Effects of combined use of ribociclib with PARP1 inhibitor on cell kinetics in breast cancer

  • Authors:
    • Ercan Pulat
    • Mehmet R. Topçul
  • View Affiliations

  • Published online on: April 3, 2024     https://doi.org/10.3892/ol.2024.14376
  • Article Number: 243
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In the present study, antiproliferative and anticancer effects of Valamor (VLM), which contains the active component ribociclib, and DPQ, a poly(ADP‑ribose) polymerase 1 inhibitor, alone and in combination were evaluated in the MCF‑7 and MDA‑MB‑231 breast cancer cell lines in vitro. VLM was applied at concentrations of 40, 80 and 160 µg/ml, and DPQ was used at concentrations of 3, 6 and 9 µg/ml. The proliferation rate, cell index obtained from the real‑time cell analysis system, mitosis activity, bromodeoxyuridine cell proliferation and caspase activity parameters were determined. In conclusion, the results obtained from cell kinetics parameters demonstrated the anticancer and antiproliferative effects of the combination of VLM and DPQ on breast cancer cells.

Introduction

In the past, breast cancer (BC) was the most prevalent illness affecting women globally (14). Since the time of the ancient Egyptians, attempts have been made to eradicate BC. More advanced surgical procedures are now being used to decrease the psychological toll of treatment, while medicinal therapies such as mastectomy and chemotherapy have significantly enhanced patient survival (57). However, preventative effectiveness and treatment options can never be fully effective without a thorough grasp of the pathophysiology and underlying mechanisms.

BC is a form of cancer that manifests itself differently in various individuals (8). Women all across the world are affected by BC, a prevalent type of cancer (9). BC can be classified into three groups based on molecular and histological evidence: BC expressing human epidermal growth factor receptor 2 (HER2+), triple-negative BC [TNBC; estrogen receptor (ER)-, progesterone receptor (PR)- and HER2-] and BC expressing hormone receptors, ER+ or PR+ (10,11).

The methods of treatment should be determined by the molecular features of BC. TNBC has also been classified into six groups: Luminal androgen receptor, immunomodulatory, mesenchymal, mesenchymal stem cell-like, basal-like 1 (BL-1) and BL-2 (10). These categories are not always useful because a single BC may have a variety of distinct cell types (4,1215).

The majority of BC-related fatalities result from metastases. At 3 years after the main tumor was first discovered, 10–15% of patients with BC had distant metastases (16). At 10 years after the first diagnosis, it is common to observe the appearance of micrometastases in distant places. As a result, individuals with BC have a lifetime risk of developing metastases (1618).

Third-generation cyclin-dependent kinase (CDK)4/6 inhibitor ribociclib (LEE011) is highly selective and inhibits CDK4/6 by competitively interacting with its ATP binding sites. By inhibiting the CDK4/6-cyclin D-retinoblastoma (Rb)-E2F axis, this strategy may stop unchecked cell division and tumor progression. CDK4 and 6 are crucial for the development and division of cancer cells, and ribociclib may limit the development of cancerous cells and stop the spread of the disease by inhibiting these enzymes (19).

Poly(ADP-ribose) polymerase (PARP) has crucial roles in DNA repair, apoptosis, cell regulation, cell division, differentiation, transcriptional regulation and chromosome maintenance (20,21). PARP1 repairs single-strand breaks in DNA through a base truncation repair mechanism. PARP-1 inhibition is a potent cancer death pathway (21). DNA damage occurs with cancer treatments such as temozolamide, platinum compounds, topoisomerase inhibitors and radiation therapy. Treatment resistance develops due to PARP, which is involved in DNA repair. Therefore, inhibiting the PARP enzyme may increase the treatment efficacy. Tumor suppressor genes, such as mutated BRCA1 and BRCA2, are highly sensitive to PARP1 inhibition, leading to cell cycle arrest and apoptosis (22). It has been proposed that PARP limits the spread of improperly repaired DNA, serving as a connection between severe DNA damage and cell death. Berger was the first to postulate this idea, also referred to as the ‘PARP suicide hypothesis’ (23).

