Skip to main content
Erschienen in: Journal of Translational Medicine 1/2022

Open Access 01.12.2022 | Research

Treatment with eFT-508 increases chemosensitivity in breast cancer cells by modulating the tumor microenvironment

verfasst von: Zhao-ying Yang, Cheng-wei Jiang, Wen-long Zhang, Guang Sun

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2022

Abstract

Background

Patients with triple-negative breast cancer (TNBC) are better responders to neoadjuvant chemotherapy; however, they are poor in the durability of response with decreased overall and progression-free survival.

Methods

Given that significant improvements have been reported with PD-L1-PD-1 blockade in different cancers, we evaluated the in vitro and in vivo effectiveness of Tomivosertib (eFT-508), an anthracycline, adriamycin, and MNK1/2 inhibitor, which has been previously shown to inhibit translation of PD-L1 in mice model of liver cancer, alone or in combination using BC cell lines and an orthotopic xenograft mice model using the TNBC cell line MDA-MB-231.

Results

Within the context of The Cancer Genome Atlas (TCGA) dataset, expression of CD274 mRNA, which encodes programmed death-ligand 1 (PD-L1), was found to be significantly overexpressed in TNBC patients compared to patients with HER2 + or luminal breast cancer (BC). Even within TNBC sub-types, CD274 expression was significantly higher in the immune modulatory subtype (TNBC-IM).
BC cells exhibited high IC50 = 0.85 ± 0.07 nM with Adriamycin and significantly lower IC50 = 0.23 ± 0.04 nM with eFT-508 (P < 0.01). Combination treatment showed in vitro synergism on chemosensitivity. Combination therapy also exhibited a synergistic effect on inhibition of tumor growth and lung colonization in vivo. Mass cytometry-based evaluation of the tumor microenvironment revealed significant attenuation of both PD-L1 and PD-L2 following mono- or combination therapy with eFT-508.

Conclusions

Treatment with eFT-508 restored effector and cytotoxic function of tumor-infiltrating CD8 + T cells in mice. The remarkable efficacy observed both in vitro and in vivo, and clinical synergism with adriamycin, highlights the potential of eFT-508 as an alternative, yet more efficacious, therapeutic option for patients with TNBC.

Background

Breast cancer (BC) is still the leading cause of women's fatality [1, 2]. Among them, triple-negative breast cancer (TNBC) is a subtype of BC, in which tumors do not express HER2/neu, estrogen receptor (ER), or progesterone receptor [3, 4]. TNBC accounts for about 20% of all BC cases [3, 4]. TNBC is a low-differentiated, high-grade invasive tumor with a high tendency of metastasis and poor prognosis [3, 5]. With the understanding of TNBC and the improvement of treatment methods, the survival rate of patients with BC has been improved, but the mortality caused by chemotherapy resistance and postoperative tumor metastasis is still rising [6, 7]. Therefore, it is still necessary to find more pathogenic mechanisms in a clinical setting to provide more effective treatment entry points for these patients [8].
TNBCs are highly heterogeneous and are further sub-divided into six sub-types based on the gene expression profiles [4]. The immunomodulatory sub-type (TNBC-IM) is characterized by enrichment of genes associated with antigen cross-presentation, immune, and cytokine signaling [4]. In addition, TILs often lose function due to the expression of immune checkpoint receptors, including the expression of programmed cell death protein 1 (PD-1) [9]. PD-1 binds to its two ligands, PD-L1 and PD-L2, which are expressed on the surface of tumor cells [10]. The breast tumors express PD-L1, and this expression is related to the negative receptor state [10, 11]. In addition, studies have found that the treatment effect of patients with TNBC is significantly improved using anti-PD-1/PD-L1 single drug treatment (29,424,936). However, their exact role in the pathogenesis of TNBC is still unclear.
The cells in primary tumors are mainly epithelial in nature, which are tightly bound and cannot migrate to secondary sites. They undergo phenotypic changes (lower expression of epithelial cell markers and higher expression of mesenchymal cell markers) and functional changes (increased migration and invasiveness) to make cancer cells migrate to secondary sites. This process is called epithelial to mesenchymal transition (EMT) [12, 13]. Anthracycline class drugs, such as Doxorubicin (adriamycin) are used in chemotherapy of early and metastatic BC in combination with other drugs [14]. Animal model studies have shown that the antitumor effect of anthracyclines in fully immunized mice is better than that in immunodeficient mice, indicating that the activation of this adaptive immune response depends on the role of the immune system. Anthracyclines, a class of chemotherapeutic drugs, can induce tumor cell death, activate the immune system to fight against dead cell antigens, and endow dead tumor cells with immunogenicity. However, they have low efficacy and high toxicity [15]. The inhibitors of protein translation are increasingly being tested in clinical trials to treat cancers and have shown promising initial results [16]. Mitogen-activated protein kinase (MAPK)-interacting kinase 1 and 2 (MNK1/2) phosphorylates eIF4E at Serine 209 residue. Phosphorylation of eIF4E initiates a cascade of events, leading to the initiation of translation [16]. A recently developed MNK1/2 inhibitor, Tomivosertib or eFT-508 [16], has been shown to inhibit the expression of PD-L1 and slow down tumor progression in the mouse model of liver cancer [17], but the efficacy in TNBC is still unknown.
Therefore, in the present study, we tested the in vitro and in vivo effectiveness of adriamycin and eFT-508 alone or in combination using BC cell lines and an orthotopic xenograft mice model using the TNBC cell line MDA-MB-231. EFT-508 alone showed significant efficacy both in vitro and in vivo and showed a synergistic effect with adriamycin. Thus, it acted as a viable alternative and a more effective treatment option for patients with TNBC.

