4161/cc.10.15.16776
    OpenUrlCrossRefPubMed
  41. ↵
    1. Elvin JA,
    2. Gay LM,
    3. Ort R,
    4. Shuluk J,
    5. Long J,
    6. Shelley L,
    7. Lee R,
    8. Chalmers ZR,
    9. Frampton GM,
    10. Ali SM,
    11. Schrock AB,
    12. Miller VA,
    13. Stephens PJ,
    14. Ross JS,
    15. Frank R
    : Clinical benefit in response to palbociclib treatment in refractory uterine leiomyosarcomas with a common CDKN2A alteration. Oncologist 22(4): 416-421, 2017. DOI: 10.1634/theoncologist.2016-0310
    OpenUrlAbstract/FREE Full Text
  42. ↵
    1. DeMichele A,
    2. Clark AS,
    3. Tan KS,
    4. Heitjan DF,
    5. Gramlich K,
    6. Gallagher M,
    7. Lal P,
    8. Feldman M,
    9. Zhang P,
    10. Colameco C,
    11. Lewis D,
    12. Langer M,
    13. Goodman N,
    14. Domchek S,
    15. Gogineni K,
    16. Rosen M,
    17. Fox K,
    18. O’dwyer P
    : CDK 4/6 inhibitor palbociclib (PD0332991) in Rb+ advanced breast cancer: Phase II activity, safety, and predictive biomarker assessment. Clin Cancer Res 21(5): 995-1001, 2015. DOI: 10.1158/1078-0432.Ccr-14-2258
    OpenUrlAbstract/FREE Full Text
  43. ↵
    1. Palafox M,
    2. Monserrat L,
    3. Bellet M,
    4. Villacampa G,
    5. Gonzalez-Perez A,
    6. Oliveira M,
    7. Brasó-Maristany F,
    8. Ibrahimi N,
    9. Kannan S,
    10. Mina L,
    11. Herrera-Abreu MT,
    12. Òdena A,
    13. Sánchez-Guixé M,
    14. Capelán M,
    15. Azaro A,
    16. Bruna A,
    17. Rodríguez O,
    18. Guzmán M,
    19. Grueso J,
    20. Viaplana C,
    21. Hernández J,
    22. Su F,
    23. Lin K,
    24. Clarke RB,
    25. Caldas C,
    26. Arribas J,
    27. Michiels S,
    28. García-Sanz A,
    29. Turner NC,
    30. Prat A,
    31. Nuciforo P,
    32. Dienstmann R,
    33. Verma CS,
    34. Lopez-Bigas N,
    35. Scaltriti M,
    36. Arnedos M,
    37. Saura C,
    38. Serra V
    : High p16 expression and heterozygous RB1 loss are biomarkers for CDK4/6 inhibitor resistance in ER(+) breast cancer. Nat Commun 13(1): 5258, 2022. DOI: 10.1038/s41467-022-32828-6
    OpenUrlCrossRefPubMed
  44. ↵
    1. Caldon CE,
    2. Sergio CM,
    3. Schütte J,
    4. Boersma MN,
    5. Sutherland RL,
    6. Carroll JS,
    7. Musgrove EA
    : Estrogen regulation of cyclin E2 requires cyclin D1 but not c-Myc. Mol Cell Biol 29(17): 4623-4639, 2009. DOI: 10.1128/MCB.00269-09
    OpenUrlAbstract/FREE Full Text
  45. ↵
    1. Herrera-Abreu MT,
    2. Palafox M,
    3. Asghar U,
    4. Rivas MA,
    5. Cutts RJ,
    6. Garcia-Murillas I,
    7. Pearson A,
    8. Guzman M,
    9. Rodriguez O,
    10. Grueso J,
    11. Bellet M,
    12. Cortés J,
    13. Elliott R,
    14. Pancholi S,
    15. Baselga J,
    16. Dowsett M,
    17. Martin LA,
    18. Turner NC,
    19. Serra V
    : Early adaptation and acquired resistance to CDK4/6 inhibition in estrogen receptor-positive breast cancer. Cancer Res 76(8): 2301-2313, 2016. DOI: 10.1158/0008-5472.CAN-15-0728
    OpenUrlAbstract/FREE Full Text
  46. ↵
    1. Turner NC,
    2. Liu Y,
    3. Zhu Z,
    4. Loi S,
    5. Colleoni M,
    6. Loibl S,
    7. DeMichele A,
    8. Harbeck N,
    9. André F,
    10. Bayar MA,
    11. Michiels S,
    12. Zhang Z,
    13. Giorgetti C,
    14. Arnedos M,
    15. Huang Bartlett C,
    16. Cristofanilli M
    : Cyclin E1 expression and palbociclib efficacy in previously treated hormone receptor-positive metastatic breast cancer. J Clin Oncol 37(14): 1169-1178, 2019. DOI: 10.1200/JCO.18.00925
    OpenUrlCrossRefPubMed
  47. ↵
    1. Arnedos M,
    2. Bayar M,
    3. Cheaib B,
    4. Scott V,
    5. Bouakka I,
    6. Valent A,
    7. Adam J,
    8. Leroux-Kozal V,
    9. Marty V,
    10. Rapinat A,
    11. Mazouni C,
    12. Sarfati B,
    13. Bieche I,
    14. Balleyguier C,
    15. Gentien D,
    16. Delaloge S,
    17. Lacroix-Triki M,
    18. Michiels S,
    19. Andre F
    : Modulation of Rb phosphorylation and antiproliferative response to palbociclib: the preoperative-palbociclib (POP) randomized clinical trial. Ann Oncol 29(8): 1755-1762, 2018. DOI: 10.1093/annonc/mdy202
    OpenUrlCrossRefPubMed
  48. ↵
    1. Caldon CE,
    2. Sergio CM,
    3. Kang J,
    4. Muthukaruppan A,
    5. Boersma MN,
    6. Stone A,
    7. Barraclough J,
    8. Lee CS,
    9. Black MA,
    10. Miller LD,
    11. Gee JM,
    12. Nicholson RI,
    13. Sutherland RL,
    14. Print CG,
    15. Musgrove EA
    : Cyclin e2 overexpression is associated with endocrine resistance but not insensitivity to cdk2 inhibition in human breast cancer cells. Mol Cancer Ther 11(7): 1488-1499, 2012. DOI: 10.1158/1535-7163.Mct-11-0963
    OpenUrlAbstract/FREE Full Text
  49. ↵
    1. Si W,
    2. Shen J,
    3. Zheng H,
    4. Fan W
    : The role and mechanisms of action of microRNAs in cancer drug resistance. Clin Epigenetics 11(1): 25, 2019. DOI: 10.1186/s13148-018-0587-8
    OpenUrlCrossRefPubMed
  50. ↵
    1. Singh R,
    2. Mo YY
    : Role of microRNAs in breast cancer. Cancer Biol Ther 14(3): 201-212, 2013. DOI: 10.4161/cbt.23296
    OpenUrlCrossRefPubMed
  51. ↵
    1. Andrikopoulou A,
    2. Shalit A,
    3. Zografos E,
    4. Koutsoukos K,
    5. Korakiti AM,
    6. Liontos M,
    7. Dimopoulos MA,
    8. Zagouri F
    : MicroRNAs as potential predictors of response to CDK4/6 inhibitor treatment. Cancers (Basel) 13(16): 4114, 2021. DOI: 10.3390/cancers13164114
    OpenUrlCrossRefPubMed
    1. Ji W,
    2. Zhang W,
    3. Wang X,
    4. Shi Y,
    5. Yang F,
    6. Xie H,
    7. Zhou W,
    8. Wang S,
    9. Guan X
    : c-myc regulates the sensitivity of breast cancer cells to palbociclib via c-myc/miR-29b-3p/CDK6 axis. Cell Death Dis 11(9): 760, 2020. DOI: 10.1038/s41419-020-02980-2
    OpenUrlCrossRefPubMed
  52. ↵
    1. Yu Y,
