Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with unfavorable outcomes. Developing therapeutic targets for TNBC remains a challenge. Here, we identified that acetyl-CoA acyltransferase 1 (ACAA1) is highly expressed in the luminal androgen receptor (LAR) subtype of TNBC compared with adjacent normal tissues in our TNBC proteomics dataset. Inhibition of ACAA1 restrained TNBC proliferation and potentiated the response to the cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitor abemaciclib. Mechanistically, ACAA1 interacted with CDK4, and the inhibition of ACAA1 blocked RB transcriptional corepressor 1 (RB1) phosphorylation, resulting in G1–S cell-cycle arrest. Importantly, trimetazidine, a traditional drug for ischemic heart disease, caused a decrease in ACAA1 protein levels and enhanced the efficacy of abemaciclib in preclinical TNBC models. In conclusion, this study identifies that ACAA1 is a therapeutic target in TNBC and suggests the combination of trimetazidine and abemaciclib could be beneficial for ACAA1-high TNBCs.

Significance:

ACAA1 is highly expressed in TNBC, serving as a potential therapeutic target in ACAA1-high tumors and a predictive biomarker of resistance to CDK4/6 inhibitors for RB1-proficient patients.

Triple-negative breast cancer (TNBC) is characterized by the absence of the estrogen receptor (ER) and progesterone receptor and the lack of HER2 overexpression (1). TNBC accounts for approximately 10%–20% of newly diagnosed breast cancers and is associated with aggressive behavior and poor prognosis (2). In the past decades, advances in omics technologies have led to a comprehensive view of TNBC tumors and microenvironment heterogeneity (3). Although multiple compounds have been evaluated in clinical trials, chemotherapy remains the mainstay systemic treatment for TNBC (3). Currently, only a limited number of targeted agents have been approved for patients with breast cancer, including TNBCs. For example, PARP inhibitors olaparib and talazoparib for patients with HER2 breast cancer carrying deleterious germline in breast cancer mutations (4–6), immune checkpoint inhibitors pembrolizumab and atezolizumab for PD-L1–positive patients with TNBC (7–9), antibody–drug conjugate (ADC) sacituzumab govitecan-hziy (Trodelvy) for third-line treatment of unresectable locally advanced or metastatic TNBC (10), and fam-trastuzumab-deruxtecan-nxki (Enhertu) for HER2-low breast cancer (11). Given that the current therapeutic scenario does not benefit all TNBCs and treatment efficacy requires further improvement, exploring novel treatment strategies in TNBC remains a major challenge.

In our previous study, we constructed a cohort of 465 patients with primary TNBC and classified TNBC into four transcriptome-based subtypes (12). We further revealed the potential therapeutic targets of each subtype and conducted a phase Ib/II clinical trial (FUTURE, ClinicalTrials.gov, number: NCT03805399) to evaluate the efficacy and safety of the proposed treatment strategies (13). Although some patients experienced favorable outcomes, a low objective response rate was observed in patients with the luminal androgen receptor (LAR) subtype treated with androgen receptor (AR) inhibitor and cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitor. Currently, CDK4/6 inhibitors are not the standard of care for TNBC, and the application of CDK4/6 inhibitors is more challenging in patients with TNBC than in ER+ patients with breast cancer due to the high frequency of RB transcriptional corepressor 1 (RB1) loss/mutations and high expression of CDK2, CDK6, CCNE1, and CDKN2A in TNBCs (14). However, increasing evidence suggests that a proportion of patients with TNBC may benefit from CDK4/6 inhibitors, although the underlying mechanism remains unclear (15–17). Therefore, there is an urgent need for studies exploring potential biomarkers to select patients with TNBC most likely to benefit from CDK4/6 inhibitors and to identify novel therapeutic targets.

We conducted a proteome-centric study to explore potential protein targets for TNBC, especially for the LAR subtype, that showed a low response rate to AR inhibitor and CDK4/6 inhibitor treatment in the FUTURE trial (13, 18). We observed high expression of acetyl-CoA acyltransferase 1 (ACAA1) and other key molecules associated with fatty acid metabolism in the integrative proteomic (iP)-2 subtype, that is mainly comprised of the transcriptomic LAR subtype. In this study, we uncovered the biological functions and mechanisms by which ACAA1 promotes TNBC proliferation and diminishes the tumor response to the CDK4/6 inhibitor abemaciclib and explored the potential of ACAA1 as a therapeutic target in TNBC.

Human tissues and public datasets

Proteomic profiles of 82 TNBC tissues and 66 paired adjacent normal tissues were obtained from patients who were treated at Fudan University Shanghai Cancer Center (FUSCC; Shanghai, China) from July 2010 to September 2014 (18). Ten pairs of primary breast tumor tissues and adjacent normal tissues were obtained from patients with TNBC who underwent surgery at FUSCC from April 2017 to August 2018. In addition, fresh tumor samples from three patients with TNBC who underwent surgery at FUSCC between October 13, 2021, and December 2, 2021, were collected for the generation of patient-derived organoids. Public datasets were obtained from Kaplan–Meier plotter (http://kmplot.com/analysis/), FUSCC TNBC, and The Cancer Genome Atlas (TCGA) for analysis (12, 14, 18–21).

Cell lines

The human mammary epithelial cell lines MCF10A and HMEC, human TNBC cell lines BT-549, HCC70, Hs-578T, MDA-MB-453, HCC38, HCC1143, MDA-MB-157, MDA-MB-436, MDA-MB-468, BT-20, HCC1937, CAL-148, and human embryonic kidney cells (HEK293T) were obtained from ATCC. MFM-223 cells were obtained from the European Collection of Authenticated Cell Cultures. The human breast cancer cell line MDA-MB-231 and its lung-tropic derivative LM2–4175 were gifted from Professor Guohong Hu (Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China). Only cells within 6 months of being thawed were used and maintained at 37°C with 5% carbon dioxide (CO2). All cell lines were authenticated by short tandem repeat profiling and tested free of Mycoplasma contamination.

Organoid preparation

Generation of patient-derived organoids was performed as described in a previous study (22). Breast cancer tissues were cut into 1–3 mm3 pieces and digested in collagenase (Sigma).

Chemicals and antibodies

Abemaciclib mesylate (S7158), trimetazidine dihydrochloride (S4543), MG-132 (S2619), bafilomycin A1 (Baf-A1, S1413), and 3-methyladenine (3-MA, S2767) were purchased from Selleck. Ammonium chloride (NH4Cl, 213330) was purchased from Sigma. Protease and phosphatase inhibitors (Roche) were purchased from Merck. All primary and secondary antibodies used in this study are summarized in Supplementary Table S1.

In vitro cell viability assay

Short-term viability assays were performed by using an IncuCyte ZOOM HD/2CLR system or Cell Counting Kit-8 (CCK-8) assays. For the IncuCyte system, cells were seeded in 96-well plates and imaged using the IncuCyte ZOOM system (Essen BioScience). Frames were captured at 12-hour intervals in three separate regions per well. Cultures were maintained in a 37°C incubator with 5% CO2, and the growth rate was analyzed using IncuCyte software (2013A Rev2). For the CCK-8 assay, cells were seeded in 96-well plates and incubated with 10% CCK-8 solution (Dojindo, CK04) at 37°C for 2 hours, and then the absorbance was measured at 450 nm.

For colony formation assays, cells were seeded into 6-well or 24-well plates and cultured for 2 weeks. The cells were rinsed with PBS, fixed and stained with a 0.25% crystal violet solution in methanol for 30 minutes for colony counting.

Flow cytometry analysis

The cells were stained with propidium iodide (BD Biosciences) for 30 minutes at 37°C and were analyzed using a CytoFLEX S Flow Cytometer (Beckman Coulter) with Modfit LT5.0 software.

Growth inhibition assay and drug synergism analysis

Cells were seeded into 96-well plates (1–6 × 103 cells/well) and incubated 24 hours before the administration of drugs. The growth medium was then replaced with a medium containing sterile water or increasing concentrations of abemaciclib (9 doses ranging from 0.078 μmol/L to 20 μmol/L, 2-fold dilution) or trimetazidine (9 doses ranging from 0.156 mmol/L to 40 mmol/L, 2-fold dilution), and plates were incubated for three days. Growth was quantified using the CCK-8 assay (Dojindo, CK04) according to the manufacturer’s instructions. The IC50 values were determined by probit regression using the Logit model with SPSS version 22.0 (SPSS). The combination index (CI) value was calculated to evaluate the effects of the two drugs using the Chou–Talalay method with CompuSyn software (CI > 1, antagonism; CI = 1, addition; CI < 1, synergism; ref. 23).

Plasmids

The pcDNA3.1-Flag-ACAA1 (CN24874) and pcDNA3.1-HA-CDK4 (CN26246) vectors were purchased from Synbio Technologies. Various truncated mutants of CDK4 and ACAA1 were constructed from the full-length vector. The ACAA1 cDNA (NM_001607) was subcloned into the pCDH-CMV-MCS-EF1-puro vector to generate the Flag-ACAA1 expression vector. Human ACAA1 shRNAs in the U6-MCS-Ubiquitin-EGFP-IRES-puromycin vector (GV248) were purchased from GeneChem. The sense sequences of shRNAs are listed in Supplementary Table S2.

RNA extraction and qRT-PCR

Total RNA was extracted from cells using TRIzol Reagent (Thermo Fisher Scientific) according to the manufacturer’s protocol. First-strand cDNAs were synthesized from 1 μg of total RNA using HiScript III RT SuperMix for qPCR with genomic DNA wiper (Vazyme, R323–01). qRT-PCR was performed using ChamQ SYBR qPCR Master Mix (Vazyme, R311–02) on the QuantStudio 5 Real-Time PCR System (Thermo Fisher Scientific). The primer sequences are listed in Supplementary Table S3. The results were analyzed with QuantStudio Design & Analysis software (v1.5.1) and the 2−ΔΔCt method.