Methods

Cell culture and treatment

The human BC cell lines, MDA-MB-231, MDA-MB-468, MCF7, T47D, and SUM149 (American Type Culture Collection, Manassas, USA) were cultured in Dulbecco’s Modified Eagle Medium (DMEM, Thermo Fisher Scientific, USA). The medium was supplemented with 10% fetal bovine serum (FBS, Invitrogen, Carlsbad, CA, USA) and 1% penicillin–streptomycin solution. All cell lines were incubated in 5% CO2 at 37 °C where indicated cells were treated with indicated doses of adriamycin (Doxorubicin) and/or Tomivosertib (eFT-508) (Selleckchem) for 24 h.

Polysome profiling

Polysome profiling was done following a previously described protocol [18]. Briefly, cells were treated with 50 µg/mL with cycloheximide (Sigma-Aldrich, St. Louis, USA) for 30 min at 37 °C and then washed with cycloheximide-supplemented cold PBS. Cells were lysed, and post-nuclear extracts were layered on 10–50% sucrose gradients and centrifuged at 100,000g for 4 h. Gradient fractionation was performed using the UA-6 UV detector (Teledyne ISCO, US) based on 254 nm absorption over time.

Puromycin end-labeling assay

After the indicated treatment regimen, puromycin (C58-58–2, Sigma Aldrich) was added at a final concentration of 1 μM (100-fold dilution) for 12 h, and the cells were cultured for an additional 30 min before harvesting and cell lysate preparation. Cell lysates were subjected to immunoblot analysis. Blots were probed with an anti-puromycin antibody (Kerafast) to assay relative puromycin incorporation. Finally, blots were stained with Coomassie to confirm equal loading across different samples.

Immunoblot analysis

Normal cells and BCs were plated in 6-well plates at a density of 3.0 × 105 cells/well and cultured in CCSCs medium. After 24 h, cells were transferred to serum-free medium and incubated for 18 h. After the cells were treated according to the established groups, the cells were collected. Next, we added cell lysis buffer (50 mm tris–HCl, pH 7.5, 150 mm NaCl, 1 mm sodium-EDTA, 1 mm EDTA, 1% (v/v) Triton x-100, 2.5 mM sodium pyrophosphate, 1 mM β-glycerophosphate, 1 mM Na3VO4, 1 mM NaF, 1 μg/mL leupeptin, and 1 mM PMSF). The protein concentration was determined by the BIORAD reagent, and the loading volume of protein was calculated. Protein bands were separated based on molecular weights via 10% (w/v) SDS-PAGE (electrophoresis conditions: concentration gel 100 V, 25 min; separation gel 200 V, 40–50 min), and then transferred to PVDF membrane (transfer condition: 100 V, 2 h). The membrane was sealed with 5% (w/v) skimmed milk powder at room temperature for 1.5–2 h, followed by the addition of corresponding primary antibody and incubate at 4 °C overnight. Finally, the secondary antibody was incubated at room temperature for 1.5–2 h, and the protein image was obtained using a gel imager. The following primary antibodies were used: anti-P-eIF4E (ab1126), eIF4E (ab238519), and GAPDH (ab181603), all antibodies were purchased from Abcam China and used at 1:1000 dilution. GAPDH was used as the loading control.