    2. Liao H,
    3. Xie R,
    4. Zhang Y,
    5. Zheng R,
    6. Chen J,
    7. Zhang B
    : Overexpression of miRNA-3613-3p enhances the sensitivity of triple negative breast cancer to CDK4/6 inhibitor palbociclib. Front Oncol 10: 590813, 2020. DOI: 10.3389/fonc.2020.590813
    OpenUrlCrossRefPubMed
  53. ↵
    1. Baldassari F,
    2. Zerbinati C,
    3. Galasso M,
    4. Corrà F,
    5. Minotti L,
    6. Agnoletto C,
    7. Previati M,
    8. Croce CM,
    9. Volinia S
    : Screen for microRNA and drug interactions in breast cancer cell lines points to mir-126 as a modulator of CDK4/6 and PIK3CA inhibitors. Front Genet 9: 174, 2018. DOI: 10.3389/fgene.2018.00174
    OpenUrlCrossRefPubMed
  54. ↵
    1. Citron F,
    2. Segatto I,
    3. Vinciguerra GLR,
    4. Musco L,
    5. Russo F,
    6. Mungo G,
    7. D’Andrea S,
    8. Mattevi MC,
    9. Perin T,
    10. Schiappacassi M,
    11. Massarut S,
    12. Marchini C,
    13. Amici A,
    14. Vecchione A,
    15. Baldassarre G,
    16. Belletti B
    : Downregulation of miR-223 expression is an early event during mammary transformation and confers resistance to CDK4/6 inhibitors in luminal breast cancer. Cancer Res 80(5): 1064-1077, 2020. DOI: 10.1158/0008-5472.Can-19-1793
    OpenUrlAbstract/FREE Full Text
  55. ↵
    1. Cornell L,
    2. Wander SA,
    3. Visal T,
    4. Wagle N,
    5. Shapiro GI
    : MicroRNA-mediated suppression of the TGF-β pathway confers transmissible and reversible CDK4/6 inhibitor resistance. Cell Rep 26(10): 2667-2680.e7, 2019. DOI: 10.1016/j.celrep.2019.02.023
    OpenUrlCrossRefPubMed
  56. ↵
    1. Jin X,
    2. Ge LP,
    3. Li DQ,
    4. Shao ZM,
    5. Di GH,
    6. Xu XE,
    7. Jiang YZ
    : LncRNA TROJAN promotes proliferation and resistance to CDK4/6 inhibitor via CDK2 transcriptional activation in ER+ breast cancer. Mol Cancer 19(1): 87, 2020. DOI: 10.1186/s12943-020-01210-9
    OpenUrlCrossRefPubMed
  57. ↵
    1. Gong X,
    2. Litchfield LM,
    3. Webster Y,
    4. Chio L,
    5. Wong SS,
    6. Stewart TR,
    7. Dowless M,
    8. Dempsey J,
    9. Zeng Y,
    10. Torres R,
    11. Boehnke K,
    12. Mur C,
    13. Marugán C,
    14. Baquero C,
    15. Yu C,
    16. Bray SM,
    17. Wulur IH,
    18. Bi C,
    19. Chu S,
    20. Qian H,
    21. Iversen PW,
    22. Merzoug FF,
    23. Ye XS,
    24. Reinhard C,
    25. De Dios A,
    26. Du J,
    27. Caldwell CW,
    28. Lallena MJ,
    29. Beckmann RP,
    30. Buchanan SG
    : Genomic aberrations that activate D-type cyclins are associated with enhanced sensitivity to the CDK4 and CDK6 inhibitor abemaciclib. Cancer Cell 32(6): 761-776.e6, 2017. DOI: 10.1016/j.ccell.2017.11.006
    OpenUrlCrossRefPubMed
  58. ↵
    1. Main SC,
    2. Cescon DW,
    3. Bratman SV
    : Liquid biopsies to predict CDK4/6 inhibitor efficacy and resistance in breast cancer. Cancer Drug Resist 5(3): 727-748, 2022. DOI: 10.20517/cdr.2022.37
    OpenUrlCrossRefPubMed
  59. ↵
    1. Fusco N,
    2. Malapelle U,
    3. Fassan M,
    4. Marchiò C,
    5. Buglioni S,
    6. Zupo S,
    7. Criscitiello C,
    8. Vigneri P,
    9. Dei Tos AP,
    10. Maiorano E,
    11. Viale G
    : PIK3CA mutations as a molecular target for hormone receptor-positive, HER2-negative metastatic breast cancer. Front Oncol 11: 644737, 2021. DOI: 10.3389/fonc.2021.644737
    OpenUrlCrossRefPubMed
  60. ↵
    1. Paplomata E,
    2. O’Regan R
    : The PI3K/AKT/mTOR pathway in breast cancer: targets, trials and biomarkers. Ther Adv Med Oncol 6(4): 154-166, 2014. DOI: 10.1177/1758834014530023
    OpenUrlCrossRefPubMed
  61. ↵
    1. Miller TW,
    2. Rexer BN,
    3. Garrett JT,
    4. Arteaga CL
    : Mutations in the phosphatidylinositol 3-kinase pathway: role in tumor progression and therapeutic implications in breast cancer. Breast Cancer Res 13(6): 224, 2011. DOI: 10.1186/bcr3039
    OpenUrlCrossRefPubMed
  62. ↵
    1. Martínez-Carreres L,
    2. Puyal J,
    3. Leal-Esteban LC,
    4. Orpinell M,
    5. Castillo-Armengol J,
    6. Giralt A,
    7. Dergai O,
    8. Moret C,
    9. Barquissau V,
    10. Nasrallah A,
    11. Pabois A,
    12. Zhang L,
    13. Romero P,
    14. Lopez-Mejia IC,
    15. Fajas L
    : CDK4 regulates lysosomal function and mTORC1 activation to promote cancer cell survival. Cancer Res 79(20): 5245-5259, 2019. DOI: 10.1158/0008-5472.CAN-19-0708
    OpenUrlAbstract/FREE Full Text
  63. ↵
    1. Takeshita T,
    2. Yamamoto Y,
    3. Yamamoto-Ibusuki M,
    4. Tomiguchi M,
    5. Sueta A,
    6. Murakami K,
    7. Iwase H
    : Clinical significance of plasma cell-free DNA mutations in PIK3CA, AKT1, and ESR1 gene according to treatment lines in ER-positive breast cancer. Mol Cancer 17(1): 67, 2018. DOI: 10.1186/s12943-018-0808-y
    OpenUrlCrossRefPubMed
  64. ↵
    1. André F,
    2. Ciruelos E,
    3. Rubovszky G,
    4. Campone M,
    5. Loibl S,
    6. Rugo HS,
    7. Iwata H,
    8. Conte P,
    9. Mayer IA,
    10. Kaufman B,
    11. Yamashita T,
    12. Lu YS,
    13. Inoue K,
    14. Takahashi M,
    15. Pápai Z,
    16. Longin AS,
    17. Mills D,
    18. Wilke C,
    19. Hirawat S,
    20. Juric D
    : Alpelisib for PIK3CA-mutated, hormone receptor-positive advanced breast cancer. N Engl J Med 380(20): 1929-1940, 2019. DOI: 10.1056/NEJMoa1813904
    OpenUrlCrossRefPubMed
  65. ↵
    1. O’Brien NA,
    2. McDermott MSJ,
    3. Conklin D,
    4. Luo T,
    5. Ayala R,
    6. Salgar S,
    7. Chau K,
    8. DiTomaso E,
    9. Babbar N,
    10. Su F,
    11. Gaither A,
    12. Hurvitz SA,
    13. Linnartz R,
    14. Rose K,
    15. Hirawat S,
    16. Slamon DJ
    : Targeting activated PI3K/mTOR signaling overcomes acquired resistance to CDK4/6-based therapies in preclinical models of hormone receptor-positive breast cancer. Breast Cancer Res 22(1): 89, 2020. DOI: 10.1186/s13058-020-01320-8