Immunoblotting analysis

Proteins were separated by SDS-PAGE and transferred onto polyvinylidene difluoride (PVDF) membranes (Millipore) that were incubated with the indicated primary and secondary antibodies. Corresponding antibody-specific signals were detected with enhanced chemiluminescence substrate (Thermo Fisher Scientific). The signals were captured by an Amersham Imager 600 with AJ600 control software and quantified with ImageJ software.

Immunoprecipitation and coimmunoprecipitation assays

Cells were homogenized in lysis buffer (50 mmol/L Tris; pH 7.4, 150 mmol/L NaCl, 1% NP-40, 0.25% sodium pyrophosphate, 0.02% NaN3, 5 mmol/L EDTA, 50 mmol/L NaF, 1 mmol/L Na3VO4) containing protease and phosphatase inhibitor cocktails (Roche). For immunoprecipitation (IP) assays, cell extracts were incubated with a rabbit anti-ACAA1 antibody or normal rabbit IgG (Cell Signaling Technology, catalog no. 2729) overnight at 4°C on a rotating platform, followed by incubation with Dynabeads Protein A (Thermo Fisher Scientific) for more than 4 hours at 4°C. For coimmunoprecipitation assays, cell extracts were immunoprecipitated with anti-Flag M2 magnetic beads (Sigma) or anti-HA magnetic beads (Thermo Fisher Scientific) at 4°C overnight. The immunoprecipitates were thoroughly washed with lysis buffer five times, eluted with SDS loading buffer, boiled at 99°C for 5 minutes, and then immunoblotted with the indicated antibodies.

Immunofluorescence

Cells were rinsed in PBS three times, fixed with 4% paraformaldehyde (PFA) at room temperature for 30 minutes, and permeabilized using 0.1% Triton X-100 in 1% BSA in PBS on ice for 10 minutes. After being blocked with 5% BSA at room temperature for 1 hour, the cells were incubated overnight at 4°C with an anti-ACAA1 mouse antibody and an anti-CDK4 rabbit antibody. After incubation with the fluorescein-conjugated secondary antibody, the cells were mounted with ProLong Gold Antifade Mountant with 4',6-diamidino-2-phenylindole (DAPI; Thermo Fisher Scientific).

Xenograft studies

Animals were used in accordance with the Institutional Animal Care and Use Committee of SLAC Guide for Care and Use of Laboratory Animals (protocol number: 20210810012 and 20220808016). For the in vivo mammary fat pad xenograft model, 8 × 105 LM2–4175 cells stably expressing negative control (shNC) or shACAA1 vectors were injected into the mammary fat pads of six-week-old female NOD/SCID mice (Shanghai SLAC Laboratory Animal Co., Ltd.). Tumors were monitored until they reached an average size of 50 mm3, at which point, mice were randomly grouped (n = 8 per group) and treatments were initiated. Abemaciclib and trimetazidine were freshly dissolved in PBS at concentrations of 5 mg/mL and 50 mg/mL, respectively. Mice were administered abemaciclib (25 mg/kg; oral gavage) and trimetazidine (250 mg/kg, i.p. injection) once daily, alone or in combination. PBS was administered by oral gavage, i.p. injection, or both routes to obtain consistency among the groups. Tumor diameters were measured every other day with electric calipers, and tumor volumes were calculated as follows: tumor volume = 0.5 × (length) × (width)2. Animal weights were recorded every other day after drug administration. When the mean tumor volumes in the control group reached approximately 1000 mm3, the experiment was ceased, and tumors were harvested and imaged.

IHC

Endogenous peroxidase activity was blocked with 3% hydrogen peroxide after deparaffinization and rehydration of the slides. Antigen retrieval was performed by incubating the slides with citrate buffer (pH 6.0) at 100°C for 15 minutes. The slides were blocked with 10% normal goat serum for 1 hour at room temperature and incubated overnight at 4°C with a primary antibody. The slides were incubated for 30 minutes with horseradish peroxidase (HRP)-conjugated secondary antibody (Gene Tech) at room temperature. Sections were then incubated with 3,3′-diaminobenzidine substrate (Gene Tech) for visualization and counterstained with hematoxylin for 2 minutes. The slides were then dehydrated in an ascending ethanol series before clearing with xylene and mounting under a coverslip. Adjacent tumor sections were selected and stained with hematoxylin and eosin. The positive staining density was measured using a computerized imaging system composed of an Olympus DP27 charge-coupled device camera connected to a phase-contrast Olympus BX43 light microscope (×40/0.65 magnification). Staining scores were determined using the H-score system that takes both intensity and percentage positivity into account using the formula: 3 × (% of 3+ cells) + 2 × (% of 2+ cells) + 1 × (% of 1+ cells), with a final score ranging from 0 to 300.

Ethical approval

All procedures involving patients were conducted in accordance with the Declaration of Helsinki and were approved by the Institutional Ethics Review Board of FUSCC. Written informed consent was obtained from all of the patients.

Statistical analysis

Quantification and statistical analyses were performed with GraphPad Prism 8.0 (GraphPad Software, Inc.) and SPSS version 22.0 (SPSS). A two-tailed t test, ANOVA, or Mann–Whitney Wilcoxon test was employed for in vitro and in vivo studies (indicated in the figure legends). The correlation coefficients between ACAA1 protein and mRNA expression, as well as between ACAA1 protein levels and IC50 values of trimetazidine, were calculated using Spearman rank correlation analysis. The survival curves were constructed using the Kaplan–Meier method and compared with the log-rank test using the autoselected best cutoff value to define high and low ACAA1 expression. A P < 0.05 was considered statistically significant.

Data availability

The previously published RNA sequencing (RNA-seq) and proteomic data of the FUSCC TNBC cohort analyzed in this study were obtained from the data repository The National Omics Data Encyclopedia (NODE) at http://www.biosino.org/node/project/detail/OEP000155 and OEP002770. The ACAA1 mRNA expression data and survival information of the TCGA TNBC cohort were downloaded from cBioPortal for Cancer Genomics (http://www.cbioportal.org/). The data of the Kaplan–Meier plotter online tool analyzed in this study were obtained at http://kmplot.com/analysis/. The RNA-seq data generated in this study are publicly available in the NCBI sequence read archive (SRA) at https://www.ncbi.nlm.nih.gov/sra/PRJNA908232. The survival data of the FUSCC TNBC cohort are not publicly available due to patient privacy requirements but are available upon reasonable request from the corresponding author.

Other methods

Methods used for Supplementary figures are provided in the Supplementary Methods.

ACAA1 was highly expressed and predicted poor survival in TNBC

A total of 7,531 proteins were obtained from the proteomic profiles of 82 TNBC tumors and 66 paired adjacent normal tissues after quantification assessment and quality control using a two-component Gaussian mixed model as described in our previous study (18). Data from the FUSCC TNBC cohort were screened to uncover potential therapeutic targets for TNBC, particularly for the LAR subtype (Fig. 1A). First, proteins present in less than 30% of samples were eliminated, leaving 1,777 proteins. Then, 26 proteins with higher expression levels in the LAR subtype than in the other subtypes (log2-fold change ≥ 1, FDR ≤ 0.05) were chosen. Finally, after comparing the protein abundance of the 26 candidates between tumors and normal tissues, the top three candidates were nicotinamide phosphoribosyltransferase (NAMPT), sterol carrier protein-2 (SCP2), and ACAA1. Previous studies have reported that NAMPT and SCP2 could promote tumor progression (24, 25). Thus, we chose ACAA1 for further study, as it was an FDA-approved drug target with no TNBC-related basic research previously reported (26).

Figure 1.

ACAA1 was highly expressed and predicted poor survival in TNBC. A, Flow chart of the method used to screen potential therapeutic targets in the FUSCC TNBC cohort. B, Protein abundance of ACAA1 in the LAR subtype (n = 19) and the other subtypes (n = 62) in the FUSCC TNBC cohort. ACAA1 protein was quantified as a normalized intensity-based absolute quantification (iBAQ) value and log2 transformed. For the boxplots, the line in the box indicates the median, boxes correspond with the first and third quartiles, and dots show all data values. C, Protein abundance of ACAA1 in breast tumors (n = 81) and adjacent normal tissues (n = 52) in the FUSCC TNBC cohort. D, The mRNA expression levels of ACAA1 in the LAR subtype (n = 81) and the other subtypes (n = 279) in the FUSCC TNBC cohort. FPKM, fragments per kilobase of exon model per million mapped fragments. For the boxplots, the line in the box indicates the median; boxes correspond to the first and third quartiles, and dots show all data values. E, The mRNA expression levels of ACAA1 in breast tumors (n = 360) and adjacent normal tissues (n = 88) in the FUSCC TNBC cohort. F, Correlation between protein abundance and mRNA expression levels of ACAA1 in the FUSCC TNBC cohort, as determined by Spearman rank correlation analysis. G–I, Kaplan–Meier analysis of the relapse-free survival (G), distant metastasis-free survival (H), and OS (I) of patients with high versus low ACAA1 mRNA expression levels in the publicly available TNBC cohort. The P values are based on the log-rank test. HR, hazard ratio. J, Lysates from 10 primary TNBC tissues (T) and adjacent normal tissues (N) were immunoblotted with ACAA1 and vinculin antibodies. K, Normal human mammary epithelial cell lines and 12 TNBC cell lines were immunoblotted with antibodies against ACAA1 and α-tubulin. The P value was calculated using the Mann–Whitney U Wilcoxon test. ***, P < 0.001.