Quantification of cytokines

We added 200 μL of assay buffer to the cells in the wells of the experimental titration plate and then placed on a shaking table for 10 min. The assay buffer was removed with a vacuum pump; 25 μL of assay buffer was added into the sample hole and appropriate matrix solution was added into the standard hole and quality control hole of back measuring hole. Finally, 25 μL of the sample was added into the appropriate hole, shook, and 25 μL of the microspheres were mixed into each hole. The board was closed with special plastic film and shook at 4 °C for 18–20 h. The residual liquid was gently removed with a vacuum pump. Next, 200 μL of the wave buffer was added to each hole for cleaning, followed by the addition of 25 μL of secondary antibody into each hole. The plate was incubated in a shaker for 2 h, then 25 µL of streptavidin phycoerythrin was added into the well containing the second antibody. The cells were incubated on shaking table for 30 min at room temperature, followed by the addition of 150 µL of sheath solution to all holes. The expressions of TNFα, IL-10, IL-8, CXCL-10, and IL-6 were measured by Luminex (Millipore) in plasma samples obtained from mice in different experimental groups.

Cell isolation, culture, and treatment

Positive selection was used to isolate CD8 + T cells (104 cells/hole) from tumors using magnetic-activated cell sorting using CD3 and CD8 microbeads (Miltenyi Biotec). The cells were cultured in 6-well plates coated with OKT3 (Ortho Biotech; 1 μg/mL) and anti-CD28 (clone CD28.2, Becton Dickinson; 1 μg/mL) antibodies. The cells were cultured in DMEM medium without double antibody for 4 days, and the supernatant was aspirated, and the PCR method and culture method were used to detect whether there was mycoplasma contamination. The cells in the culture flask with good growth condition and cell fusion degree of 75–80% were washed twice with PBS buffer and digested with trypsin. The degree of cell digestion was observed under a microscope. When the cells became round, and the intercellular space became larger, an appropriate amount of 10% (v/v) FBS 1640 medium was added to stop the digestion. The cells were gently blown down using a blow tube and transferred into a 15 mL centrifuge tube. After centrifugation at 800 rpm for 5 min, the supernatant was discarded and passaged at a ratio of 1:2.

Determination of cell viability by MTT assay

The cells of each group planted in the 96-well plate were taken, and after culturing for 48 h, 20 μL of MTT solution was added to the cells, and the incubation was continued for 4 h. Remove the supernatant solution with a pipette, add 150 μL of DMSO to each well, observe the dissolution of the crystals, and measure the absorbance (A) value of each well at 490 nm. A larger A value indicates higher cell viability.

Spontaneous xenograft metastasis model

All animal experiments were approved by the Laboratory Animal Welfare and Ethics Committee of the Third Military Medical University. MDA-MB-231 cells (0.5 × 106) were trypsinized, washed with PBS, and resuspended in a 1:1 solution of PBS and Matrigel (phenol red-free and reduced growth factors, BD Biosciences) and injected into the 4th mammary fat pad of 8-week old female BALB/c mice (n = 20). Tumor volume was measured every day using a caliper. When the tumors reached 50 mm3 in size (around day 7), the mice were randomly divided into four experimental groups (n = 5/group)—vehicle (DMSO), adriamycin (3 mg/kg/day), eFT-508 (1 mg/kg/day), and eFT-508 (1 mg/kg/day) + adriamycin (3 mg/kg/day)—all injections were intratumoral. Bodyweight, tumor growth rate, and survival were analyzed for each group. The mice were sacrificed either when tumor volume reached 3000 mm3 or at the end of 8 weeks, whichever was earlier. The tumors and lungs were surgically removed, weighed, fixed in 4% polyformaldehyde, and stored at − 80 °C.