    OpenUrlCrossRefPubMed
  66. ↵
    1. Michaloglou C,
    2. Crafter C,
    3. Siersbaek R,
    4. Delpuech O,
    5. Curwen JO,
    6. Carnevalli LS,
    7. Staniszewska AD,
    8. Polanska UM,
    9. Cheraghchi-Bashi A,
    10. Lawson M,
    11. Chernukhin I,
    12. McEwen R,
    13. Carroll JS,
    14. Cosulich SC
    : Combined inhibition of mTOR and CDK4/6 is required for optimal blockade of E2F function and long-term growth inhibition in estrogen receptor-positive breast cancer. Mol Cancer Ther 17(5): 908-920, 2018. DOI: 10.1158/1535-7163.MCT-17-0537
    OpenUrlAbstract/FREE Full Text
  67. ↵
    1. Xu H,
    2. Wang Y,
    3. Han Y,
    4. Wu Y,
    5. Wang J,
    6. Xu B
    : CDK4/6 inhibitors versus PI3K/AKT/mTOR inhibitors in women with hormone receptor-positive, HER2-negative metastatic breast cancer: An updated systematic review and network meta-analysis of 28 randomized controlled trials. Front Oncol 12: 956464, 2022. DOI: 10.3389/fonc.2022.956464
    OpenUrlCrossRefPubMed
  68. ↵
    1. Pascual J,
    2. Lim JS,
    3. Macpherson IR,
    4. Armstrong AC,
    5. Ring A,
    6. Okines AF,
    7. Cutts RJ,
    8. Herrera-abreu MT,
    9. Garcia-murillas I,
    10. Pearson A,
    11. Hrebien S,
    12. Gevensleben H,
    13. Proszek PZ,
    14. Hubank M,
    15. Hills M,
    16. King J,
    17. Parmar M,
    18. Prout T,
    19. Finneran L,
    20. Malia J,
    21. Swales KE,
    22. Ruddle R,
    23. Raynaud FI,
    24. Turner A,
    25. Hall E,
    26. Yap TA,
    27. Lopez JS,
    28. Turner NC
    : Triplet therapy with palbociclib, taselisib, and fulvestrant in PIK3CA-mutant breast cancer and doublet palbociclib and taselisib in pathway-mutant solid cancers. Cancer Discov 11(1): 92-107, 2021. DOI: 10.1158/2159-8290.CD-20-0553
    OpenUrlAbstract/FREE Full Text
  69. ↵
    1. Bartek J,
    2. Lukas J
    : Chk1 and Chk2 kinases in checkpoint control and cancer. Cancer Cell 3(5): 421-429, 2003. DOI: 10.1016/s1535-6108(03)00110-7
    OpenUrlCrossRefPubMed
  70. ↵
    1. Anurag M,
    2. Punturi N,
    3. Hoog J,
    4. Bainbridge MN,
    5. Ellis MJ,
    6. Haricharan S
    : Comprehensive profiling of DNA repair defects in breast cancer identifies a novel class of endocrine therapy resistance drivers. Clin Cancer Res 24(19): 4887-4899, 2018. DOI: 10.1158/1078-0432.CCR-17-3702
    OpenUrlAbstract/FREE Full Text
  71. ↵
    1. Haricharan S,
    2. Punturi N,
    3. Singh P,
    4. Holloway KR,
    5. Anurag M,
    6. Schmelz J,
    7. Schmidt C,
    8. Lei JT,
    9. Suman V,
    10. Hunt K,
    11. Olson JA Jr.,
    12. Hoog J,
    13. Li S,
    14. Huang S,
    15. Edwards DP,
    16. Kavuri SM,
    17. Bainbridge MN,
    18. Ma CX,
    19. Ellis MJ
    : Loss of MutL disrupts CHK2-dependent cell-cycle control through CDK4/6 to promote intrinsic endocrine therapy resistance in primary breast cancer. Cancer Discov 7(10): 1168-1183, 2017. DOI: 10.1158/2159-8290.CD-16-1179
    OpenUrlAbstract/FREE Full Text
  72. ↵
    1. Matheson CJ,
    2. Backos DS,
    3. Reigan P
    : Targeting WEE1 kinase in cancer. Trends Pharmacol Sci 37(10): 872-881, 2016. DOI: 10.1016/j.tips.2016.06.006
    OpenUrlCrossRefPubMed
  73. ↵
    1. Britschgi A,
    2. Duss S,
    3. Kim S,
    4. Couto JP,
    5. Brinkhaus H,
    6. Koren S,
    7. De Silva D,
    8. Mertz KD,
    9. Kaup D,
    10. Varga Z,
    11. Voshol H,
    12. Vissieres A,
    13. Leroy C,
    14. Roloff T,
    15. Stadler MB,
    16. Scheel CH,
    17. Miraglia LJ,
    18. Orth AP,
    19. Bonamy GM,
    20. Reddy VA,
    21. Bentires-Alj M
    : The Hippo kinases LATS1 and 2 control human breast cell fate via crosstalk with ERα. Nature 541(7638): 541-545, 2017. DOI: 10.1038/nature20829
    OpenUrlCrossRefPubMed
  74. ↵
    1. Zhang K,
    2. Qi H-X,
    3. Hu Z-M,
    4. Chang Y-N,
    5. Shi Z-M,
    6. Han X-H,
    7. Han Y-W,
    8. Zhang R-X,
    9. Zhang Z,
    10. Chen T,
    11. Hong W
    : YAP and TAZ take center stage in cancer. Biochemistry 54(43): 6555-6566, 2015. DOI: 10.1021/acs.biochem.5b01014
    OpenUrlCrossRefPubMed
  75. ↵
    1. Han Y
    : Analysis of the role of the Hippo pathway in cancer. J Transl Med 17(1): 116, 2019. DOI: 10.1186/s12967-019-1869-4
    OpenUrlCrossRefPubMed
  76. ↵
    1. Zanconato F,
    2. Battilana G,
    3. Cordenonsi M,
    4. Piccolo S
    : YAP/TAZ as therapeutic targets in cancer. Curr Opin Pharmacol 29: 26-33, 2016. DOI: 10.1016/j.coph.2016.05.002
    OpenUrlCrossRefPubMed
  77. ↵
    1. Gujral TS,
    2. Kirschner MW
    : Hippo pathway mediates resistance to cytotoxic drugs. Proc Natl Acad Sci USA 114(18): E3729-E3738, 2017. DOI: 10.1073/pnas.1703096114
    OpenUrlAbstract/FREE Full Text
  78. ↵
    1. Shi P,
    2. Feng J,
    3. Chen C
    : Hippo pathway in mammary gland development and breast cancer. Acta Biochim Biophys Sin (Shanghai) 47(1): 53-59, 2015. DOI: 10.1093/abbs/gmu114
    OpenUrlCrossRefPubMed
  79. ↵
    1. Xie Q,
    2. Chen J,
    3. Feng H,
    4. Peng S,
    5. Adams U,
    6. Bai Y,
    7. Huang L,
    8. Li J,
    9. Huang J,
    10. Meng S,
    11. Yuan Z
    : YAP/TEAD-mediated transcription controls cellular senescence. Cancer Res 73(12): 3615-3624, 2013. DOI: 10.1158/0008-5472.Can-12-3793
    OpenUrlAbstract/FREE Full Text
  80. ↵
    1. Sahores A,
    2. May M,
    3. Sequeira GR,
    4. Fuentes C,
    5. Jacobsen B,
    6. Lanari C,
    7. Lamb CA
    : Targeting FGFR with BGJ398 in breast cancer: Effect on tumor growth and metastasis. Curr Cancer Drug Targets 18(10): 979-987, 2018. DOI: 10.2174/1568009618666171214114706
    OpenUrlCrossRefPubMed
  81. ↵
    1. Roberto M,