In this study, the antiproliferative and anticancer effects of Ribociclib and PARP1 inhibitor alone and together on the Luminal A type breast cancer MCF-7 cell line and triple negative cancer type MDA-MB-231 cell line were investigated.

Materials and methods

Cell culture

The TNBC cell line MDA-MB-231 and the luminal A BC cell line MCF-7 (European Cell Culture Collection) were both employed. The two cell lines were kept in DMEM (Gibco: Thermo Fisher Scientific, Inc.) tissue culture medium containing 100 µg/ml Streptomycin sulfate (I.E. Ulugay), 100 IU/ml penicillin (Pronapen; Pfizer), Amphotericin B (Sigma-Aldrich; Merck KGaA), 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) with the pH adjusted to 7.2 with 4.4% NaHCO3.

Ribociclib and PARP1 concentration

In the experiments, for both cell lines, 40, 80 and 160 µg/ml concentrations of VLM (FARMANOVA), which contains the active ingredient ribociclib, and 3, 6 and 9 µg/ml concentrations of DPQ (Sigma-Aldrich; Merck KGaA), a PARP1 inhibitor, were used. These concentrations are based on preliminary experiments performed by our group.

Cell viability assay

The MTT assay was used to investigate the cytotoxicity of VLM and DPQ on the cells as a consequence of the application of the planned doses. For this application, MCF-7 cells were seeded into 96-well plates at a density of 2×104 cells per well and incubated overnight. The cells were then treated with VLM and DPQ concentrations for 24 h. At the end of the experimental period, the medium in each well was removed and 40 µl fresh MTT solution (Sigma-Aldrich; Merck KGaA) was added into each well and cells were incubated at 37°C for 4 h. Subsequently, 160 µl DMSO was added. By using the 690 nm wavelength as a reference, a spectrophotometer was used to measure the absorbance values of the experimental groups at 570 nm.

Cell index

Cell index values determine the cytotoxic effect of agents applied to the cell by monitoring cell reproduction, cell size or morphology in real time. The cytotoxic impact of VLM and DPQ on the designated cell lines was assessed by xCELLigence DP (Acea Biosciences, Inc.). Before taking any measurements, each well of a 16-well E-plate was filled with 100 µl of the appropriate medium. Subsequently, 100 µl cell suspension was added to each well of the E-plate. In each well, 10.000 cells for the MCF-7 cell line and 5.000 cells for the MDA-MB-231 cell line were seeded. The E-plates were kept at room temperature in a sterile environment for 20 min and then placed in the stations in the device and the experiment was continued at 37°C and 5% CO2 under saturated humidity ambient conditions. The device was set to measure every 15 min. After an overnight incubation, drugs were added and measurements were continued (24).

Bromodeoxyuridine (BrdU) incorporation assay

VLM and DPQ were used in the experiments in line with the kit's protocol (BrdU Cell Proliferation Assay Kit; cat. no. 2750; EMD Millipore) at the optimum combination concentrations.

Mitotic index

Experimental results were obtained following the kit protocol with VLM and DPQ at the specified doses (Mitotic assay kit; cat. no. 18021; Active Motif).

Caspase activity

In line with the kit instructions (CaspaTag Caspase 3, 7 in situ assay kit; cat. no. APT403; EMD Millipore), experimental findings were obtained.

Statistical analysis

All parameters of cell kinetics were evaluated comparing multiple independent groups. Comparisons between groups were performed using one-way ANOVA and Dunnett's tests. Statistical evaluations of the cell index were made by the xCelligence device. Dunnett's post-hoc test was used for all other parameters. Statistical analyses were reviewed twice in line with referee opinions. Experiments were performed in triplicate. The statistical analyses were performed using SPSS statistics software (v22.0; IBM). P<0.05 was considered to indicate a statistically significant difference.