Figure 1.

ACAA1 was highly expressed and predicted poor survival in TNBC. A, Flow chart of the method used to screen potential therapeutic targets in the FUSCC TNBC cohort. B, Protein abundance of ACAA1 in the LAR subtype (n = 19) and the other subtypes (n = 62) in the FUSCC TNBC cohort. ACAA1 protein was quantified as a normalized intensity-based absolute quantification (iBAQ) value and log2 transformed. For the boxplots, the line in the box indicates the median, boxes correspond with the first and third quartiles, and dots show all data values. C, Protein abundance of ACAA1 in breast tumors (n = 81) and adjacent normal tissues (n = 52) in the FUSCC TNBC cohort. D, The mRNA expression levels of ACAA1 in the LAR subtype (n = 81) and the other subtypes (n = 279) in the FUSCC TNBC cohort. FPKM, fragments per kilobase of exon model per million mapped fragments. For the boxplots, the line in the box indicates the median; boxes correspond to the first and third quartiles, and dots show all data values. E, The mRNA expression levels of ACAA1 in breast tumors (n = 360) and adjacent normal tissues (n = 88) in the FUSCC TNBC cohort. F, Correlation between protein abundance and mRNA expression levels of ACAA1 in the FUSCC TNBC cohort, as determined by Spearman rank correlation analysis. G–I, Kaplan–Meier analysis of the relapse-free survival (G), distant metastasis-free survival (H), and OS (I) of patients with high versus low ACAA1 mRNA expression levels in the publicly available TNBC cohort. The P values are based on the log-rank test. HR, hazard ratio. J, Lysates from 10 primary TNBC tissues (T) and adjacent normal tissues (N) were immunoblotted with ACAA1 and vinculin antibodies. K, Normal human mammary epithelial cell lines and 12 TNBC cell lines were immunoblotted with antibodies against ACAA1 and α-tubulin. The P value was calculated using the Mann–Whitney U Wilcoxon test. ***, P < 0.001.

Close modal

The protein abundance and mRNA expression levels of ACAA1 were higher in the LAR subtype (n = 19 for protein; n = 81 for mRNA) than in other subtypes and were higher in TNBC tumors than in adjacent normal tissues (Fig. 1BE, Supplementary Fig. S1A and S1B). The ACAA1 protein abundance and mRNA expression levels were positively correlated according to Spearman rank correlation analysis (R = 0.386, P < 0.001; Fig. 1F). Kaplan–Meier survival analysis using public databases indicated that higher ACAA1 mRNA expression levels were associated with worse relapse-free survival, distant metastasis-free survival and overall survival (OS) rates in patients with TNBC (Fig. 1GI). Similarly, higher ACAA1 protein abundance was associated with reduced OS in the publicly available breast cancer cohort (Supplementary Fig. S1C), and higher ACAA1 mRNA expression in the TCGA TNBC cohort was associated with shorter relapse-free survival and OS (Supplementary Fig. S1D and S1E). To confirm the ACAA1 protein expression pattern, we collected 10 pairs of primary TNBC tumors and adjacent normal tissues. Quantitative analysis showed that ACAA1 protein levels were much higher in TNBC tumors than in the corresponding normal tissues (Fig. 1J; Supplementary Fig. S1F). We examined ACAA1 protein levels in two human mammary epithelial cell lines and 12 TNBC cell lines using immunoblotting. We observed that ACAA1 was highly expressed in several TNBC cell lines compared with that in HMECs and MCF10A cells (Fig. 1K; Supplementary Fig. S1G). In our following experiments, different cell lines were chosen on the basis of the purpose of each experiment and the expression pattern of ACAA1 or other certain proteins. In summary, our findings demonstrate that ACAA1 is highly expressed in TNBC and is associated with poor prognosis.

Inhibition of ACAA1 restrained TNBC proliferation by blocking the G1–S cell-cycle transition

We performed in vitro cell proliferation and colony formation assays in ACAA1-overexpressing and ACAA1-downregulated TNBC cells to explore the biological functions of ACAA1 in TNBC. ACAA1 was found to be expressed at relatively higher levels in LM2–4175, MFM-223 and MDA-MB-453 cell lines (Fig. 1K; Supplementary Figs. S1G and S2A), leading us to choose these cell lines for downregulation experiments, while ACAA1 low-expressing MDA-MB-231 and BT-549 cell lines were used for overexpression experiments. Overexpression of ACAA1 increased the growth rate and clonogenic ability of MDA-MB-231 and BT-549 cells (Fig. 2AC; Supplementary Fig. S2B), whereas downregulation of ACAA1 with shRNAs significantly impaired the proliferative potential and clonogenic ability of TNBC cells (Fig. 2DF; Supplementary Fig. S2C). Flow cytometry revealed a decreased proportion of S-phase cells and an increased proportion of G1 phase cells when ACAA1 was downregulated (Fig. 2G). RB1 plays a key role in regulating G1–S cell-cycle progression (27). Immunoblotting was used to analyze the expression pattern and phosphorylation status of RB1. We observed higher RB1 phosphorylation (Ser780) in cells stably expressing ACAA1 and decreased levels of total and phosphorylated RB1 when ACAA1 was downregulated (Fig. 2H and I; Supplementary Fig. S2D and S2E).

Figure 2.

Inhibition of ACAA1 restrained TNBC proliferation by blocking the G1–S cell-cycle transition. A, MDA-MB-231 and BT-549 cells stably expressing an empty control vector (Vec) or vector expressing Flag-tagged ACAA1 (Flag-ACAA1) were immunoblotted with antibodies against ACAA1 and vinculin. B,In vitro growth curves of MDA-MB-231 and BT-549 cells expressing vector or Flag-ACAA1. Data are presented as the mean ± SD; n = 3 independent experiments. Two-way ANOVA was used. ***, P < 0.001. C, Representative images of surviving colonies of MDA-MB-231 and BT-549 cells stably expressing empty vector or Flag-ACAA1 are shown. n = 3 independent experiments. P values were determined using a two-tailed unpaired Student t test. **, P < 0.01; ***, P < 0.001. D, LM2–4175, MFM-223, and MDA-MB-453 cells stably expressing shNC or two shRNAs targeting ACAA1 (shACAA1) were immunoblotted with antibodies against ACAA1 and vinculin. E,In vitro growth curves of LM2–4175 and MFM-223 cells stably expressing shNC or shACAA1 vectors. The data are presented as the mean ± SD; n = 3 independent experiments. Two-way ANOVA was used. ***, P < 0.001. F, Representative images of surviving colonies of LM2–4175 and MDA-MB-453 cells stably expressing shNC or two shACAA1 vectors are shown. n = 3 independent experiments. P values were determined using a two-tailed unpaired Student t test. ***, P < 0.001. G, Flow cytometry analysis showing the cell-cycle distribution of LM2–4175 and MFM-223 cells expressing shNC or shACAA1 expression vectors. The data are presented as the mean ± SD; n = 3 independent experiments. P values were determined using a two-tailed unpaired Student t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001. H, MDA-MB-231 and BT-549 cells stably expressing vector or Flag-ACAA1 were immunoblotted with the indicated antibodies. I, LM2–4175 cells expressing shNC or shACAA1 vectors were immunoblotted with the indicated antibodies. J, MFM-223 and HCC1143 cells were treated with increasing doses of trimetazidine (TMZ) for 48 hours and analyzed using immunoblotting with the indicated antibodies. K, MFM-223 and HCC1143 cells were treated with increasing doses of trimetazidine for 24 hours and subjected to qRT-PCR analysis. Data are presented as the mean ± SD; n = 3 independent experiments. P values were determined using a two-tailed unpaired Student t test. ns, not significant.

Figure 2.

Inhibition of ACAA1 restrained TNBC proliferation by blocking the G1–S cell-cycle transition. A, MDA-MB-231 and BT-549 cells stably expressing an empty control vector (Vec) or vector expressing Flag-tagged ACAA1 (Flag-ACAA1) were immunoblotted with antibodies against ACAA1 and vinculin. B,In vitro growth curves of MDA-MB-231 and BT-549 cells expressing vector or Flag-ACAA1. Data are presented as the mean ± SD; n = 3 independent experiments. Two-way ANOVA was used. ***, P < 0.001. C, Representative images of surviving colonies of MDA-MB-231 and BT-549 cells stably expressing empty vector or Flag-ACAA1 are shown. n = 3 independent experiments. P values were determined using a two-tailed unpaired Student t test. **, P < 0.01; ***, P < 0.001. D, LM2–4175, MFM-223, and MDA-MB-453 cells stably expressing shNC or two shRNAs targeting ACAA1 (shACAA1) were immunoblotted with antibodies against ACAA1 and vinculin. E,In vitro growth curves of LM2–4175 and MFM-223 cells stably expressing shNC or shACAA1 vectors. The data are presented as the mean ± SD; n = 3 independent experiments. Two-way ANOVA was used. ***, P < 0.001. F, Representative images of surviving colonies of LM2–4175 and MDA-MB-453 cells stably expressing shNC or two shACAA1 vectors are shown. n = 3 independent experiments. P values were determined using a two-tailed unpaired Student t test. ***, P < 0.001. G, Flow cytometry analysis showing the cell-cycle distribution of LM2–4175 and MFM-223 cells expressing shNC or shACAA1 expression vectors. The data are presented as the mean ± SD; n = 3 independent experiments. P values were determined using a two-tailed unpaired Student t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001. H, MDA-MB-231 and BT-549 cells stably expressing vector or Flag-ACAA1 were immunoblotted with the indicated antibodies. I, LM2–4175 cells expressing shNC or shACAA1 vectors were immunoblotted with the indicated antibodies. J, MFM-223 and HCC1143 cells were treated with increasing doses of trimetazidine (TMZ) for 48 hours and analyzed using immunoblotting with the indicated antibodies. K, MFM-223 and HCC1143 cells were treated with increasing doses of trimetazidine for 24 hours and subjected to qRT-PCR analysis. Data are presented as the mean ± SD; n = 3 independent experiments. P values were determined using a two-tailed unpaired Student t test. ns, not significant.