    2. Astone A,
    3. Botticelli A,
    4. Carbognin L,
    5. Cassano A,
    6. D’Auria G,
    7. Fabbri A,
    8. Fabi A,
    9. Gamucci T,
    10. Krasniqi E,
    11. Minelli M,
    12. Orlandi A,
    13. Pantano F,
    14. Paris I,
    15. Pizzuti L,
    16. Portarena I,
    17. Salesi N,
    18. Scagnoli S,
    19. Scavina P,
    20. Tonini G,
    21. Vici P,
    22. Marchetti P
    : CDK4/6 inhibitor treatments in patients with hormone receptor positive, Her2 negative advanced breast cancer: Potential molecular mechanisms, clinical implications and future perspectives. Cancers (Basel) 13(2): 332, 2021. DOI: 10.3390/cancers13020332
    OpenUrlCrossRefPubMed
  82. ↵
    1. Formisano L,
    2. Stauffer KM,
    3. Young CD,
    4. Bhola NE,
    5. Guerrero-Zotano AL,
    6. Jansen VM,
    7. Estrada MM,
    8. Hutchinson KE,
    9. Giltnane JM,
    10. Schwarz LJ,
    11. Lu Y,
    12. Balko JM,
    13. Deas O,
    14. Cairo S,
    15. Judde JG,
    16. Mayer IA,
    17. Sanders M,
    18. Dugger TC,
    19. Bianco R,
    20. Stricker T,
    21. Arteaga CL
    : Association of FGFR1 with ERα maintains ligand-independent ER transcription and mediates resistance to estrogen deprivation in ER(+) breast cancer. Clin Cancer Res 23(20): 6138-6150, 2017. DOI: 10.1158/1078-0432.CCR-17-1232
    OpenUrlAbstract/FREE Full Text
  83. ↵
    1. Mao P,
    2. Cohen O,
    3. Kowalski KJ,
    4. Kusiel JG,
    5. Buendia-Buendia JE,
    6. Cuoco MS,
    7. Exman P,
    8. Wander SA,
    9. Waks AG,
    10. Nayar U,
    11. Chung J,
    12. Freeman S,
    13. Rozenblatt-Rosen O,
    14. Miller VA,
    15. Piccioni F,
    16. Root DE,
    17. Regev A,
    18. Winer EP,
    19. Lin NU,
    20. Wagle N
    : Acquired FGFR and FGF alterations confer resistance to estrogen receptor (ER) targeted therapy in ER+ metastatic breast cancer. Clin Cancer Res 26(22): 5974-5989, 2020. DOI: 10.1158/1078-0432.Ccr-19-3958
    OpenUrlAbstract/FREE Full Text
  84. ↵
    1. Formisano L,
    2. Lu Y,
    3. Servetto A,
    4. Hanker AB,
    5. Jansen VM,
    6. Bauer JA,
    7. Sudhan DR,
    8. Guerrero-Zotano AL,
    9. Croessmann S,
    10. Guo Y,
    11. Ericsson PG,
    12. Lee KM,
    13. Nixon MJ,
    14. Schwarz LJ,
    15. Sanders ME,
    16. Dugger TC,
    17. Cruz MR,
    18. Behdad A,
    19. Cristofanilli M,
    20. Bardia A,
    21. O’Shaughnessy J,
    22. Nagy RJ,
    23. Lanman RB,
    24. Solovieff N,
    25. He W,
    26. Miller M,
    27. Su F,
    28. Shyr Y,
    29. Mayer IA,
    30. Balko JM,
    31. Arteaga CL
    : Aberrant FGFR signaling mediates resistance to CDK4/6 inhibitors in ER+ breast cancer. Nat Commun 10(1): 1373, 2019. DOI: 10.1038/s41467-019-09068-2
    OpenUrlCrossRefPubMed
  85. ↵
    1. O’Leary B,
    2. Cutts R,
    3. Huang X,
    4. Hrebien S,
    5. Liu Y,
    6. Garcia-Murillas I,
    7. Andre F,
    8. Loi S,
    9. Loibl S,
    10. Cristofanilli M,
    11. Bartlett CH,
    12. Turner NC
    : Genomic markers of early progression on fulvestrant with or without palbociclib for ER+ advanced breast cancer in the PALOMA-3 trial. J Clin Oncol 37(15_suppl): 1010-1010, 2019. DOI: 10.1200/JCO.2019.37.15_suppl.1010
    OpenUrlCrossRef
  86. ↵
    1. Dhillon AS,
    2. Hagan S,
    3. Rath O,
    4. Kolch W
    : MAP kinase signalling pathways in cancer. Oncogene 26(22): 3279-3290, 2007. DOI: 10.1038/sj.onc.1210421
    OpenUrlCrossRefPubMed
  87. ↵
    1. de Leeuw R,
    2. McNair C,
    3. Schiewer MJ,
    4. Neupane NP,
    5. Brand LJ,
    6. Augello MA,
    7. Li Z,
    8. Cheng LC,
    9. Yoshida A,
    10. Courtney SM,
    11. Hazard ES,
    12. Hardiman G,
    13. Hussain MH,
    14. Diehl JA,
    15. Drake JM,
    16. Kelly WK,
    17. Knudsen KE
    : MAPK reliance via acquired CDK4/6 inhibitor resistance in cancer. Clin Cancer Res 24(17): 4201-4214, 2018. DOI: 10.1158/1078-0432.CCR-18-0410
    OpenUrlAbstract/FREE Full Text
  88. ↵
    1. Pandey P,
    2. Khan F,
    3. Upadhyay TK,
    4. Sharangi AB
    : Deciphering the immunomodulatory role of cyclin-dependent kinase 4/6 inhibitors in the tumor microenvironment. Int J Mol Sci 24(3): 2236, 2023. DOI: 10.3390/ijms24032236
    OpenUrlCrossRefPubMed
  89. ↵
    1. Goel S,
    2. DeCristo MJ,
    3. Watt AC,
    4. BrinJones H,
    5. Sceneay J,
    6. Li BB,
    7. Khan N,
    8. Ubellacker JM,
    9. Xie S,
    10. Metzger-Filho O,
    11. Hoog J,
    12. Ellis MJ,
    13. Ma CX,
    14. Ramm S,
    15. Krop IE,
    16. Winer EP,
    17. Roberts TM,
    18. Kim HJ,
    19. McAllister SS,
    20. Zhao JJ
    : CDK4/6 inhibition triggers anti-tumour immunity. Nature 548(7668): 471-475, 2017. DOI: 10.1038/nature23465
    OpenUrlCrossRefPubMed
  90. ↵
    1. Vilgelm AE,
    2. Johnson CA,
    3. Prasad N,
    4. Yang J,
    5. Chen SC,
    6. Ayers GD,
    7. Pawlikowski JS,
    8. Raman D,
    9. Sosman JA,
    10. Kelley M,
    11. Ecsedy JA,
    12. Shyr Y,
    13. Levy SE,
    14. Richmond A
    : Connecting the dots: Therapy-induced senescence and a tumor-suppressive immune microenvironment. J Natl Cancer Inst 108(6): djv406, 2015. DOI: 10.1093/jnci/djv406
    OpenUrlCrossRefPubMed
  91. ↵
    1. Wang X,
    2. Lang M,
    3. Zhao T,
    4. Feng X,
    5. Zheng C,
    6. Huang C,
    7. Hao J,
    8. Dong J,
    9. Luo L,
    10. Li X,
    11. Lan C,
    12. Yu W,
    13. Yu M,
    14. Yang S,
    15. Ren H
    : Cancer-FOXP3 directly activated CCL5 to recruit FOXP3(+)Treg cells in pancreatic ductal adenocarcinoma. Oncogene 36(21): 3048-3058, 2017. DOI: 10.1038/onc.2016.458
    OpenUrlCrossRef
  92. ↵
    1. Vijayaraghavan S,
    2. Doostan I,
    3. Carey JP,
    4. Keyomarsi K
    : Abstract 2060: Characterizing acquired resistance to palbociclib in breast cancer. Cancer Res 77(13_Suppl): 2060-2060, 2017. DOI: 10.1158/1538-7445.AM2017-2060