Results

Cell viability

When the MCF-7 cell absorbance levels following treatment for 40, 80 and 160 µg/ml VLM for 24 h were analyzed, it was observed that the 40 µg/ml VLM decreased the viability of MCF-7 cells to 85%, the 80 µg/ml VLM concentration decreased cell viability to 49.19% and the 160 µg/ml VLM concentration decreased cell viability to 13.79% compared to the control group, which was set as 100% (Fig. 1A).

Analysis of the absorbance values following DPQ treatment of MCF-7 cells at concentrations of 3, 6 and 9 µg/ml for 24 h showed that the 3 µg/ml DPQ concentration decreased the viability of MCF-7 cells to 65%, the 6 µg/ml DPQ concentration decreased cell viability to 57% and the 9 µg/ml DPQ concentration decreased cell viability to 51% compared to the control group, which was set as 100% (Fig. 1B).

Analysis of the absorbance rates after 40, 80 and 160 µg/ml VLM treatment of MDA-MB-231 cells for 24 h indicated that the 40 µg/ml VLM concentration decreased the viability of MDA-MB-231 cells to 46.86% compared to the control group, which was considered 100%. The 80 µg/ml VLM concentration reduced the cell viability to 45.71%, while the 160 µg/ml VLM concentration reduced the cell viability to 25.42% (Fig. 1C).

When the absorbance values were analyzed after DPQ was applied to MDA-MB-231 cells at concentrations of 3, 6 and 9 µg/ml for 24 h, it was observed that 3 µg/ml MDA-MB-231 cells had a 63.52% vitality after exposure to DPQ, the 6 µg/ml DPQ concentration decreased cell viability to 25.93% and the 9 µg/ml DPQ concentration decreased cell viability to 12.48% compared to the control group, which was set as 100% (Fig. 1D).

Cell index

Cell index values obtained from the xCelligence real-time cell analysis system (Acea Biosciences Inc.) monitor cell reproduction, cell size or morphology in real time. Acquired cell index values from the real-time cell analysis (RTCA) after treatment with VLM in MCF-7 and MDA-MB-231 cells at 40, 80 and 160 µg/ml showed that the drug had antiproliferative effects in both cell lines. When the curves obtained from the cell index graphs of MCF-7 cells are examined, different effects are observed at different concentrations. While no effect was observed at a concentration of 40 µg/ml, an antimitotic effect was observed at a concentration of 80 µg/ml and DNA damage was observed at a concentration of 160 µg/ml (Fig. 2). When the curves obtained from the cell index graphs of MDA-MB-231 cells are examined, all concentrations applied appeared to have an antiproliferative effect and cause DNA damage to the cells (Fig. 3).

The cell index values obtained from the RTAC system after DPQ treatment of cells at concentrations of 3, 6 and 9 µg/ml showed that the PARP inhibitor had antiproliferative effects on both cell lines. When the curves obtained from the cell index graphs of MCF-7 cells were examined, DNA damage was observed at all concentrations (Fig. 4). When the curves obtained from the cell index graphs of MDA-MB-231 cells were examined, it was observed that all concentrations applied had antiproliferative effects and caused DNA damage to the cells (Fig. 5).

Cell index values obtained from the RTAC system showed antiproliferative effects on both MCF-7 and MDA-MB-231 cells as a result of the combined application of VLM and DPQ at concentrations of 2.5 µg/ml VLM + 1.5 µg/ml DPQ, 20 µg/ml VLM + 4.5 µg/ml DPQ and 10 µg/ml VLM + 4.5 µg/ml DPQ. The combined concentrations showed DNA damage in MCF-7 cells (Fig. 6) and DNA damage in MDA-MB-231 cells (Fig. 7).

BrdU incorporation

Combining VLM with DPQ to treat MCF-7 and MDA-MB-231 cells (2.5 µg/ml VLM + 1.5 µg/ml DPQ) for 0–72 h resulted in labeling of cells with BrdU at the synthesis stage and the absorbance values obtained are presented in Tables I and II, respectively. There was a significant difference between the proliferation of control and experimental groups (P<0.05).