Close modal

Given the similarity between ACAA1 and ACAA2, we next investigated the effects of trimetazidine, an approved ACAA2 inhibitor, on TNBC cell lines (28). We detected the IC50 values of trimetazidine in 12 TNBC cell lines and observed that several TNBC cell lines were vulnerable to trimetazidine (Supplementary Fig. S3A). However, no significant correlation was observed between the ACAA1 protein level and sensitivity to trimetazidine (Supplementary Fig. S3B).

To explore the role of trimetazidine on ACAA1, we performed qRT-PCR and immunoblotting analyses in ACAA1 high-expressing MFM-223 and HCC1143 cells treated with trimetazidine. Interestingly, we observed a dose-dependent decrease in the protein level but not in the mRNA expression of ACAA1 in trimetazidine-treated cells (Fig. 2J and K; Supplementary Fig. S4A). ACAA1 high-expressing MFM-223 and HCC70 cells were treated with the proteasome inhibitor MG-132 to determine the degradation pathway of ACAA1. ACAA1 did not accumulate, but a significant increase in p21, a known substrate of the ubiquitin–proteasome system (29), was detected (Supplementary Fig. S4B). Meanwhile, the lysosomal inhibitors Baf-A1 and NH4Cl resulted in the accumulation of ACAA1 in MFM-223 cells (Supplementary Fig. S4C and S4D), suggesting that ACAA1 was degraded in the lysosomes. Three types of autophagy are involved in lysosomal degradation: macroautophagy, microautophagy, and chaperone-mediated autophagy (CMA; ref. 30). CMA may be induced by serum starvation (31). Further studies showed that serum starvation and siRNA-mediated knockdown of two key molecules involved in CMA, HSP family A (HSP70) member 8 (HSPA8) and lysosome-associated membrane protein type 2a (LAMP2A), had no significant effects on ACAA1 protein levels, indicating that CMA was not responsible for ACAA1 protein degradation (Supplementary Fig. S4E–G). Furthermore, treatment with 1 mmol/L 3-MA, a selective inhibitor of macroautophagy (32), resulted in a significant increase in ACAA1 protein levels in MFM-223 and HCC70 cells, indicating that macroautophagy was responsible for ACAA1 protein degradation (Supplementary Fig. S4H).

We further explored whether trimetazidine could inhibit tumor growth through regulation of ACAA2. First, we explored the role of ACAA2 in TNBC and evaluated the effect of trimetazidine on ACAA2. The siRNA-mediated knockdown of ACAA2 had no significant effect on the growth of Hs-578T cells with high ACAA2 expression (Supplementary Fig. S5A–S5C). In addition, no decreases in ACAA2 mRNA expression or protein levels in trimetazidine-treated Hs-578T, MFM-223, or HCC1143 cells were observed (Supplementary Fig. S5D and E). In summary, ACAA2 had no effect on TNBC cell growth, and trimetazidine had no effect on the protein level or mRNA expression of ACAA2.

Overall, our findings suggest that ACAA1 promotes TNBC progression through the G1–S cell-cycle transition and is a potential therapeutic target for TNBC, because treatment with trimetazidine causes ACAA1 protein degradation and inhibits TNBC cell proliferation.

ACAA1 interacted with CDK4 in TNBC

We performed stable isotope labeling by amino acids in cell culture (SILAC)-based quantitative proteomics to identify the ACAA1-associated proteins and uncover the underlying molecular mechanisms of ACAA1 in TNBC (Fig. 3A). After excluding three HSPs that are well-known regulators of the stress response (33), four potential ACAA1-associated proteins (PEX7, FKBP10, CDK4, and ARHGAP21) were identified on the basis of the unique peptide numbers, log2 (ratio H/L) and “significance B” values (Fig. 3B). We focused on CDK4 as the key ACAA1-associated protein because its role in cell-cycle progression is similar to that of ACAA1 in TNBC (34). We verified the interaction between ACAA1 and CDK4 through IP assays in HEK293T, BT-549, HCC70, and HCC38 cells but observed no interaction between ACAA1 and D-cyclins (cyclin D1, D2, and D3; Fig. 3C and D; Supplementary Fig. S6A). Immunofluorescence (IF) staining showed that CDK4 localized primarily in the nucleus, while ACAA1 localized both in the cytoplasm and in the nucleus (Fig. 3E).

Figure 3.

ACAA1 interacted with CDK4 in TNBC. A, Flowchart of SILAC combined with LC/MS-MS for the comparative analysis of protein expression in BT-549 cells overexpressing ACAA1. B, The top five proteins with accurate quantification and significance B. C, HEK293T and BT-549 cells stably expressing an empty vector or vector expressing Flag-tagged ACAA1 (Flag-ACAA1) were subjected to IP and immunoblotting analyses with the indicated antibodies. D, HCC70 and HCC38 cell lysates were subjected to IP assays with control IgG and an ACAA1 antibody, followed by immunoblot analysis with the indicated antibodies. E, IF staining for ACAA1 (green) and CDK4 (red) in LM2–4175, MFM-223, HCC38, and HCC1143 cells. Scale bar, 10 μm. F, Schematic diagram of HA-tagged full-length (FL) human CDK4 and its truncated mutants. G, Schematic diagram of Flag-tagged full-length human ACAA1 and its truncated mutants. H, HEK293T cells were cotransfected with HA-tagged full-length CDK4 or one of its truncated mutants (Δ1 to Δ5), as well as Flag-ACAA1. After 48 hours of transfection, cell lysates were immunoprecipitated with beads coated with a Flag antibody. Specific interactions were analyzed by immunoblotting with the indicated antibodies. I, Flag-tagged full-length ACAA1 or one of its truncated mutants (Δ1 to Δ4) were cotransfected with HA-CDK4 in HEK293T cells. After 48 hours, extracts were immunoprecipitated with beads coated with a Flag antibody. Specific interactions were analyzed using immunoblotting with the indicated antibodies.

Figure 3.

ACAA1 interacted with CDK4 in TNBC. A, Flowchart of SILAC combined with LC/MS-MS for the comparative analysis of protein expression in BT-549 cells overexpressing ACAA1. B, The top five proteins with accurate quantification and significance B. C, HEK293T and BT-549 cells stably expressing an empty vector or vector expressing Flag-tagged ACAA1 (Flag-ACAA1) were subjected to IP and immunoblotting analyses with the indicated antibodies. D, HCC70 and HCC38 cell lysates were subjected to IP assays with control IgG and an ACAA1 antibody, followed by immunoblot analysis with the indicated antibodies. E, IF staining for ACAA1 (green) and CDK4 (red) in LM2–4175, MFM-223, HCC38, and HCC1143 cells. Scale bar, 10 μm. F, Schematic diagram of HA-tagged full-length (FL) human CDK4 and its truncated mutants. G, Schematic diagram of Flag-tagged full-length human ACAA1 and its truncated mutants. H, HEK293T cells were cotransfected with HA-tagged full-length CDK4 or one of its truncated mutants (Δ1 to Δ5), as well as Flag-ACAA1. After 48 hours of transfection, cell lysates were immunoprecipitated with beads coated with a Flag antibody. Specific interactions were analyzed by immunoblotting with the indicated antibodies. I, Flag-tagged full-length ACAA1 or one of its truncated mutants (Δ1 to Δ4) were cotransfected with HA-CDK4 in HEK293T cells. After 48 hours, extracts were immunoprecipitated with beads coated with a Flag antibody. Specific interactions were analyzed using immunoblotting with the indicated antibodies.

Close modal

We constructed five truncated mutations of CDK4 and four truncated mutations of ACAA1 to identify the region of each protein responsible for this interaction (Fig. 3F and G). Next, we performed coimmunoprecipitation assays in HEK293T cells and showed that deleting the domain (residues 161–245) following the Asp158-Phe159-Gly160 (Deutsche Forschungsgemeinschaft; DFG) motif in CDK4 abolished the binding between CDK4 and ACAA1 (Fig. 3H). This domain contains both the T-loop region (residues 161–171) and CDK4 phosphorylation site (Thr172), which is critical to the function of CDK4 (35). In addition, we identified the N-terminal thiolase domain (residues 38–161) of ACAA1 to be critical in its interaction with CDK4 (Fig. 3I). However, further immunoblotting analyses showed that overexpression or downregulation of ACAA1 did not alter CDK4 protein expression, phosphorylation (Thr172) or nucleoplasmic distribution (Supplementary Fig. S6B–D). Likewise, overexpression of CDK4 did not affect ACAA1 protein expression (Supplementary Fig. S6E). Altogether, these data suggest that ACAA1 binds to CDK4 but has no effect on its protein expression, phosphorylation status, or nucleoplasmic distribution.

Downregulation of ACAA1 facilitated the efficacy of the CDK4/6 inhibitor abemaciclib in TNBC

We detected the IC50 values of abemaciclib and analyzed the protein expression pattern by immunoblotting using a panel of 13 TNBC cell lines with different ACAA1 protein levels and RB1 statuses [wild-type (WT), deleted, or mutant] to determine the sensitivity to abemaciclib (Fig. 4A and B). We observed heterogeneous sensitivities to abemaciclib, with IC50 values ranging from 0.157 μmol/L (Hs-578T) to 11.658 μmol/L (HCC1937; Fig. 4A). Consistent with a previous study, we observed that cells with deleted or mutant RB1 (the group shown in black) were relatively resistant to abemaciclib (36). Among the WT cell lines, cells with lower ACAA1 protein levels (ACAA1low, the blue group) had lower IC50 values than those with higher ACAA1 protein levels (ACAA1high, the red group). These data suggested a possible correlation between ACAA1 expression and sensitivity to the CDK4/6 inhibitor abemaciclib.