    OpenUrlCrossRef
  93. ↵
    1. Zhang Z,
    2. Li J,
    3. Ou Y,
    4. Yang G,
    5. Deng K,
    6. Wang Q,
    7. Wang Z,
    8. Wang W,
    9. Zhang Q,
    10. Wang H,
    11. Sun W,
    12. Sun P,
    13. Yang S
    : CDK4/6 inhibition blocks cancer metastasis through a USP51-ZEB1-dependent deubiquitination mechanism. Signal Transduct Target Ther 5(1): 25, 2020. DOI: 10.1038/s41392-020-0118-x
    OpenUrlCrossRefPubMed
  94. ↵
    1. Xi J,
    2. Ma CX
    : Sequencing endocrine therapy for metastatic breast cancer: What do we do after disease progression on a CDK4/6 inhibitor? Curr Oncol Rep 22(6): 57, 2020. DOI: 10.1007/s11912-020-00917-8
    OpenUrlCrossRefPubMed
  95. ↵
    1. Portman N,
    2. Alexandrou S,
    3. Carson E,
    4. Wang S,
    5. Lim E,
    6. Caldon CE
    : Overcoming CDK4/6 inhibitor resistance in ER-positive breast cancer. Endocr Relat Cancer 26(1): R15-R30, 2019. DOI: 10.1530/ERC-18-0317
    OpenUrlCrossRefPubMed
  96. ↵
    1. Lane AA,
    2. Chabner BA
    : Histone deacetylase inhibitors in cancer therapy. J Clin Oncol 27(32): 5459-5468, 2009. DOI: 10.1200/jco.2009.22.1291
    OpenUrlAbstract/FREE Full Text
  97. ↵
    1. Carcagno AL,
    2. Marazita MC,
    3. Ogara MF,
    4. Ceruti JM,
    5. Sonzogni SV,
    6. Scassa ME,
    7. Giono LE,
    8. Cánepa ET
    : E2F1-mediated upregulation of p19INK4d determines its periodic expression during cell cycle and regulates cellular proliferation. PLoS One 6(7): e21938, 2011. DOI: 10.1371/journal.pone.0021938
    OpenUrlCrossRefPubMed
  98. ↵
    1. Schachter MM,
    2. Merrick KA,
    3. Larochelle S,
    4. Hirschi A,
    5. Zhang C,
    6. Shokat KM,
    7. Rubin SM,
    8. Fisher RP
    : A Cdk7-Cdk4 T-loop phosphorylation cascade promotes G1 progression. Mol Cell 50(2): 250-260, 2013. DOI: 10.1016/j.molcel.2013.04.003
    OpenUrlCrossRefPubMed
  99. ↵
    1. Ji Q,
    2. Qi R,
    3. Zhang L,
    4. Zhang Y,
    5. Xu J,
    6. Yuan W,
    7. Cheng T,
    8. Gao Y
    : Effect of PD0332991 on biological activity of hematopoietic stem cells in mice. Zhonghua Xue Ye Xue Za Zhi 35(2): 157-161, 2014. DOI: 10.3760/cma.j.issn.0253-2727.2014.02.021
    OpenUrlCrossRefPubMed
  100. ↵
    1. Del Re M,
    2. Bertolini I,
    3. Crucitta S,
    4. Fontanelli L,
    5. Rofi E,
    6. De Angelis C,
    7. Diodati L,
    8. Cavallero D,
    9. Gianfilippo G,
    10. Salvadori B,
    11. Fogli S,
    12. Falcone A,
    13. Scatena C,
    14. Naccarato AG,
    15. Roncella M,
    16. Ghilli M,
    17. Morganti R,
    18. Fontana A,
    19. Danesi R
    : Overexpression of TK1 and CDK9 in plasma-derived exosomes is associated with clinical resistance to CDK4/6 inhibitors in metastatic breast cancer patients. Breast Cancer Res Treat 178(1): 57-62, 2019. DOI: 10.1007/s10549-019-05365-y
    OpenUrlCrossRefPubMed
  101. ↵
    1. Ji W,
    2. Shi Y,
    3. Wang X,
    4. He W,
    5. Tang L,
    6. Tian S,
    7. Jiang H,
    8. Shu Y,
    9. Guan X
    : Combined Androgen receptor blockade overcomes the resistance of breast cancer cells to palbociclib. Int J Biol Sci 15(3): 522-532, 2019. DOI: 10.7150/ijbs.30572
    OpenUrlCrossRefPubMed
  102. ↵
    1. Seki H,
    2. Sakurai T,
    3. Maeda Y,
    4. Oki N,
    5. Aoyama M,
    6. Yamaguchi R,
    7. Tokuda T,
    8. Kaburagi T,
    9. Okumura T,
    10. Karahashi T,
    11. Nakajima K,
    12. Higeta K,
    13. Shimizu K
    : Efficacy and safety of palbociclib and fulvestrant in Japanese patients with ER+/HER2− advanced/metastatic breast cancer. In Vivo 33(6): 2037-2044, 2019. DOI: 10.21873/invivo.11701
    OpenUrlAbstract/FREE Full Text
  103. ↵
    1. Kalinsky K,
    2. Accordino MK,
    3. Chiuzan C,
    4. Mundi PS,
    5. Trivedi MS,
    6. Novik Y,
    7. Tiersten A,
    8. Raptis G,
    9. Baer LN,
    10. Young Oh S,
    11. Zelnak AB,
    12. Wisinski KB,
    13. Andreopoulou E,
    14. Gradishar WJ,
    15. Stringer-Reasor E,
    16. Reid SA,
    17. O’Dea A,
    18. O’Regan R,
    19. Crew KD,
    20. Hershman DL
    : A randomized, phase II trial of fulvestrant or exemestane with or without ribociclib after progression on anti-estrogen therapy plus cyclin-dependent kinase 4/6 inhibition (CDK 4/6i) in patients (pts) with unresectable or hormone receptor-positive (HR+), HER2-negative metastatic breast cancer (MBC): MAINTAIN trial. J Clin Oncol 40(17_suppl): LBA1004-LBA1004, 2022. DOI: 10.1200/JCO.2022.40.17_suppl.LBA1004
    OpenUrlCrossRef
  104. ↵
    1. Pacilio C,
    2. Rosati G,
    3. Crispo A,
    4. Bimonte S,
    5. DI Rella F,
    6. Nuzzo F,
    7. DE Laurentiis M
    : An overview of the roles of CDK4/6 inhibitors in metastatic breast cancer elderly patients. In Vivo 37(4): 1445-1449, 2023. DOI: 10.21873/invivo.13228
    OpenUrlAbstract/FREE Full Text
  105. ↵
    1. Bardia A,
    2. Aftimos P,
    3. Bihani T,
    4. Anderson-Villaluz AT,
    5. Jung J,
    6. Conlan MG,
    7. Kaklamani VG
    : EMERALD: Phase III trial of elacestrant (RAD1901) vs endocrine therapy for previously treated ER+ advanced breast cancer. Fut Oncol 15(28): 3209-3218, 2019. DOI: 10.2217/fon-2019-0370
    OpenUrlCrossRefPubMed
  106. ↵
    1. Bidard FC,
    2. Kaklamani VG,
    3. Neven P,
    4. Streich G,
    5. Montero AJ,
    6. Forget F,
    7. Mouret-Reynier MA,
    8. Sohn JH,
    9. Taylor D,
    10. Harnden KK,
    11. Khong H,
    12. Kocsis J,
    13. Dalenc F,
    14. Dillon PM,
    15. Babu S,
    16. Waters S,
    17. Deleu I,
    18. García Sáenz JA,
    19. Bria E,
    20. Cazzaniga M,
    21. Lu J,
    22. Aftimos P,
    23. Cortés J,
    24. Liu S,
    25. Tonini G,
    26. Laurent D,
    27. Habboubi N,