Hematoxylin & eosin (H&E) and Immunohistochemistry (IHC) staining

Next, 4 µm sections were either processed for H&E staining using routine methodologies or IHC. The sections were dewaxed using xylene I and II for 10 min, respectively. A cover glass was prepared in advance, covered with water, 100% (I, II), 90%, 80%, and 70% alcohol for 5 min, respectively, and rinsed with tap water for 15 min; dyed with hematoxylin for 5 min. The dyeing time could be appropriately increased or reduced. After differentiation with 5% acetic acid for 1 min, the sections were rinsed with running water, followed by the addition of acetic acid with a pipette, and the tissue was covered on the slide. After differentiation, the color became light and blue; Blue returning: Blue returning liquid, which is not available in the laboratory, can also be used; the sections were stained with Eosin for 1 min, dehydrated using 70%, 80%, 90%, and 100% alcohol for 10 s and xylene for 1 min. The sections were dried naturally in the fume hood and then sealed for about 5 min. Next, a drop of neutral gum was dropped to seal the film, and a straw was used to drop a very small drop; the tissue was covered after pressing the film to avoid bubbles in the middle. For IHC, sections were treated for antigen retrieval and incubated with primary antibodies (P-eIF4E, 1:200; Cell Signaling Technology) overnight at 4 °C. The images were obtained using an Olympus light microscope. We randomly selected three visual fields and analyzed three slices per sample at 20×magnification.

Mass cytometry (CyTOF) staining

Cells were stained with cisplatin (DVS-Sciences) to identify live cells. Then chemoresistant cell lines were incubated with 19 metal-tagged anti-PD-L1 antibodies (Biolegend, 124,314), followed by an intercalator (DVS Sciences). Staining was done following guidelines of the Maxpar cell staining protocol (Fluidigm Corporation) and then incubated at 4 °C for 30 min [17]. CyTOF-II mass cytometer (Fluidigm) was used for sample runs. Post-run, the EQ Four Element Calibration Beads (EQ Beads, 201,078, Fluidigm) were used to normalize the signal. Events were initially gated for viability using Ir191 vs. Ir193, and double-positive cells were used in downstream analyses to identify different sub-populations post-dynamic downsampling.

Mixed lymphocyte reaction (MLR) assay

Responders CD3 + CD8 + T cells, sorted from tumor-infiltrating immune cells, were stained with CFSE (Thermo Fisher Scientific) and seeded into 96-well plate format in RPMI. Also, 10% AB serum either alone or in combination with irradiated (40 Gy) allogeneic PBMCs (stimulators) at three different ratios—1:1, 2:1, and 5:1. CD14+ antigen was present in all wells. T cell proliferation was calculated by analyzing the CFSE dilution of the responders after 72 h.

Effector T cell functional assay

Sorter tumor-infiltrating CD3 + CD8 + cells were cultured in medium ± PMA (20 ng/mL; Sigma-Aldrich, St. Louis, MO) and Ionomycin (1 µg/mL; Sigma-Aldrich). Luminex (Millipore) was used to quantify the levels of indicated cytokines in the supernatants collected before or after 3 days of PMA/Ionomycin stimulation.

Data mining

Transcriptomic analyses of CD274 expression data in invasive breast carcinoma from The Cancer Genome Atlas (TCGA) were performed using the interactive web resource UALCAN (http://​ualcan.​path.​uab.​edu; Accession date: November 8, 2020) [19]. Survival probability based on low and high expression of CD274 was computed by Kaplan–Meier curve analysis and Log-rank text using USALCAN interactive portal.

Statistical analysis

Quantitative data were presented as mean ± standard deviation (SD). Statistical analysis between groups was performed using Student’s t-test (two-group comparison) or ANOVA (multiple group comparisons). Kaplan–Meier curves were computed to analyze survival rates, and Log-rank (Mantel-Cox) test was used to evaluate statistical significance. A P-value < 0.05 was considered to be statistically significant.