Figure 4.

Downregulation of ACAA1 facilitated the efficacy of abemaciclib in vitro and in vivo in TNBC. A, Cell viability of a panel of 13 TNBC cell lines with varying endogenous RB1 gene statuses treated with abemaciclib. The IC50 values are shown. Data are presented as the mean ± SD; n = 3 independent experiments. The P value was determined using an unpaired two-tailed Student t test. B, Cell lysates of a panel of 13 TNBC cell lines with varying endogenous RB1 status were immunoblotted with the indicated antibodies. Mut, mutant; Del, deleted. C, LM2–4175 cells stably expressing shNC or shACAA1 expression vectors were treated with increasing concentrations of abemaciclib for 72 hours. n = 3 independent experiments. D, LM2–4175 cells stably expressing shNC or shACAA1 expression vectors were treated with increasing concentrations of abemaciclib for 24 hours. Cell lysates were immunoblotted with the indicated antibodies. E–G, LM2–4175 cells (8 × 105) stably expressing shNC or shACAA1 expression vectors were injected into the fourth mammary fat pad of 6-week-old female NOD/SCID mice to construct an orthotopic xenograft model. After 19 days of injection, the tumors reached approximately 50 mm3, and the mice were randomly assigned to two treatment groups: the abemaciclib (Abema) and vehicle groups. Abemaciclib and PBS were administered by oral gavage once daily for 11 consecutive days. Mice were sacrificed on day 30 post injection, and tumors were excised. Representative tumor images (E), endpoint tumor weight (F), and tumor growth curves (G) are shown and compared among the groups. Data are presented as the mean ± SD (n = 8 per group). H, Two TNBC patient-derived organoids were treated with 0.5 μmol/L abemaciclib or vehicle. Representative brightfield images of organoids after drug treatment and the 3D cell viability assay results are shown. Scale bars, 200 μm. Data are presented as the mean ± SD; n = 3 biological replicates. I, Hematoxylin and eosin (H&E) staining and IHC staining for ACAA1, RB1, and p-RB (Ser780) in the two TNBC patient-derived organoid models are shown. Scale bars, 100 μm. The inset shows an image detail. P values were determined using a two-tailed unpaired Student t test. **, P < 0.01; ***, P < 0.001.

Figure 4.

Downregulation of ACAA1 facilitated the efficacy of abemaciclib in vitro and in vivo in TNBC. A, Cell viability of a panel of 13 TNBC cell lines with varying endogenous RB1 gene statuses treated with abemaciclib. The IC50 values are shown. Data are presented as the mean ± SD; n = 3 independent experiments. The P value was determined using an unpaired two-tailed Student t test. B, Cell lysates of a panel of 13 TNBC cell lines with varying endogenous RB1 status were immunoblotted with the indicated antibodies. Mut, mutant; Del, deleted. C, LM2–4175 cells stably expressing shNC or shACAA1 expression vectors were treated with increasing concentrations of abemaciclib for 72 hours. n = 3 independent experiments. D, LM2–4175 cells stably expressing shNC or shACAA1 expression vectors were treated with increasing concentrations of abemaciclib for 24 hours. Cell lysates were immunoblotted with the indicated antibodies. E–G, LM2–4175 cells (8 × 105) stably expressing shNC or shACAA1 expression vectors were injected into the fourth mammary fat pad of 6-week-old female NOD/SCID mice to construct an orthotopic xenograft model. After 19 days of injection, the tumors reached approximately 50 mm3, and the mice were randomly assigned to two treatment groups: the abemaciclib (Abema) and vehicle groups. Abemaciclib and PBS were administered by oral gavage once daily for 11 consecutive days. Mice were sacrificed on day 30 post injection, and tumors were excised. Representative tumor images (E), endpoint tumor weight (F), and tumor growth curves (G) are shown and compared among the groups. Data are presented as the mean ± SD (n = 8 per group). H, Two TNBC patient-derived organoids were treated with 0.5 μmol/L abemaciclib or vehicle. Representative brightfield images of organoids after drug treatment and the 3D cell viability assay results are shown. Scale bars, 200 μm. Data are presented as the mean ± SD; n = 3 biological replicates. I, Hematoxylin and eosin (H&E) staining and IHC staining for ACAA1, RB1, and p-RB (Ser780) in the two TNBC patient-derived organoid models are shown. Scale bars, 100 μm. The inset shows an image detail. P values were determined using a two-tailed unpaired Student t test. **, P < 0.01; ***, P < 0.001.

Close modal

We performed growth inhibition assays using LM2–4175 and HCC1143 cells and found that downregulation of ACAA1 sensitized RB1-WT TNBC cells to abemaciclib (Fig. 4C; Supplementary Fig. S7A). Consistently, a substantially lower dose of abemaciclib suppressed RB1 phosphorylation and E2F-targeted protein (PLK1 and cyclin A2) levels in ACAA1 knockdown LM2–4175 and HCC1143 cells (Fig. 4D; Supplementary Fig. S7B and C). However, the potentiating effect was not observed in RB1-mutant HCC70 cell lines (Supplementary Fig. S7D and S7E). We used the mammary fat pad injection model in female NOD/SCID mice to investigate the effects of ACAA1 on TNBC proliferation and the response to abemaciclib in vivo. In accordance with the in vitro results, downregulation of ACAA1 inhibited xenograft tumor growth, and the tumors were more sensitive to abemaciclib (Fig. 4EG). We observed a 24.08% decrease in tumor weight after treatment with abemaciclib and a 51.75% decrease in tumor weight after abemaciclib treatment combined with ACAA1 knockdown (Fig. 4F). Likewise, we observed a 55.83% decrease in the tumor volume and a 43.21% decrease in the tumor volume following abemaciclib treatment with or without downregulation of ACAA1 (Fig. 4G). We also observed that treatment with abemaciclib delayed tumor growth for 6 days in LM2–4175 xenograft models and extended to 15 days when abemaciclib was combined with ACAA1 knockdown (Supplementary Fig. S8A). Treatment toxicity evaluated by body weight, white blood cell (WBC) counts, hemoglobin levels, and platelet counts was limited (Supplementary Fig. S8B–S8E). We subsequently performed 3D cell viability assays using two TNBC patient-derived organoid models treated with or without 0.5 μmol/L abemaciclib. The transcriptomic subtype of those two patients was defined as the basal-like and immune-suppressed (BLIS) subtype for PDO811 and the LAR subtype for PDO845 on the basis of our previously defined IHC method (37). We observed that the organoid (PDO811) derived from the patient with weaker ACAA1 and RB1 phosphorylation (Ser780) IHC staining was more sensitive to abemaciclib (Fig. 4H and I). Overall, our data reveal that ACAA1 may be used to stratify RB1-proficient TNBC for CDK4/6 inhibitor treatment and that inhibition of ACAA1 potentiates the tumor response to the CDK4/6 inhibitor abemaciclib.

Trimetazidine potentiated the antitumor efficacy of the CDK4/6 inhibitor abemaciclib in vitro and in vivo

As downregulation of ACAA1 potentiates the response to the CDK4/6 inhibitor abemaciclib and ACAA1 protein levels are decreased by trimetazidine, we performed drug combination assays and calculated the CI values using the Chou–Talalay method (38). As expected, the combination of abemaciclib with trimetazidine exhibited significant synergy in three RB1 WT TNBC cell lines: MFM-223 (CI = 0.48 ± 0.21), LM2–4175 (CI = 0.52 ± 0.21), and HCC1143 (CI = 0.29 ± 0.21; Fig. 5A; Supplementary Fig. S9A and S9B). The growth rate of MFM-223, LM2–4175, and HCC1143 cells was slightly or moderately inhibited using a single agent (apart from 1 mmol/L trimetazidine in MFM-223 cells) but was further inhibited by the combination of abemaciclib and trimetazidine (Fig. 5B; Supplementary Fig. S9C–D). Notably, a synergistic effect was observed even on RB1-deficient HCC1937 cells (Supplementary Fig. S9E). Moreover, immunoblot analysis showed that RB1 and PLK1 protein levels and RB1 phosphorylation (Ser780) levels decreased significantly following treatment with trimetazidine and abemaciclib, indicating that combined treatment further enhanced cell-cycle arrest compared with single-agent treatment (Fig. 5C; Supplementary Fig. S9F). Next, we evaluated the efficacy and toxicity of trimetazidine and abemaciclib alone or in combination in vivo. The combination of abemaciclib and trimetazidine induced greater tumor regression in LM2–4175 xenograft models than abemaciclib or trimetazidine alone (Fig. 5DF). We observed 31.46%, 35.93%, and 62.13% decreases in tumor weight following treatment with abemaciclib, trimetazidine, and their combination, respectively (Fig. 5E). Likewise, we observed 43.25%, 45.75%, and 74.26% decreases in tumor volumes after treatment with abemaciclib, trimetazidine and their combination, respectively (Fig. 5F). Importantly, no significant body weight loss was observed in the combination group compared with the single-agent groups (Fig. 5G). We also observed that treatment with abemaciclib or trimetazidine alone delayed tumor growth for 6 days, that extended to 15 days when these drugs were administered in combination (Supplementary Fig. S10A). Toxicity evaluated by body weight, WBC counts, hemoglobin levels, platelet counts, and hepatorenal indicators was limited (Supplementary Fig. S10B–S10H). A significant decrease in phosphorylated RB1 (Ser780) was observed in the IHC staining of xenograft tumors from mice that underwent combined treatment (Fig. 5H; Supplementary Fig. S11A). Likewise, significantly weaker IHC staining for ACAA1 was observed after in vivo trimetazidine treatment (Fig. 5I; Supplementary Fig. S11B). We performed RNA-seq and gene set enrichment analysis (GSEA) on xenograft tumors to investigate the influence of trimetazidine in vivo. Consistent with the in vitro results, no significant decrease in ACAA1 mRNA expression was observed (Supplementary Fig. S12A and S12B). GSEA revealed downregulation of cell-cycle–related gene sets (such as the mitotic G1 phase, G1–S transition, and E2F targets gene sets) after in vivo trimetazidine treatment (Supplementary Fig. S12C and S12D). Finally, we verified the synergy of 1 μmol/L abemaciclib and 1 mmol/L trimetazidine in an organoid model derived from one patient with TNBC (Fig. 5J). Altogether, these findings reveal that trimetazidine downregulates ACAA1 protein levels and the G1–S cell-cycle transition in vivo, supporting trimetazidine combined with abemaciclib as a potential strategy for TNBC with high ACAA1 expression levels.