    28. Conlan MG,
    29. Bardia A
    : Elacestrant (oral selective estrogen receptor degrader) versus standard endocrine therapy for estrogen receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer: results from the randomized phase III EMERALD trial. J Clin Oncol 40(28): 3246-3256, 2022. DOI: 10.1200/JCO.22.00338
    OpenUrlCrossRefPubMed
  107. ↵
    1. Bardia A,
    2. Hurvitz SA,
    3. DeMichele A,
    4. Clark AS,
    5. Zelnak A,
    6. Yardley DA,
    7. Karuturi M,
    8. Sanft T,
    9. Blau S,
    10. Hart L,
    11. Ma C,
    12. Rugo HS,
    13. Purkayastha D,
    14. Moulder S
    : Phase I/II trial of exemestane, ribociclib, and everolimus in women with HR(+)/HER2(−) advanced breast cancer after progression on CDK4/6 inhibitors (TRINITI-1). Clin Cancer Res 27(15): 4177-4185, 2021. DOI: 10.1158/1078-0432.CCR-20-2114
    OpenUrlAbstract/FREE Full Text
  108. ↵
    1. Stokoe D,
    2. Stephens LR,
    3. Copeland T,
    4. Gaffney PRJ,
    5. Reese CB,
    6. Painter GF,
    7. Holmes AB,
    8. McCormick F,
    9. Hawkins PT
    : Dual role of phosphatidylinositol-3,4,5-trisphosphate in the activation of protein kinase B. Science 277(5325): 567-570, 1997. DOI: 10.1126/science.277.5325.567
    OpenUrlAbstract/FREE Full Text
  109. ↵
    1. Vicier C,
    2. Sfumato P,
    3. Isambert N,
    4. Dalenc F,
    5. Robert M,
    6. Levy C,
    7. Rezai K,
    8. Provansal M,
    9. Adélaïde J,
    10. Garnier S,
    11. Guille A,
    12. Carbuccia N,
    13. Popovici C,
    14. Charafe-Jauffret E,
    15. Chaffanet M,
    16. Birnbaum D,
    17. Pakradouni J,
    18. Bertucci F,
    19. Boher JM,
    20. Sabatier R,
    21. Gonçalves A
    : TAKTIC: A prospective, multicentre, uncontrolled, phase IB/II study of LY2780301, a p70S6K/AKT inhibitor, in combination with weekly paclitaxel in HER2-negative advanced breast cancer patients. Eur J Cancer 159: 205-214, 2021. DOI: 10.1016/j.ejca.2021.09.040
    OpenUrlCrossRefPubMed
  110. ↵
    1. Contreras-Leal E,
    2. Hernández-Oliveras A,
    3. Flores-Peredo L,
    4. Zarain-Herzberg Á,
    5. Santiago-García J
    : Histone deacetylase inhibitors promote the expression ofATP2A3gene in breast cancer cell lines. Mol Carcinog 55(10): 1477-1485, 2016. DOI: 10.1002/mc.22402
    OpenUrlCrossRefPubMed
  111. ↵
    1. Chatterjee N,
    2. Wang WL,
    3. Conklin T,
    4. Chittur S,
    5. Tenniswood M
    : Histone deacetylase inhibitors modulate miRNA and mRNA expression, block metaphase, and induce apoptosis in inflammatory breast cancer cells. Cancer Biol Ther 14(7): 658-671, 2013. DOI: 10.4161/cbt.25088
    OpenUrlCrossRefPubMed
  112. ↵
    1. Li L,
    2. Sun Y,
    3. Liu J,
    4. Wu X,
    5. Chen L,
    6. Ma L,
    7. Wu P
    : Histone deacetylase inhibitor sodium butyrate suppresses DNA double strand break repair induced by etoposide more effectively in MCF-7 cells than in HEK293 cells. BMC Biochem 16: 2, 2015. DOI: 10.1186/s12858-014-0030-5
    OpenUrlCrossRefPubMed
  113. ↵
    1. Jiang Z,
    2. Li W,
    3. Hu X,
    4. Zhang Q,
    5. Sun T,
    6. Cui S,
    7. Wang S,
    8. Ouyang Q,
    9. Yin Y,
    10. Geng C,
    11. Tong Z,
    12. Cheng Y,
    13. Pan Y,
    14. Sun Y,
    15. Wang H,
    16. Ouyang T,
    17. Gu K,
    18. Feng J,
    19. Wang X,
    20. Wang S,
    21. Liu T,
    22. Gao J,
    23. Cristofanilli M,
    24. Ning Z,
    25. Lu X
    : Tucidinostat plus exemestane for postmenopausal patients with advanced, hormone receptor-positive breast cancer (ACE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 20(6): 806-815, 2019. DOI: 10.1016/s1470-2045(19)30164-0
    OpenUrlCrossRefPubMed
  114. ↵
    1. Kawiak A,
    2. Kostecka A
    : Regulation of Bcl-2 family proteins in estrogen receptor-positive breast cancer and their implications in endocrine therapy. Cancers (Basel) 14(2): 279, 2022. DOI: 10.3390/cancers14020279
    OpenUrlCrossRefPubMed
  115. ↵
    1. Lindeman GJ,
    2. Fernando TM,
    3. Bowen R,
    4. Jerzak KJ,
    5. Song X,
    6. Decker T,
    7. Boyle F,
    8. McCune S,
    9. Armstrong A,
    10. Shannon C,
    11. Bertelli G,
    12. Chang CW,
    13. Desai R,
    14. Gupta K,
    15. Wilson TR,
    16. Flechais A,
    17. Bardia A
    : VERONICA: Randomized phase II study of fulvestrant and venetoclax in ER-positive metastatic breast cancer post-CDK4/6 inhibitors - efficacy, safety, and biomarker results. Clin Cancer Res 28(15): 3256-3267, 2022. DOI: 10.1158/1078-0432.CCR-21-3811
    OpenUrlCrossRefPubMed
  116. ↵
    1. Schaer DA,
    2. Beckmann RP,
    3. Dempsey JA,
    4. Huber L,
    5. Forest A,
    6. Amaladas N,
    7. Li Y,
    8. Wang YC,
    9. Rasmussen ER,
    10. Chin D,
    11. Capen A,
    12. Carpenito C,
    13. Staschke KA,
    14. Chung LA,
    15. Litchfield LM,
    16. Merzoug FF,
    17. Gong X,
    18. Iversen PW,
    19. Buchanan S,
    20. de Dios A,
    21. Novosiadly RD,
    22. Kalos M
    : The CDK4/6 inhibitor abemaciclib induces a T cell inflamed tumor microenvironment and enhances the efficacy of PD-L1 checkpoint blockade. Cell Rep 22(11): 2978-2994, 2018. DOI: 10.1016/j.celrep.2018.02.053
    OpenUrlCrossRefPubMed
  117. ↵
    1. Cohen I,
    2. Tagliaferri M,
    3. Tripathy D
    : Traditional Chinese medicine in the treatment of breast cancer. Semin Oncol 29(6): 563-574, 2002. DOI: 10.1053/sonc.2002.50005
    OpenUrlCrossRefPubMed
  118. ↵
    1. Hu M,
    2. Zhao M,
    3. An C,
    4. Yang M,
    5. Li Q,
    6. Zhang Y,
    7. Suetsugu A,
    8. Tome Y,
    9. Yano S,
    10. Fu Y,
    11. Hoffman RM,
    12. Hu K
    : Real-time imaging of apoptosis induction of human breast cancer cells by the traditional chinese medicinal herb tubeimu. Anticancer Res 32(7): 2509-2514, 2012.