Table I.

Absorbance values at 450–655 nm (emission) of MCF-7 cells treated with 2.5 µg/ml VLM + 1.5 µg/ml DPQ in the bromodeoxyuridine assay (×10−3).

Table I.

Absorbance values at 450–655 nm (emission) of MCF-7 cells treated with 2.5 µg/ml VLM + 1.5 µg/ml DPQ in the bromodeoxyuridine assay (×10−3).

Time, hControlCombination treatment
24502±3398±2a
48507±4257±3a
72496±4203±2a

a P<0.05 vs. control.

Table II.

Absorbance values at 450–655 nm (emission) of MDA-MB-231 cells treated with 2.5 µg/ml Valamor + 1.5 µg/ml DPQ in the bromodeoxyuridine assay (×10−3).

Table II.

Absorbance values at 450–655 nm (emission) of MDA-MB-231 cells treated with 2.5 µg/ml Valamor + 1.5 µg/ml DPQ in the bromodeoxyuridine assay (×10−3).

Time, hControlCombination treatment
24625±3609±2a
48639±4355±3a
72635±4300±2a

a P<0.05 vs. control.

Mitotic activity

The absorbance values reflecting mitotic activity of MCF-7 and MDA-MB-231 cells treated with a combination of VLM and DPQ (2.5 µg/ml VLM + 1.5 µg/ml DPQ) for 0–72 h are presented in Tables III and IV, respectively. There was a significant decrease in the experimental group according to the control (P<0.05).

Table III.

Absorbance values at 450–655 nm (emission) of MCF-7 cells treated with 2.5 µg/ml Valamor + 1.5 µg/ml DPQ reflecting mitotic activity (×10−3).

Table III.

Absorbance values at 450–655 nm (emission) of MCF-7 cells treated with 2.5 µg/ml Valamor + 1.5 µg/ml DPQ reflecting mitotic activity (×10−3).

Time, hControlCombination treatment
24169±397±2a
48165±0483±3a
72163±0432±2a

a P<0.05 vs. control.

Table IV.

Absorbance values at 450–655 nm (emission) of MDA-MB-231 cells treated with 2.5 µg/ml Valamor + 1.5 µg/ml DPQ reflecting mitotic activity (×10−3).

Table IV.

Absorbance values at 450–655 nm (emission) of MDA-MB-231 cells treated with 2.5 µg/ml Valamor + 1.5 µg/ml DPQ reflecting mitotic activity (×10−3).

Time, hControlCombination treatment
2484±352±2a
4885±428±3a
7282±416±2a

a P<0.05 vs. control.

Caspase activity

In Tables V and VI, the respective fluorescence amounts of caspase activity of MCF-7 and MDA-MB-231 cells after treatment with a combination of VLM and DPQ (2.5 µg/ml VLM + 1.5 µg/ml DPQ) for 0–72 h are presented. There was a significant increase in experimental group according to control (P<0.05).

Table V.

Fluorescence values at 450–655 nm (emission) of MCF-7 cells treated with 2.5 µg/ml Valamor + 1.5 µg/ml DPQ reflecting caspase activity.

Table V.

Fluorescence values at 450–655 nm (emission) of MCF-7 cells treated with 2.5 µg/ml Valamor + 1.5 µg/ml DPQ reflecting caspase activity.

Time, hControlCombination treatment
24204±12302±14a
48212±11328±15a
72307±11518±13a

a P<0.05 vs. control.

Table VI.

Fluorescence values at 450–655 nm (emission) of MDA-MB-231 cells treated with 2.5 µg/ml Valamor + 1.5 µg/ml DPQ reflecting caspase activity.

Table VI.

Fluorescence values at 450–655 nm (emission) of MDA-MB-231 cells treated with 2.5 µg/ml Valamor + 1.5 µg/ml DPQ reflecting caspase activity.

Time, hControlCombination treatment
24224±12361±14a
48226±11427±15a
72231±11453±13a

a P<0.05 vs. control.