Results

Expression of mRNA encoding PD-L1 is higher in TNBC-IM subtypes

We initially determined CD274 (encodes PD-L1) mRNA expression within the TCGA dataset in invasive breast carcinoma (BRCA). The expression of CD274 mRNA was slightly significantly lower in primary tumor tissues compared to the normal tissues (Fig. 1A; P = 0.049305). However, there was no correlation between CD274 expression and the overall survival in patients with BC (Fig. 1B; P = 0.8). There was no correlation between CD274 expression and disease stage (Additional file 1: S1A) and race (Additional file 1: Figure S1B).
Next, we evaluated CD274 expression in BRCA based on BC subclasses, luminal, HER2 + , and TNBC. The expression of CD274 was higher compared to both normal (P = 0.0101171) and luminal (P = 0.023656), but not HER2 + (P = 0.103021) (Fig. 2A). Then we determined CD274 expression within the different subtypes of TNBC. The expression of CD274 was statistically significant different in TNBC-IM compared to normal (P = 0.0138987), luminal (P = 0.016004), HER2 + (P = 0.020079), TNBC-BL1 (P = 0.025676), TNBC-LAR (P = 0.010937), TNBC-MSL (P = 0.0119235), and TNBC-UNS (P = 0.030065) (Fig. 2B). Thus, PD-L1 mRNA expression had a significant correlation with the receptor-negative status [13, 14], especially to the TNBC-IM subtype.

Combination treatment with eFT-508 and adriamycin exhibits a synergistic effect on in vitro chemosensitivity of BC cell lines

Since PD-L1 mRNA expression was significantly higher in TNBC patient samples, we next evaluated if cancer cell lines representing different subtypes had different chemosensitivity to adriamycin and eFT-508 alone and in combination. Luminal cell lines MCF7 and T47D, basal-like, EGFR + SUM149, and TNBC cell lines MDA-MB-231 and MDA-MB-468 cells were used. The exponentially growing cultures of these cell lines were treated with increasing concentrations of adriamycin for 24 h. All cell lines were chemoresistant with a high IC50 ranging from 48.3 to 49.2 nM (Fig. 3A). However, when these cells were treated with increasing concentration of eFT-508, MDA-MB-231 (IC50 = 0.31 ± 0.02 nM) and MDA-MB-468 (IC50 = 1.21 ± 0.01 nM) cell lines exhibited significantly higher chemosensitivity compared to MCF7 (IC50 = 11.38 ± 1.02 nM), T47D (IC50 = 10.19 ± 0.01 nM), or SUM149 (IC50 = 9.14 ± 0.14 nM) cell lines (Fig. 3B; P < 0.05 in each case). MDA-MB-231 cells were significantly more chemosensitive compared to MDA-MB-468 cells (Fig. 3B). Combination treatment with adriamycin and eFT-508 revealed higher in vitro chemosensitivity with significantly lower IC50 in all cell lines compared to either monotherapy (Fig. 3C). Again, the IC50 was significantly lower in MDA-MB-231 cells compared to each of the other four cell lines (Fig. 3C). Western blot results showed that PD-L1 (Proteintech, Mouse Monoclonal| Catalog number: 66248–1-Ig, Fig. 3D) and PD-L2 (Proteintech, Rabbit Polyclonal, Catalog number: 18251–1-AP, Fig. 3E) were highly expressed in BC cells. In addition, five cell lines were treated with eFT-508; the results of WB showed a decreased expression of PD-L1 and PD-L2 (Fig. 3F). Immunoblot analysis confirmed a robust decrease in P-eIF4E (S209) in each of the cell lines treated with eFT-508 without any change in total eIF4E expression (Fig. 3G). Polysome profiling indicated the inhibition of global translation following eFT-508 treatment in these cells, as evident by a decrease in both 80S and polysome peaks (Fig. 3H and data not shown). Polysome profiling results were further corroborated by puromycin labeling. Treatment with eFT-508 (Fig. 3I) but not Adriamycin (Fig. 3J) resulted in a robust decrease in puromycin labeling in both MDA-MB-231 and MCF7 cells. To determine if the observed results were due to clonality of the cell lines tested, we next determined the response of the parental MDA-MB-231 and multidrug resistant MDA-MB-231/ADR cells to adriamycin and eFT-508 treatment, alone or in combination. Indeed, the synergistic effect of adriamycin and eFT-508 was conserved in this model too (Fig. 4A–C). Again, the difference in chemosensitivity was not due to the differential effect of eFT-508 on translational inhibition as evidenced by puromycin labeling (Fig. 4D). Hence, the observed difference in chemosensitivity of the TNBC cell line MDA-MB-231 compared to the other cell lines tested was not due to the differential effect of eFT-508 on eIF4E phosphorylation and translational inhibition, indicating that the difference was instead dictated by the receptor negativity of the BC cell lines.