    OpenUrlAbstract/FREE Full Text
  119. ↵
    1. Xie XH,
    2. Zhao H,
    3. Hu YY,
    4. Gu XD
    : Germacrone reverses Adriamycin resistance through cell apoptosis in multidrug-resistant breast cancer cells. Exp Ther Med 8(5): 1611-1615, 2014. DOI: 10.3892/etm.2014.1932
    OpenUrlCrossRefPubMed
  120. ↵
    1. Ezzati M,
    2. Yousefi B,
    3. Velaei K,
    4. Safa A
    : A review on anti-cancer properties of Quercetin in breast cancer. Life Sci 248: 117463, 2020. DOI: 10.1016/j.lfs.2020.117463
    OpenUrlCrossRefPubMed
  121. ↵
    1. Li F,
    2. Wang Y,
    3. Wang X,
    4. Li J,
    5. Cui H,
    6. Niu M
    : Ganoderic acids suppress growth and angiogenesis by modulating the NF-κB signaling pathway in breast cancer cells. Int J Clin Pharmacol Ther 50(10): 712-721, 2012. DOI: 10.5414/cp201663
    OpenUrlCrossRefPubMed
  122. ↵
    1. Wu J,
    2. Liu Y,
    3. Fang C,
    4. Zhao L,
    5. Lin L,
    6. Lu L
    : Traditional Chinese medicine preparation combined therapy may improve chemotherapy efficacy: a systematic review and meta-analysis. Evid Based Complement Alternat Med 2019: 5015824, 2019. DOI: 10.1155/2019/5015824
    OpenUrlCrossRefPubMed
  123. ↵
    1. Guo HY,
    2. Chen ZA,
    3. Shen QK,
    4. Quan ZS
    : Application of triazoles in the structural modification of natural products. J Enzyme Inhib Med Chem 36(1): 1115-1144, 2021. DOI: 10.1080/14756366.2021.1890066
    OpenUrlCrossRefPubMed
  124. ↵
    1. Hou W,
    2. Luo Z,
    3. Zhang G,
    4. Cao D,
    5. Li D,
    6. Ruan H,
    7. Ruan BH,
    8. Su L,
    9. Xu H
    : Click chemistry-based synthesis and anticancer activity evaluation of novel C-14 1,2,3-triazole dehydroabietic acid hybrids. Eur J Med Chem 138: 1042-1052, 2017. DOI: 10.1016/j.ejmech.2017.07.049
    OpenUrlCrossRefPubMed
  125. ↵
    1. Zhu X,
    2. Wong ILK,
    3. Chan KF,
    4. Cui J,
    5. Law MC,
    6. Chong TC,
    7. Hu X,
    8. Chow LMC,
    9. Chan TH
    : Triazole bridged flavonoid dimers as potent, nontoxic, and highly selective breast cancer resistance protein (BCRP/ABCG2) inhibitors. J Med Chem 62(18): 8578-8608, 2019. DOI: 10.1021/acs.jmedchem.9b00963
    OpenUrlCrossRefPubMed
  126. ↵
    1. Chong TC,
    2. Wong ILK,
    3. Cui J,
    4. Law MC,
    5. Zhu X,
    6. Hu X,
    7. Kan JWY,
    8. Yan CSW,
    9. Chan TH,
    10. Chow LMC
    : Characterization of a potent, selective, and safe inhibitor, Ac15(Az8)(2), in reversing multidrug resistance mediated by breast cancer resistance protein (BCRP/ABCG2). Int J Mol Sci 23(21): 13261, 2022. DOI: 10.3390/ijms232113261
    OpenUrlCrossRefPubMed
    1. Finn RS,
    2. Crown JP,
    3. Lang I,
    4. Boer K,
    5. Bondarenko IM,
    6. Kulyk SO,
    7. Ettl J,
    8. Patel R,
    9. Pinter T,
    10. Schmidt M,
    11. Shparyk Y,
    12. Thummala AR,
    13. Voytko NL,
    14. Fowst C,
    15. Huang X,
    16. Kim ST,
    17. Randolph S,
    18. Slamon DJ
    : 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 16(1): 25-35, 2015. DOI: 10.1016/s1470-2045(14)71159-3
    OpenUrlCrossRefPubMed
    1. Rugo HS,
    2. Finn RS,
    3. Diéras V,
    4. Ettl J,
    5. Lipatov O,
    6. Joy AA,
    7. Harbeck N,
    8. Castrellon A,
    9. Iyer S,
    10. Lu DR,
    11. Mori A,
    12. Gauthier ER,
    13. Bartlett CH,
    14. Gelmon KA,
    15. Slamon DJ
    : Palbociclib plus letrozole as first-line therapy in estrogen receptor-positive/human epidermal growth factor receptor 2-negative advanced breast cancer with extended follow-up. Breast Cancer Res Treat 174(3): 719-729, 2019. DOI: 10.1007/s10549-018-05125-4
    OpenUrlCrossRefPubMed
    1. Verma S,
    2. Bartlett CH,
    3. Schnell P,
    4. DeMichele AM,
    5. Loi S,
    6. Ro J,
    7. Colleoni M,
    8. Iwata H,
    9. Harbeck N,
    10. Cristofanilli M,
    11. Zhang K,
    12. Thiele A,
    13. Turner NC,
    14. Rugo HS
    : Palbociclib in combination with fulvestrant in women with hormone receptor-positive/HER2-negative advanced metastatic breast cancer: Detailed safety analysis from a multicenter, randomized, placebo-controlled, phase III study (PALOMA-3). Oncologist 21(10): 1165-1175, 2016. DOI: 10.1634/theoncologist.2016-0097
    OpenUrlAbstract/FREE Full Text
    1. Xu B,
    2. Hu X,
    3. Li W,
    4. Sun T,
    5. Shen K,
    6. Wang S,
    7. Cheng Y,
    8. Zhang Q,
    9. Cui S,
    10. Tong Z,
    11. Geng C,
    12. Song E,
    13. Huang CS,
    14. Sriuranpong V,
    15. Ngan RK,
    16. Chia YH,
    17. Wang X,
    18. Zhao H
    : Palbociclib plus letrozole versus placebo plus letrozole in Asian postmenopausal women with oestrogen receptor-positive/human epidermal growth factor receptor 2-negative advanced breast cancer: Primary results from PALOMA-4. Eur J Cancer 175: 236-245, 2022. DOI: 10.1016/j.ejca.2022.08.012
    OpenUrlCrossRefPubMed
    1. Johnston S,
    2. Martin M,
    3. Di Leo A,
    4. Im SA,
    5. Awada A,
    6. Forrester T,
    7. Frenzel M,
    8. Hardebeck MC,
    9. Cox J,
    10. Barriga S,
    11. Toi M,
    12. Iwata H,
    13. Goetz MP
    : MONARCH 3 final PFS: a randomized study of abemaciclib as initial therapy for advanced breast cancer. NPJ Breast Cancer 5: 5, 2019. DOI: 10.1038/s41523-018-0097-z
    OpenUrlCrossRefPubMed
    1. Zhang QY,
    2. Sun T,
    3. Yin YM,
    4. Li HP,
    5. Yan M,
    6. Tong ZS,
    7. Oppermann CP,
    8. Liu YP,
    9. Costa R,
    10. Li M,
    11. Cheng Y,
    12. Ouyang QC,
    13. Chen X,
    14. Liao N,
    15. Wu XH,
    16. Wang XJ,
    17. Feng JF,
    18. Hegg R,
    19. Kanakasetty GB,
    20. Coccia-Portugal MA,
    21. Han RB,
    22. Lu Y,
    23. Chi HD,
    24. Jiang ZF,
    25. Hu XC
    : MONARCH plus: abemaciclib plus endocrine therapy in women with HR+/HER2− advanced breast cancer: the multinational randomized phase III study. Ther Adv Med Oncol 12: 1758835920963925, 2020. DOI: 10.1177/1758835920963925