Figure 5.

Effect of trimetazidine on potentiating the antitumor efficacy of abemaciclib in TNBC in vitro and in vivo. A, The cell viability inhibition rate and CI value of abemaciclib (Abema) and trimetazidine (TMZ) in MFM-223 cells at different drug concentrations are shown. Abemaciclib (μmol/L) was diluted 2/3 times from left to right, and trimetazidine (mmol/L) was diluted 1/2 times from top to bottom. B,In vitro growth curves of abemaciclib (2 μmol/L) and trimetazidine (1 mmol/L) alone or in combination in MFM-223 cells. The data are presented as the mean ± SD; n = 3 independent experiments. C, MFM-223 cells were treated with 1 μmol/L abemaciclib and/or 1 mmol/L trimetazidine for 12 hours. Cell lysates were subjected to immunoblotting with the indicated antibodies. D–G, Representative tumor images (D), quantification of tumor weight (E), tumor volume (F), and relative body weight (G) in LM2–4175 xenograft mice following treatment with abemaciclib (25 mg/kg; n = 8) and trimetazidine (250 mg/kg; n = 8), individually or in combination (n = 8), or vehicle (n = 8) are shown. Abemaciclib was administered by oral gavage and trimetazidine was administered via intraperitoneal injection once daily (arrow, first and last administration of treatment). H, Hematoxylin and eosin (H&E) staining and IHC staining for p-RB (Ser780) in the xenograft tumors. The H-score is shown. Scale bars, 100 μm. The data are presented as the mean ± SD; n = 8 biological replicates. I, Hematoxylin and eosin staining and IHC staining for ACAA1 in xenograft tumors treated with vehicle or trimetazidine. The H-score is shown. Scale bars, 100 μm. The data are presented as the means ± SD; n = 8 biological replicates. J, One TNBC patient-derived organoid was treated with 1 μmol/L abemaciclib and/or 1 mmol/L trimetazidine. Representative brightfield images of organoids after drug treatment and the results of the 3D cell viability assay are shown. Scale bars, 200 μm. Data are presented as the mean ± SD; n = 3 biological replicates. P values were determined using a two-tailed unpaired Student t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant.

Figure 5.

Effect of trimetazidine on potentiating the antitumor efficacy of abemaciclib in TNBC in vitro and in vivo. A, The cell viability inhibition rate and CI value of abemaciclib (Abema) and trimetazidine (TMZ) in MFM-223 cells at different drug concentrations are shown. Abemaciclib (μmol/L) was diluted 2/3 times from left to right, and trimetazidine (mmol/L) was diluted 1/2 times from top to bottom. B,In vitro growth curves of abemaciclib (2 μmol/L) and trimetazidine (1 mmol/L) alone or in combination in MFM-223 cells. The data are presented as the mean ± SD; n = 3 independent experiments. C, MFM-223 cells were treated with 1 μmol/L abemaciclib and/or 1 mmol/L trimetazidine for 12 hours. Cell lysates were subjected to immunoblotting with the indicated antibodies. D–G, Representative tumor images (D), quantification of tumor weight (E), tumor volume (F), and relative body weight (G) in LM2–4175 xenograft mice following treatment with abemaciclib (25 mg/kg; n = 8) and trimetazidine (250 mg/kg; n = 8), individually or in combination (n = 8), or vehicle (n = 8) are shown. Abemaciclib was administered by oral gavage and trimetazidine was administered via intraperitoneal injection once daily (arrow, first and last administration of treatment). H, Hematoxylin and eosin (H&E) staining and IHC staining for p-RB (Ser780) in the xenograft tumors. The H-score is shown. Scale bars, 100 μm. The data are presented as the mean ± SD; n = 8 biological replicates. I, Hematoxylin and eosin staining and IHC staining for ACAA1 in xenograft tumors treated with vehicle or trimetazidine. The H-score is shown. Scale bars, 100 μm. The data are presented as the means ± SD; n = 8 biological replicates. J, One TNBC patient-derived organoid was treated with 1 μmol/L abemaciclib and/or 1 mmol/L trimetazidine. Representative brightfield images of organoids after drug treatment and the results of the 3D cell viability assay are shown. Scale bars, 200 μm. Data are presented as the mean ± SD; n = 3 biological replicates. P values were determined using a two-tailed unpaired Student t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant.

Close modal

In this study, we screened ACAA1 on the basis of our proteomic and transcriptomic data and identified that ACAA1 inhibition restrained tumor proliferation and facilitated the response to the CDK4/6 inhibitor abemaciclib in TNBC. Further study indicated that trimetazidine downregulates ACAA1 protein levels and gene sets of G1–S transition and downstream E2F targets. Mechanistic investigations demonstrated the role of ACAA1 in regulating the G1–S cell-cycle transition and the interaction between ACAA1 and CDK4 in the nucleus (Fig. 6). Importantly, we revealed that trimetazidine potentiated the antitumor efficacy of abemaciclib in TNBC cell lines, patient-derived organoids, and xenograft models.

Figure 6.

Schematic of ACAA1 proliferation regulation and the response to CDK4/6 inhibitors through interaction with CDK4 in TNBC.

Figure 6.

Schematic of ACAA1 proliferation regulation and the response to CDK4/6 inhibitors through interaction with CDK4 in TNBC.

Close modal

The LAR subtype remains a challenge in TNBC with a relatively poor prognosis, as evidenced by the low pathologic complete response rates after neoadjuvant chemotherapy and low objective response rates in metastatic TNBC after targeted therapy, including an AR inhibitor (13, 39). The LAR subtype and proteomic iP-2 subtype overlap significantly, with 60% of the iP-2 subtype clustered in the FUSCC subtype LAR (18). We demonstrated that ACAA1 was expressed relatively higher in both the LAR subtype and proteomic iP-2 subtype and confirmed its association with poor survival in TNBCs. ACAA1 encodes human peroxisomal 3-oxoacyl-CoA thiolase, an enzyme responsible for the thiolytic cleavage of straight-chain 3-oxoacyl-CoAs and the generation of acetyl-CoA and acyl-CoA in the peroxisomal beta-oxidation system (40). ACAA1 is a key member of the PPAR and fatty acid metabolism pathways, and its expression is upregulated or downregulated in various tumor tissues compared with the corresponding normal tissues (41–44). A previous study showed that five ACAA1 SNPs are associated with breast cancer susceptibility (45).

Three pharmacologic inhibitors of CDK4/6 (palbociclib, ribociclib, and abemaciclib) have been widely implemented in clinical practice as treatments for hormone receptor–positive and HER2-negative advanced or metastatic breast cancer (46). As a well-known tumor suppressor, RB1 plays an important role in restricting cell-cycle progression by regulating E2F transcription factors, and RB1 aberrations have been reported as mechanisms of preexisting and acquired resistance to CDK4/6 inhibitors (47–50). Comprehensive profiles of breast cancer revealed that a high frequency of RB1 loss and mutations were detected in a considerable proportion of TNBC tissues (12, 14). In this study, we observed total and phosphorylated blockages of RB1 following ACAA1 downregulation and discovered an interaction between ACAA1 and CDK4 in TNBC. CDK4 is a key regulator of the G1–S cell-cycle transition and is crucial in the growth and development of many cancer types, including breast cancer (51). Many studies have documented the synergistic effects of combining CDK4/6 inhibitors with other targeted therapies (such as inhibitors of PARP, AR, BET, EGFR, and PI3Kα) in certain TNBCs (52–57). However, the translation of these preclinical studies into clinical benefits is an ongoing challenge.

Trimetazidine, an anti-ischemic agent, reduces long-chain fatty acid oxidation by inhibiting the 3-ketoacyl-coenzyme A thiolase enzyme (58). Previous studies reported that the combination of trimetazidine with other drugs, such as the PPARγ agonist troglitazone, the glutaminase C inhibitor C.968, and chemotherapy, induces apoptosis in cancer cells (59, 60). In this study, we utilized trimetazidine’s ability to reduce ACAA1 protein levels to target ACAA1. We observed a similar role of trimetazidine to CDK4/6 inhibitors and discovered its effect on potentiating the efficacy of abemaciclib in RB1-proficient TNBC models. We demonstrate that ACAA1 regulates TNBC proliferation via the CDK4/6-RB1-E2F signaling pathway and that inhibiting ACAA1 with trimetazidine enhances the response to the CDK4/6 inhibitor abemaciclib. However, we speculate that other mechanisms beyond the CDK4-RB1-E2F pathway may also be involved, and further studies are still required to investigate the role of trimetazidine in treating breast cancer.