Combination treatment with eFT-508 and adriamycin exhibits a synergistic effect on in vivo chemosensitivity of breast tumor xenografts

An orthotopic model of spontaneous metastasis was established by injecting MDA-MB-231 cells in the mammary fat pad of BALB/c mice. Once tumors reached a volume of 50 mm3, mice were randomly divided into four groups (n = 5/group)—vehicle (DMSO), adriamycin (3 mg/kg/day), eFT-508 (1 mg/kg/day), and eFT-508 (1 mg/kg/day) + adriamycin (3 mg/kg/day)—all injections were intratumoral. Tumors reached the size of 50 mm3 at approximately day 7 in all mice. Tumor growth, as well as metastatic dissemination, was followed for up to 8 weeks after the initial injection. Tissues were harvested either when mice were dead or when tumor volume reached 3000 mm3. All mice in the vehicle group died by the end of week 6. Hence body weights were measured for 5 weeks. No significant difference in body weight was observed among the animals from the different experimental groups (Fig. 4E). The tumor growth rate was significantly higher in the vehicle and adriamycin groups compared to eFT-508 or combination therapy groups (P < 0.0001 in each case). Combination therapy exhibited synergism in attenuating tumor growth rate compared to eFT-508 monotherapy (Fig. 4F; P < 0.05).
After the animals were sacrificed, we counted the micrometastatized lung nodules and plotted them. Both eFT-508 monotherapy and combination therapy significantly reduced the incidence of macrometastatic lung nodules (Fig. 4G; P < 0.0001 compared to either vehicle or adriamycin group). No statistical difference was observed between eFT-508 monotherapy and combination groups (Fig. 4G; P > 0.05). Similarly, eFT-508 monotherapy or combination therapy significantly improved survival compared to vehicle or adriamycin groups (Fig. 4H). Thus, these results showed potent anti-tumorigenic and anti-metastatic effects of eFT-508 and revealed synergism with adriamycin only in the context of tumor growth rate. H&E staining of lung tissues (Fig. 5A–D). H&E staining showed that the combined drug group had fewer lung metastases than the single adriamycin group, which was consistent with the results in Fig. 4G. IHC staining confirmed the suppression of phosphorylation of eIF4E in the eFT-508 monotherapy (Fig. 5G) and combination (Fig. 5H) groups compared to vehicle (Fig. 5E) and adriamycin (Fig. 5F) groups.

Combination therapy had a synergistic effect on the expression of PD-L1 and PD-L2 by the tumor cells

Next, we evaluated the tumor microenvironment for PD-L1 and PD-L2 expression using mass cytometry. Compared to tumors in the vehicle (Fig. 6A, E) or adriamycin treatment group (Fig. 6B, E), both eFT-508 monotherapy (Fig. 6C, E) and combination therapy (Fig. 6D, E ) significantly downregulated PD-L1 + and PD-L2 + breast tumor cells. Even though there was no significant difference in PD-L1 + and PD-L2 + breast tumor cells between vehicle and adriamycin groups, combination therapy with adriamycin and eFT-508 showed a synergistic effect compared to both adriamycin and eFT-508 monotherapy groups (Fig. 6C–E). These results indicated that treatment with eFT-508 was perhaps inducing immune modulation potentiating tumor cell clearance.