Discussion

The primary goal of treatment for advanced or metastatic BC is to slow the disease's progression, ideally using patient-friendly anticancer medications that do not cause any undue damage (25). Cell cycle progression is deregulated in cancer, which is characterized by unchecked cell proliferation (26). The mitotic cell division cycle consists of four phases: Mitosis (M), the phase during which cellular DNA is synthesized (S), the first gap phase (G1) between the M and S phases, and the G2 phase between the S and M phases. To facilitate cell cycle progression, which is essential for mammalian cell cycle regulation, cell cycle-related proteins are phosphorylated by cyclins A, B, D and E, and their associated CDK1, −2, −4 and −6 (27,28). Although several distinct genes encode the 3 D-type cyclins, D1, D2 and D3 have a common set of amino acids (on average 57% across the coding area) (29). CDK4/6 inhibitors are a novel family of pharmaceuticals that decrease cell cycle progression. Tumor cell growth is halted in this manner. Palbociclib, ribociclib and abemaciclib are three such inhibitors that have been authorized lately for the treatment of BC in different contexts and combination regimens (30).

After having demonstrated noticeably improved progression-free survival outcomes in comparison to standard therapy, ribociclib is one of three selective small-molecule inhibitors of CDK4/6 that are currently approved for the treatment of advanced hormone receptor-positive, HER2-negative BC (3133). Third-generation CDK4/6 inhibitor ribociclib (LEE011) is highly selective and inhibits CDK4/6 by competitively interacting with its ATP binding sites (19). Ribociclib can halt the spread of cancer by inhibiting these enzymes, which also reduce the proliferation of cancer cells (34).

Beyond the locally advanced/metastatic scenario, the introduction of PARP inhibitors may offer advantages for the treatment of BC (35). The oral version of PARP inhibitors has the potential to enhance patient experience and adherence (36).

The activity of CDKs is required for DNA end resection. Numerous investigations revealed that CDKs were crucial to PARP inhibitor resistance (3742).

TNBC cells that had been resistant to niraparib were made sensitive again by the CDK inhibitor dinaciclib. The experiment, which included dinaciclib and niraparib, was effective not just in TNBC cells but also in cells from the pancreas, ovary, prostate, colon and lung cancers (43).

Furthermore, out of all the functioning cell-cycle complexes, the CDK4/6 complex had the strongest negative connection with mutations, indicating that combination suppression of CDK4/6 and PARP may work in concert. In addition, combined therapy demonstrated a reactive oxygen species-dependent synergy in both Rb-proficient and Rb-deficient BC cells. These results point to a possible therapeutic approach to increase the effectiveness of PARP and CDK4/6 inhibitors in the treatment of cancer (44).

In a study conducted with the HCC1937 cell line, a CDK4/6 inhibitor was used together with a PARP inhibitor. The results of the study showed that the combination of CDK4/6 inhibitor and PARP inhibitor may expand the use of these inhibitors in patients with TNBC and potentially overcome PARP inhibitor resistance (45). Several recent studies have shown how CDK4/6 and PARP inhibitors work synergistically in various cancer cells (4648).

In the current study, consistent with the studies mentioned above, VLM and DPQ showed antiproliferative effects on both MCF-7 and MDA-MB-231 cells at the lowest concentration combinations used. The combined concentrations were shown to induce DNA damage in both target cell lines. In conclusion, the findings of this research investigating the inhibitory effects of VLM and demonstrated significant efficacy to reduce the viability of MCF-7 and MDA-MB231 cell lines, the cell index, mitotic signs and BrdU labeling, as well as a significant increase in caspase activity in these cell lines. The lack of cell proliferation after 72 h of treatment may be due to the cells becoming stable, or it may be due to the cells entering the process of apoptosis when caspase activity is taken into account.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Scientific Research Projects Coordination Unit of Istanbul University (project no. FYL-2022-39390).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