    OpenUrlCrossRefPubMed
    1. Hortobagyi GN,
    2. Stemmer SM,
    3. Burris HA,
    4. Yap YS,
    5. Sonke GS,
    6. Paluch-Shimon S,
    7. Campone M,
    8. Petrakova K,
    9. Blackwell KL,
    10. Winer EP,
    11. Janni W,
    12. Verma S,
    13. Conte P,
    14. Arteaga CL,
    15. Cameron DA,
    16. Mondal S,
    17. Su F,
    18. Miller M,
    19. Elmeliegy M,
    20. Germa C,
    21. O’Shaughnessy J
    : Updated results from MONALEESA-2, a phase III trial of first-line ribociclib plus letrozole versus placebo plus letrozole in hormone receptor-positive, HER2-negative advanced breast cancer. Ann Oncol 29(7): 1541-1547, 2018. DOI: 10.1093/annonc/mdy155
    OpenUrlCrossRefPubMed
    1. Slamon DJ,
    2. Neven P,
    3. Chia S,
    4. Fasching PA,
    5. De Laurentiis M,
    6. Im SA,
    7. Petrakova K,
    8. Bianchi GV,
    9. Esteva FJ,
    10. Martín M,
    11. Nusch A,
    12. Sonke GS,
    13. De la Cruz-Merino L,
    14. Beck JT,
    15. Pivot X,
    16. Vidam G,
    17. Wang Y,
    18. Rodriguez Lorenc K,
    19. Miller M,
    20. Taran T,
    21. Jerusalem G
    : Phase III randomized study of ribociclib and fulvestrant in hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer: MONALEESA-3. J Clin Oncol 36(24): 2465-2472, 2018. DOI: 10.1200/jco.2018.78.9909
    OpenUrlCrossRefPubMed
    1. Rugo HS,
    2. Lerebours F,
    3. Ciruelos E,
    4. Drullinsky P,
    5. Ruiz-Borrego M,
    6. Neven P,
    7. Park YH,
    8. Prat A,
    9. Bachelot T,
    10. Juric D,
    11. Turner N,
    12. Sophos N,
    13. Zarate JP,
    14. Arce C,
    15. Shen Y,
    16. Turner S,
    17. Kanakamedala H,
    18. Hsu W,
    19. Chia S
    : Alpelisib plus fulvestrant in PIK3CA-mutated, hormone receptor-positive advanced breast cancer after a CDK4/6 inhibitor (BYLieve): one cohort of a phase 2, multicentre, open-label, non-comparative study. Lancet Oncol 22(4): 489-498, 2021. DOI: 10.1016/s1470-2045(21)00034-6
    OpenUrlCrossRefPubMed
    1. André F,
    2. Ciruelos EM,
    3. Juric D,
    4. Loibl S,
    5. Campone M,
    6. Mayer IA,
    7. Rubovszky G,
    8. Yamashita T,
    9. Kaufman B,
    10. Lu YS,
    11. Inoue K,
    12. Pápai Z,
    13. Takahashi M,
    14. Ghaznawi F,
    15. Mills D,
    16. Kaper M,
    17. Miller M,
    18. Conte PF,
    19. Iwata H,
    20. Rugo HS
    : Alpelisib plus fulvestrant for PIK3CA-mutated, hormone receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: final overall survival results from SOLAR-1. Ann Oncol 32(2): 208-217, 2021. DOI: 10.1016/j.annonc.2020.11.011
    OpenUrlCrossRefPubMed
    1. Piccart M,
    2. Hortobagyi GN,
    3. Campone M,
    4. Pritchard KI,
    5. Lebrun F,
    6. Ito Y,
    7. Noguchi S,
    8. Perez A,
    9. Rugo HS,
    10. Deleu I,
    11. Burris HA 3rd.,
    12. Provencher L,
    13. Neven P,
    14. Gnant M,
    15. Shtivelband M,
    16. Wu C,
    17. Fan J,
    18. Feng W,
    19. Taran T,
    20. Baselga J
    : Everolimus plus exemestane for hormone-receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: overall survival results from BOLERO-2†. Ann Oncol 25(12): 2357-2362, 2014. DOI: 10.1093/annonc/mdu456
    OpenUrlCrossRefPubMed
    1. Wander SA,
    2. Micalizzi DS,
    3. Dubash T,
    4. Juric D,
    5. Spring LM,
    6. Vidula N,
    7. Keenan J,
    8. Beeler M,
    9. Viscosi E,
    10. Che D,
    11. Fisher EL,
    12. Hepp RA,
    13. Moy B,
    14. Isakoff SJ,
    15. Ellisen LW,
    16. Supko JG,
    17. Maheswaran S,
    18. Haber DA,
    19. Bardia A
    : Abstract P1-18-22: AKT inhibition in combination with endocrine therapy and a CDK4/6 inhibitor (CDK4/6i) in patients with hormone receptor positive (HR+)/HER2 negative metastatic breast cancer (MBC) and prior CDK4/6i exposure: A translational investigation. Cancer Res 82(4_Suppl): P1-18-22-P1-18-22, 2022. DOI: 10.1158/1538-7445.SABCS21-P1-18-22
    OpenUrlCrossRef
PreviousNext
Back to top

In this issue

Anticancer Research: 43 (12)
Anticancer Research
Vol. 43, Issue 12
December 2023
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Mechanism of CDK4/6 Inhibitor Resistance in Hormone Receptor-positive Breast Cancer and Alternative Treatment Strategies
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
3 + 5 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Mechanism of CDK4/6 Inhibitor Resistance in Hormone Receptor-positive Breast Cancer and Alternative Treatment Strategies
CHUN-MING CHANG, HO YIN PEKKLE LAM
Anticancer Research Dec 2023, 43 (12) 5283-5298; DOI: 10.21873/anticanres.16732

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Mechanism of CDK4/6 Inhibitor Resistance in Hormone Receptor-positive Breast Cancer and Alternative Treatment Strategies
CHUN-MING CHANG, HO YIN PEKKLE LAM
Anticancer Research Dec 2023, 43 (12) 5283-5298; DOI: 10.21873/anticanres.16732
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • CDK4/6 Inhibitors
    • CDK4/6 Inhibitor Resistance
    • Intrinsic Resistance Mechanism
    • Acquired Resistance Mechanism
    • Other Possible Mechanisms
    • Potential Strategies to Overcome CDK4/6 Inhibitor Resistance in the Future
    • Conclusion
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • Intratumoral Microbiota: Unraveling their Oncogenic Impact on Cancer Progression With Focus on Breast Cancer Therapeutic Outcomes
  • Generative Modelling of Oncogene-carrying Extrachromosomal Circular DNA Biogenesis and Dynamics in Cells
  • Google Scholar

More in this TOC Section

  • Management of Bladder Cancer During Pregnancy: A Narrative Review
  • Mendelian Randomization Studies on Actinic Keratosis
  • Clinical Applications of Artificial Intelligence in Uveal Melanoma
Show more Review

Similar Articles

Keywords

  • CDK4/6 inhibitors-resistance
  • Breast cancer
  • palbociclib
  • ribociclib
  • abemaciclib
  • review
Anticancer Research

© 2025 Anticancer Research

Powered by HighWire