Several limitations of this study need to be noted. First, ACAA1 was detected in the nucleus of TNBC cell lines. It lacks the canonical nuclear localization signal sequences, and the mechanisms underlying its nuclear localization and potential effects remain unclear, warranting further investigation. Second, the dosage of trimetazidine used in our study was higher than the clinical dose (20 mg, orally, three times a day) used to treat heart-related conditions. Finally, our study lacks paired biopsies and detection of ACAA1 in patients with TNBC before and after treatment with CDK4/6 inhibitors, as CDK4/6 inhibitors are not currently used to treat patients with TNBC. Hence, a well-designed clinical trial enrolling patients with TNBC with heterogeneous ACAA1 expression status is required to verify our conclusions in the clinic.

In conclusion, our data suggest that the combination of trimetazidine with abemaciclib may be a promising treatment strategy for ACAA1-high TNBCs. Furthermore, our study highlights the importance of investigating ACAA1 protein levels in further clinical trials involving CDK4/6 inhibitors in TNBC.

No disclosures were reported.

W.T. Peng: Conceptualization, resources, data curation, software, formal analysis, investigation, visualization, methodology, writing–original draft, writing–review and editing. X. Jin: Conceptualization, resources, data curation, software, formal analysis, funding acquisition, visualization, methodology, writing–original draft, writing–review and editing. X.E. Xu: Data curation, methodology, project administration, writing–review and editing. Y.S. Yang: Software, investigation, methodology, writing–review and editing. D. Ma: Data curation, formal analysis, methodology, writing–review and editing. Z.M. Shao: Conceptualization, resources, supervision, funding acquisition, validation, investigation, project administration, writing–review and editing. Y.Z. Jiang: Conceptualization, data curation, supervision, funding acquisition, investigation, methodology, project administration, writing–review and editing.

This work was supported by grants from the National Key Research and Development Project of China (2020YFA0112304 to Z.-M. Shao), the National Natural Science Foundation of China (91959207 and 92159301 to Z.-M. Shao), the Program of Shanghai Academic/Technology Research Leader (20XD1421100 to Y.-Z. Jiang), the Natural Science Foundation of Shanghai (22ZR1479200 to Y.-Z. Jiang), the Shanghai Key Laboratory of Breast Cancer (12DZ2260100 to Z.-M. Shao), Youth Talent Program of Shanghai Health Commission (2022YQ012 to X. Jin), the SHDC Municipal Project for Developing Emerging and Frontier Technology in Shanghai Hospitals (SHDC12021103 to Z.-M. Shao), and the Clinical Research Plan of SHDC (SHDC2020CR4002 to Y.-Z. Jiang; SHDC2020CR5005 to Z.-M. Shao).