EP and MT performed the experiments. EP and MT wrote and edited the manuscript. EP and MT confirm the authenticity of all the raw data. Both authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin. 68:7–30. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Veronesi U, Boyle P, Goldhirsch A, Orecchia R and Viale G: Breast cancer. Lancet. 365:1727–1741. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Cliffton EE and Young LE: Carcinoma of the breast; five to twenty-year follow-up following radical mastectomy. Am J Surg. 82:185–190. 1951. View Article : Google Scholar : PubMed/NCBI

6 

Figueiredo MI, Cullen J, Hwang YT, Rowland JH and Mandelblatt JS: Breast cancer treatment in older women: Does getting what you want improve your long-term body image and mental health? J Clin Oncol. 22:4002–4009. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Mathews FS: The ten-year survivors of radical mastectomy. Ann Surg. 98:635–643. 1993. View Article : Google Scholar : PubMed/NCBI

8 

Nagarajan D and McArdle SEB: Immune landscape of breast cancers. Biomedicines. 6:202018. View Article : Google Scholar : PubMed/NCBI

9 

Makhoul I, Atiq M, Alwbari A and Kieber-Emmons T: Breast cancer immunotherapy: An update. Breast Cancer (Auckl). 12:11782234187748022018.PubMed/NCBI

10 

Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y and Pietenpol JA: Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Investig. 121:2750–2767. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Liedtke C, Mazouni C, Hess KR, André F, Tordai A, Mejia JA, Symmans WF, Gonzalez-Angulo AM, Hennessy B, Green M, et al: Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J Clin Oncol. 26:1275–1281. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Chen X, Xu D, Li X, Zhang J, Xu W, Hou J, Zhang W and Tang J: Latest overview of the cyclindependent kinases 4/6 inhibitors in breast cancer: The past, the present, and the future. J Cancer. 10:6608–6617. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Hulka BS: Epidemiology of susceptibility to breast cancer. Prog Clin Biol Res. 395:159–174. 1996.PubMed/NCBI

14 

Reinert T and Barrios CH: Optimal management of hormone receptor positive metastatic breast cancer in 2016. Ther Adv Med Oncol. 7:304–320. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Wuerstlein R and Harbeck N: Neoadjuvant therapy for HER2-positive breast cancer. Rev Recent Clin Trials. 12:81–92. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Riggi N, Aguet M and Stamenkovic I: Cancer metastasis: A reappraisal of its underlying mechanisms and their relevance to treatment. Annu Rev Pathol. 13:117–140. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Weigelt B, Peterse JL and van't Veer LJ: Breast cancer metastasis: Markers and models. Nat Rev Cancer. 5:591–602. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Scully OJ, Bay BH, Yip G and Yu Y: Breast cancer metastasis. Cancer Genomics Proteomics. 9:311–320. 2012.PubMed/NCBI

19 

Poratti M and Marzaro G: Third-generation CDK inhibitors: A review on the synthesis and binding modes of palbociclib, ribociclib, and abemaciclib. Eur J Med Chem. 172:143–153. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Luo X and Kraus WL: A one and a two … expanding roles for poly(ADP-ribose) polymerases in metabolism. Cell Metab. 13:353–355. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Zhou D, Chu W, Xu J, Jones LA, Peng X, Li S, Chen DL and Mach RH: Synthesis, [18F] radiolabeling, and evaluation of poly (ADP-ribose) polymerase-1 (PARP-1) inhibitors for in vivo imaging of PARP-1 using positron emission tomography. Bioorg Med Chem Lett. 22:1700–1707. 2014. View Article : Google Scholar

22 

Giannini G, Battistuzzi G, Vesci L, Milazzo FM, Paolis FD, Barbarino M, Guglielmi MB, Carollo V, Gallo G, Artali R and Dallavalle S: Novel PARP-1 inhibitors based on a 2-propanoyl-3H-quinazolin-4-one scaffold. Bioorg Med Chem Lett. 24:462–466. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Berger NA: Poly(ADP-ribose) in the cellular response to DNA damage. Radiat Res. 101:4–15. 1985. View Article : Google Scholar : PubMed/NCBI