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

1.
Dent
R
,
Trudeau
M
,
Pritchard
KI
,
Hanna
WM
,
Kahn
HK
,
Sawka
CA
, et al
.
Triple-negative breast cancer: clinical features and patterns of recurrence
.
Clin Cancer Res
2007
;
13
:
4429
34
.
2.
Foulkes
WD
,
Smith
IE
,
Reis-Filho
JS
.
Triple-negative breast cancer
.
N Engl J Med
2010
;
363
:
1938
48
.
3.
Bianchini
G
,
De Angelis
C
,
Licata
L
,
Gianni
L
.
Treatment landscape of triple-negative breast cancer - expanded options, evolving needs
.
Nat Rev Clin Oncol
2022
;
19
:
91
113
.
4.
Robson
M
,
Im
SA
,
Senkus
E
,
Xu
B
,
Domchek
SM
,
Masuda
N
, et al
.
Olaparib for metastatic breast cancer in patients with a germline BRCA mutation
.
N Engl J Med
2017
;
377
:
523
33
.
5.
Litton
JK
,
Rugo
HS
,
Ettl
J
,
Hurvitz
SA
,
Goncalves
A
,
Lee
KH
, et al
.
Talazoparib in patients with advanced breast cancer and a germline BRCA mutation
.
N Engl J Med
2018
;
379
:
753
63
.
6.
Tutt
ANJ
,
Garber
JE
,
Kaufman
B
,
Viale
G
,
Fumagalli
D
,
Rastogi
P
, et al
.
Adjuvant olaparib for patients with BRCA1- or BRCA2-mutated breast cancer
.
N Engl J Med
2021
;
384
:
2394
405
.
7.
Schmid
P
,
Cortes
J
,
Pusztai
L
,
McArthur
H
,
Kummel
S
,
Bergh
J
, et al
.
Pembrolizumab for early triple-negative breast cancer
.
N Engl J Med
2020
;
382
:
810
21
.
8.
Cortes
J
,
Cescon
DW
,
Rugo
HS
,
Nowecki
Z
,
Im
S-A
,
Yusof
MM
, et al
.
Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial
.
Lancet
2020
;
396
:
1817
28
.
9.
Schmid
P
,
Adams
S
,
Rugo
HS
,
Schneeweiss
A
,
Barrios
CH
,
Iwata
H
, et al
.
Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer
.
N Engl J Med
2018
;
379
:
2108
21
.
10.
Bardia
A
,
Mayer
IA
,
Vahdat
LT
,
Tolaney
SM
,
Isakoff
SJ
,
Diamond
JR
, et al
.
Sacituzumab govitecan-hziy in refractory metastatic triple-negative breast cancer
.
N Engl J Med
2019
;
380
:
741
51
.
11.
Modi
S
,
Jacot
W
,
Yamashita
T
,
Sohn
J
,
Vidal
M
,
Tokunaga
E
, et al
.
Trastuzumab deruxtecan in previously treated HER2-low advanced breast cancer
.
N Engl J Med
2022
;
387
:
9
20
.
12.
Jiang
YZ
,
Ma
D
,
Suo
C
,
Shi
J
,
Xue
M
,
Hu
X
, et al
.
Genomic and transcriptomic landscape of triple-negative breast cancers: subtypes and treatment strategies
.
Cancer Cell
2019
;
35
:
428
40
.
13.
Jiang
YZ
,
Liu
Y
,
Xiao
Y
,
Hu
X
,
Jiang
L
,
Zuo
WJ
, et al
.
Molecular subtyping and genomic profiling expand precision medicine in refractory metastatic triple-negative breast cancer: the FUTURE trial
.
Cell Res
2021
;
31
:
178
86
.
14.
Cancer Genome Atlas N
.
Comprehensive molecular portraits of human breast tumours
.
Nature
2012
;
490
:
61
70
.
15.
Hollestelle
A
,
Nagel
JHA
,
Smid
M
,
Lam
S
,
Elstrodt
F
,
Wasielewski
M
, et al
.
Distinct gene mutation profiles among luminal-type and basal-type breast cancer cell lines
.
Breast Cancer Res Tr
2010
;
121
:
53
64
.
16.
Asghar
US
,
Barr
AR
,
Cutts
R
,
Beaney
M
,
Babina
I
,
Sampath
D
, et al
.
Single-cell dynamics determines response to CDK4/6 inhibition in triple-negative breast cancer
.
Clin Cancer Res
2017
;
23
:
5561
72
.
17.
Hu
Y
,
Gao
J
,
Wang
M
,
Li
M
.
Potential prospect of CDK4/6 inhibitors in triple-negative breast cancer
.
Cancer Manag Res
2021
;
13
:
5223
37
.
18.
Gong
T-Q
,
Jiang
Y-Z
,
Shao
C
,
Peng
W-T
,
Liu
M-W
,
Li
D-Q
, et al
.
Proteome-centric cross-omics characterization and integrated network analyses of triple-negative breast cancer
.
Cell Rep
2022
;
38
:
110460
.
19.
Ciriello
G
,
Gatza
ML
,
Beck
AH
,
Wilkerson
MD
,
Rhie
SK
,
Pastore
A
, et al
.
Comprehensive molecular portraits of invasive lobular breast cancer
.
Cell
2015
;
163
:
506
19
.
20.
Gyorffy
B
.
Survival analysis across the entire transcriptome identifies biomarkers with the highest prognostic power in breast cancer
.
Comput Struct Biotechnol J
2021
;
19
:
4101
9
.
21.
Tang
W
,
Zhou
M
,
Dorsey
TH
,
Prieto
DA
,
Wang
XW
,
Ruppin
E
, et al
.
Integrated proteotranscriptomics of breast cancer reveals globally increased protein-mRNA concordance associated with subtypes and survival
.
Genome Med
2018
;
10
:
94
.
22.
Sachs
N
,
de Ligt
J
,
Kopper
O
,
Gogola
E
,
Bounova
G
,
Weeber
F
, et al
.
A living biobank of breast cancer organoids captures disease heterogeneity
.
Cell
2018
;
172
:
373
86
.
23.
Chou
TC
.
Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies
.
Pharmacol Rev
2006
;
58
:
621
81
.
24.
Zhang
H
,
Zhang
N
,
Liu
Y
,
Su
P
,
Liang
Y
,
Li
Y
, et al
.
Epigenetic regulation of NAMPT by NAMPT-AS drives metastatic progression in triple-negative breast cancer
.
Cancer Res
2019
;
79
:
3347
59
.
25.
Ding
X
,
Fan
K
,
Hu
J
,
Zang
Z
,
Zhang
S
,
Zhang
Y
, et al
.
SCP2-mediated cholesterol membrane trafficking promotes the growth of pituitary adenomas via Hedgehog signaling activation
.
J Exp Clin Cancer Res
2019
;
38
:
404
.
26.
Wishart
DS
,
Knox
C
,
Guo
AC
,
Shrivastava
S
,
Hassanali
M
,
Stothard
P
, et al
.
DrugBank: a comprehensive resource for in silico drug discovery and exploration
.
Nucleic Acids Res
2006
;
34
:
D668
72
.
27.
Dyson
NJ
.
RB1: a prototype tumor suppressor and an enigma
.
Genes Dev
2016
;
30
:
1492
502
.
28.
Kantor
PF
,
Lucien
A
,
Kozak
R
,
Lopaschuk
GD
.
The antianginal drug trimetazidine shifts cardiac energy metabolism from fatty acid oxidation to glucose oxidation by inhibiting mitochondrial long-chain 3-ketoacyl coenzyme A thiolase
.
Circ Res
2000
;
86
:
580
8
.
29.
Zhu
Q
,
Wani
G
,
Yao
J
,
Patnaik
S
,
Wang
Q-E
,
El-Mahdy
M
, et al
.
The ubiquitin–proteasome system regulates p53-mediated transcription at p21waf1 promoter
.
Oncogene
2007
;
26
:
4199
208
.
30.
Mizushima
N
,
Levine
B
,
Cuervo
AM
,
Klionsky
DJ
.
Autophagy fights disease through cellular self-digestion
.
Nature
2008
;
451
:
1069
75
.
31.
Orenstein
SJ
,
Cuervo
AM
.
Chaperone-mediated autophagy: molecular mechanisms and physiological relevance
.
Semin Cell Dev Biol
2010
;
21
:
719
26
.
32.
Seglen
PO
,
Gordon
PB
.
3-Methyladenine: specific inhibitor of autophagic/lysosomal protein degradation in isolated rat hepatocytes
.
Proc Natl Acad Sci U S A
1982
;
79
:
1889
92
.
33.
Feder
ME
,
Hofmann
GE
.
Heat-shock proteins, molecular chaperones, and the stress response: evolutionary and ecological physiology
.
Annu Rev Physiol
1999
;
61
:
243
82
.
34.
Baker
SJ
,
Reddy
EP
.
CDK4: a key player in the cell cycle, development, and cancer
.
Genes Cancer
2012
;
3
:
658
69
.
35.
Day
PJ
,
Cleasby
A
,
Tickle
IJ
,
O’Reilly
M
,
Coyle
JE
,
Holding
FP
, et al
.
Crystal structure of human CDK4 in complex with a D-type cyclin
.
Proc Natl Acad Sci U S A
2009
;
106
:
4166
70
.
36.
Sobhani
N
,
D’Angelo
A
,
Pittacolo
M
,
Roviello
G
,
Miccoli
A
,
Corona
SP
, et al
.
Updates on the CDK4/6 inhibitory strategy and combinations in breast cancer
.
Cells
2019
;
8
:
321
.
37.
Zhao
S
,
Ma
D
,
Xiao
Y
,
Li
XM
,
Ma
JL
,
Zhang
H
, et al
.
Molecular subtyping of triple-negative breast cancers by immunohistochemistry: molecular basis and clinical relevance
.
Oncologist
2020
;
25
:
e1481
e91
.
38.
Chou
TC
.
Drug combination studies and their synergy quantification using the Chou-Talalay method
.
Cancer Res
2010
;
70
:
440
6
.
39.
Masuda
H
,
Baggerly
KA
,
Wang
Y
,
Zhang
Y
,
Gonzalez-Angulo
AM
,
Meric-Bernstam
F
, et al
.
Differential response to neoadjuvant chemotherapy among 7 triple-negative breast cancer molecular subtypes
.
Clin Cancer Res
2013
;
19
:
5533
40
.
40.
Schram
AW
,
Goldfischer
S
,
Van Roermund
C
,
Brouwer-Kelder
EM
,
Collins
J
,
Hashimoto
T
, et al
.
Human peroxisomal 3-oxoacyl-coenzyme A thiolase deficiency
.
Proc Natl Acad Sci U S A
1987
;
84
:
2494
6
.
41.
Colas
E
,
Perez
C
,
Cabrera
S
,
Pedrola
N
,
Monge
M
,
Castellvi
J
, et al
.
Molecular markers of endometrial carcinoma detected in uterine aspirates
.
Int J Cancer
2011
;
129
:
2435
44
.
42.
Yan
H
,
Li
Z
,
Shen
Q
,
Wang
Q
,
Tian
J
,
Jiang
Q
, et al
.
Aberrant expression of cell cycle and material metabolism related genes contributes to hepatocellular carcinoma occurrence
.
Pathol Res Pract
2017
;
213
:
316
21
.
43.
Wang
S
,
Fei
R
,
Xu
X
,
Xu
J
,
Yao
Y
,
Lu
J
, et al
.
25-hydroxycholesterol regulates migration, invasion and EMT of colorectal cancer through miR-92a-3p/ACAA1/NF-κB Pathway
.
2021
.
44.
Wang
CY
,
Chao
YJ
,
Chen
YL
,
Wang
TW
,
Phan
NN
,
Hsu
HP
, et al
.
Upregulation of peroxisome proliferator-activated receptor-alpha and the lipid metabolism pathway promotes carcinogenesis of ampullary cancer
.
Int J Med Sci
2021
;
18
:
256
69
.
45.
Deming
S
,
Gao
Y-T
,
Long
J
,
Shu
X-O
,
Beeghly-Fadiel
A
,
Cai
Q
, et al
.
Abstract# 3945: Genetic variation in estrogen esterification pathway genes ACAA1, ACAA2, and LCAT and breast cancer risk
.
AACR
;
2009
.
46.
Fassl
A
,
Geng
Y
,
Sicinski
P
.
CDK4 and CDK6 kinases: From basic science to cancer therapy
.
Science
2022
;
375
:
eabc1495
.
47.
Dick
FA
,
Goodrich
DW
,
Sage
J
,
Dyson
NJ
.
Non-canonical functions of the RB protein in cancer
.
Nat Rev Cancer
2018
;
18
:
442
51
.
48.
Vijayaraghavan
S
,
Karakas
C
,
Doostan
I
,
Chen
X
,
Bui
T
,
Yi
M
, et al
.
CDK4/6 and autophagy inhibitors synergistically induce senescence in Rb positive cytoplasmic cyclin E negative cancers
.
Nat Commun
2017
;
8
:
15916
.
49.
Dean
JL
,
McClendon
AK
,
Hickey
TE
,
Butler
LM
,
Tilley
WD
,
Witkiewicz
AK
, et al
.
Therapeutic response to CDK4/6 inhibition in breast cancer defined by ex vivo analyses of human tumors
.
Cell Cycle
2012
;
11
:
2756
61
.
50.
Li
Z
,
Razavi
P
,
Li
Q
,
Toy
W
,
Liu
B
,
Ping
C
, et al
.
Loss of the FAT1 tumor suppressor promotes resistance to CDK4/6 inhibitors via the hippo pathway
.
Cancer Cell
2018
;
34
:
893
905
.
51.
Goel
S
,
Bergholz
JS
,
Zhao
JJ
.
Targeting CDK4 and CDK6 in cancer
.
Nat Rev Cancer
2022
;
22
:
356
72
.
52.
Zhu
X
,
Chen
L
,
Huang
B
,
Li
X
,
Yang
L
,
Hu
X
, et al
.
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
2021
;
40
:
122
.
53.
Christenson
JL
,
O’Neill
KI
,
Williams
MM
,
Spoelstra
NS
,
Jones
KL
,
Trahan
GD
, et al
.
Activity of combined androgen receptor antagonism and cell cycle inhibition in androgen receptor positive triple negative breast cancer
.
Mol Cancer Ther
2021
;
20
:
1062
71
.
54.
Ge
JY
,
Shu
S
,
Kwon
M
,
Jovanovic
B
,
Murphy
K
,
Gulvady
A
, et al
.
Acquired resistance to combined BET and CDK4/6 inhibition in triple-negative breast cancer
.
Nat Commun
2020
;
11
:
2350
.
55.
Ni
Y
,
Schmidt
KR
,
Werner
BA
,
Koenig
JK
,
Guldner
IH
,
Schnepp
PM
, et al
.
Death effector domain-containing protein induces vulnerability to cell cycle inhibition in triple-negative breast cancer
.
Nat Commun
2019
;
10
:
2860
.
56.
Foidart
P
,
Yip
C
,
Radermacher
J
,
Blacher
S
,
Lienard
M
,
Montero-Ruiz
L
, et al
.
Expression of MT4-MMP, EGFR, and RB in triple-negative breast cancer strongly sensitizes tumors to erlotinib and palbociclib combination therapy
.
Clin Cancer Res
2019
;
25
:
1838
50
.
57.
Teo
ZL
,
Versaci
S
,
Dushyanthen
S
,
Caramia
F
,
Savas
P
,
Mintoff
CP
, et al
.
Combined CDK4/6 and PI3Kalpha inhibition is synergistic and immunogenic in triple-negative breast cancer
.
Cancer Res
2017
;
77
:
6340
52
.
58.
McClellan
KJ
,
Plosker
GL
.
Trimetazidine. A review of its use in stable angina pectoris and other coronary conditions
.
Drugs
1999
;
58
:
143
57
.
59.
Andela
VB
,
Altuwaijri
S
,
Wood
J
,
Rosier
RN
.
Inhibition of beta-oxidative respiration is a therapeutic window associated with the cancer chemo-preventive activity of PPARgamma agonists
.
FEBS Lett
2005
;
579
:
1765
9
.
60.
Atli Sekeroglu
Z
,
Sekeroglu
V
,
Isik
S
,
Aydin
B
.
Trimetazidine alone or in combination with gemcitabine and/or abraxane decreased cell viability, migration and ATP levels and induced apoptosis of human pancreatic cells
.
Clin Res Hepatol Gastroenterol
2021
;
45
:
101632
.