24 

Topçul M, Çeti N İL, Özbaş Turan S and Kolusayin Ozar MÖ: In vitro cytotoxic effect of PARP inhibitor alone and in combination with nab-paclitaxel on triple-negative and luminal A breast cancer cells. Oncol Rep. 40:527–535. 2018.PubMed/NCBI

25 

Silberholz J, Bertsimas D and Vahdat L: Clinical benefit, toxicity and cost of metastatic breast cancer therapies: Systematic review and meta-analysis. Breast Cancer Res Treat. 176:535–543. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Malumbres M and Barbacid M: Cell cycle, CDKs, and cancer: A changing paradigm. Nat Rev Cancer. 9:153–166. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Sherr CJ, Beach D and Shapiro GI: Targeting CDK4 and CDK6: From discovery to therapy. Cancer Discov. 6:353–367. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Xiong Y, Menninger J, Beach D and Ward DC: Molecular cloning and chromosomal mapping of CCND genes encoding human D-type cyclins. Genomics. 13:575–584. 1992. View Article : Google Scholar : PubMed/NCBI

30 

Braal CL, Jongbloed EM, Wilting SM, Mathijssen RHJ, Koolen SLW and Jager A: Inhibiting CDK4/6 in breast cancer with palbociclib, ribociclib, and abemaciclib: Similarities and differences. Drugs. 81:317–331. 2021. View Article : Google Scholar : PubMed/NCBI

31 

Hortobagyi GN, Stemmer SM, Burris HA, Yap YS, Sonke GS, Paluch-Shimon S, Campone M, Petrakova K, Blackwell KL, Winer EP, et al: 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. 29:1541–1547. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Slamon DJ, Neven P, Chia S, Fasching PA, De Laurentiis M, Im SA, Petrakova K, Bianchi GV, Esteva FJ, Martín M, et al: 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. 36:2465–2472. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Tripathy D, Im SA, Colleoni M, Franke F, Bardia A, Harbeck N, Hurvitz SA, Chow L, Sohn J, Lee KS, et al: Ribociclib plus endocrine therapy for premenopausal women with hormone-receptor-positive, advanced breast cancer (MONALEESA-7): A randomised phase 3 trial. Lancet Oncol. 19:904–915. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Fang H, Huang D, Yang F and Guan X: Potential biomarkers of CDK4/6 inhibitors in hormone receptor-positive advanced breast cancer. Breast Cancer Res Treat. 168:287–297. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Cortesi L, Rugo HS and Jackisch C: An overview of PARP ınhibitors for the treatment of breast cancer. Target Oncol. 16:255–282. 2021. View Article : Google Scholar : PubMed/NCBI

36 

Eek D, Krohe M, Mazar I, Horsfeld A, Pompilus F, Friebe R and Shields AL: Patient-reported preferences for oral versus intravenous administration for the treatment of cancer: A review of the literature. Patient Prefer Adherence. 10:1609–1621. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Tomimatsu N, Mukherjee B, Catherine Hardebeck M, Ilcheva M, Vanessa Camacho C, Louise Harris J, Porteus M, Llorente B, Khanna KK and Burma S: Phosphorylation of EXO1 by CDKs 1 and 2 regulates DNA end resection and repair pathway choice. Nat Commun. 5:35612014. View Article : Google Scholar : PubMed/NCBI

38 

Bajrami I, Frankum JR, Konde A, Miller RE, Rehman FL, Brough R, Campbell J, Sims D, Rafiq R, Hooper S, et al: Genome-wide profiling of genetic synthetic lethality identifies CDK12 as a novel determinant of PARP1/2 inhibitor sensitivity. Cancer Res. 74:287–297. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Joshi PM, Sutor SL, Huntoon CJ and Karnitz LM: Ovarian cancer-associated mutations disable catalytic activity of CDK12, a kinase that promotes homologous recombination repair and resistance to cisplatin and poly(ADP-ribose) polymerase inhibitors. J Biol Chem. 289:9247–9253. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Johnson SF, Cruz C, Greifenberg AK, Dust S, Stover DG, Chi D, Primack B, Cao S, Bernhardy AJ, Coulson R, et al: CDK12 ınhibition reverses de novo and acquired PARP inhibitor resistance in BRCA wild-type and mutated models of triple-negative breast cancer. Cell Rep. 17:2367–2381. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Ning JF, Stanciu M, Humphrey MR, Gorham J, Wakimoto H, Nishihara R, Lees J, Zou L, Martuza RL, Wakimoto H and Rabkin SD: Myc targeted CDK18 promotes ATR and homologous recombination to mediate PARP inhibitor resistance in glioblastoma. Nat Commun. 10:29102019. View Article : Google Scholar : PubMed/NCBI

42 

Militello AM, Zielli T, Boggiani D, Michiara M, Naldi N, Bortesi B, Zanelli P, Uliana V, Giuliotti S and Musolino A: Mechanism of action and clinical efficacy of CDK4/6 inhibitors in BRCA-mutated, estrogen receptor-positive breast cancers: Case report and literature review. Front Oncol. 9:7592019. View Article : Google Scholar : PubMed/NCBI

43 

Carey JPW, Karakas C, Bui T, Chen X, Vijayaraghavan S, Zhao Y, Wang J, Mikule K, Litton JK, Hunt KK and Keyomarsi K: Synthetic lethality of PARP inhibitors in combination with MYC blockade is independent of BRCA status in triple-negative breast Cancer. Cancer Res. 78:742–757. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Li S, Zhang Y, Wang N, Guo R, Liu Q, Lv C, Wang J, Wang L and Yang Q: Pan-cancer analysis reveals synergistic effects of CDK4/6i and PARPi combination treatment in RB-proficient and RB-deficient breast cancer cells. Cell Death Dis. 11:2192020. View Article : Google Scholar : PubMed/NCBI

45 

Eskiler GG, Ozman Z, Haciefendi A and Cansaran-Duman D: Novel combination treatment of CDK 4/6 inhibitors with PARP inhibitors in triple negative breast cancer cells. Naunyn Schmiedebergs Arch Pharmacol. 396:1031–1041. 2023. View Article : Google Scholar : PubMed/NCBI

46 

Li H, Liu ZY, Wu N, Chen YC, Cheng Q and Wang J: PARP inhibitor resistance: The underlying mechanisms and clinical implications. Mol Cancer. 19:1072020. View Article : Google Scholar : PubMed/NCBI

47 

Klein FG, Granier C, Zhao Y, Pan Q, Tong Z, Gschwend JE, Holm PS and Nawroth R: Combination of talazoparib and palbociclib as a potent treatment strategy in bladder cancer. J Pers Med. 11:3402021. View Article : Google Scholar : PubMed/NCBI

48 

Zhu X, Chen L, Huang B, Li X, Yang L, Hu X, Jiang Y, Shao Z and Wang Z: Efficacy and mechanism of the combination of PARP and CDK4/6 inhibitors in the treatment of triple-negative breast cancer. J Exp Clin Cancer Res. 40:1222021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2024
Volume 27 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Pulat E and Pulat E: Effects of combined use of ribociclib with PARP1 inhibitor on cell kinetics in breast cancer. Oncol Lett 27: 243, 2024
APA
Pulat, E., & Pulat, E. (2024). Effects of combined use of ribociclib with PARP1 inhibitor on cell kinetics in breast cancer. Oncology Letters, 27, 243. https://doi.org/10.3892/ol.2024.14376
MLA
Pulat, E., Topçul, M. R."Effects of combined use of ribociclib with PARP1 inhibitor on cell kinetics in breast cancer". Oncology Letters 27.6 (2024): 243.
Chicago
Pulat, E., Topçul, M. R."Effects of combined use of ribociclib with PARP1 inhibitor on cell kinetics in breast cancer". Oncology Letters 27, no. 6 (2024): 243. https://doi.org/10.3892/ol.2024.14376