Treatment with eFT-508 restores the functional activity of tumor-infiltrating cytotoxic T cells

Since the overexpression of PD-L1 was associated with tumor exhaustion and functional impairment [20, 21], we hypothesized that eFT-508-mediated inhibition of PD-L1 expression could restore the effector function of tumor-infiltrating CD8 + T cells (TILs). Hence, the functional capacity of tumor-infiltrating CD8 + T cells in mice treated with vehicle and eFT-508 monotherapy was assessed. CD8 + T cells isolated from mice treated with vehicle demonstrated attenuated expression of Th1/Th2 cytokines, interleukin (IL)-17, interleukin (IL)-10 interferon-gamma (IFN- γ), and tumor necrosis factor-alpha (TNFα), both in the native state and after polyclonal stimulation with PMA and ionomycin (Fig. 7A). In comparison, the expression of all four cytokines was higher in tumor-infiltrating T cells isolated from mice treated with eFT-508 in the native state and significantly induced after polyclonal stimulation (Fig. 7A). Furthermore, the effector function of tumor-infiltrating CD8 + T was determined using the mixed lymphocyte reaction (MLR) assay. The tumor-infiltrating CD8 + T cells in mice treated with vehicle control had impaired alloreactivity compared to those in mice treated with eFT-508 (Fig. 7B), consistent with the altered effector function. Thus, these results established that treatment with eFT-508 alone or in combination with adriamycin had a synergistic effect on tumor growth, metastatic disease progression, potentially due to its ability to restore effector function of tumor-infiltrating cytotoxic CD8 + T cells.

Discussion

Patients with TNBC responded better to neoadjuvant chemotherapy; however, OS and PFS still decreased [11, 22]. Therefore, there was an urgent need to find alternative treatment strategies. The evasion of immune surveillance is a crucial adaptive mechanism for tumor progression [23]. The expression of immune checkpoint receptors by tumor-infiltrating T cells and their cognate ligands by tumor cells is a potent mechanism to drive T cell exhaustion, resulting in impaired T cell function and immune evasion [20, 21]. In this study, we found that eFT-508, an MNK1/2 inhibitor, enhanced the sensitivity of in vitro chemotherapy and prevented tumor growth and metastasis in vivo by inhibiting the expression of PD-L1. Our results indicated that eFT-508 mediated its anti-tumorigenic activity partially by inhibiting PD-L1 expression and restoring the effector function of tumor-infiltrating cytotoxic T cells. Of note, MDA-MB-231 cells were more chemosensitive compared to the MDA-MB-468 cells, despite both of them being TNBC cell lines. This is potentially due to the fact that MDA-MB-468 is a basal BC cell line whereas MDA-MB-231 is a claudin-low cell line. MDA-MB-231 is characterized by low expression of claudin 3, 4, and 7, as well as low expression of Ki67 and the epithelial cell marker E-cadherin. In contrast expression of each one of these markers is higher in the MDA-MB-468 cells. So, the observed difference among these two TNBC cell lines can be due to relative enrichment of mesenchymal cell markers and cancer stem cells [2426].
PD-L1/PD1 is a key molecule in tumor development. Despite significant improvements in PD-L1-PD-1 blockade in different cancers, only a fraction of patients have been found to show durable and objective clinical response among the high incidence of intrinsic and acquired therapy resistance [24]. Thus, it was essential to understand how the expression of PD-L1 was controlled in chronic conditions to tackle these challenges. PD-L1 has been shown to be regulated at genetic, epigenetic, transcriptional, translational, and posttranslational levels [2737], often in a context-dependent fashion. Therefore, we targeted PD-L1 to detect the effect of drugs on tumors. Additionally, PI3K/Akt activation, MAPK signaling, as well as transforming growth factor-beta (TGFβ) and tumor necrosis factor-alpha (TNFα) are known oncogenic signals that can induce PD-L1 expression [27, 3133, 38, 39]. Indeed, PD-L1 expression is known to be inversely regulated by the genetic ablation of PTEN in patients with TNBC [39]. MicroRNA (miR)-200 has been shown to inhibit translation of CD274 mRNA (which encodes PD-L1) [32, 36]. The miR-200 family is known to be regulated by TGFβ signaling pathway and functions as an inhibitor of EMT and metastatic progression in BC [12, 13]. Thus, the loss of miR-200 expression could induce the expression of PD-L1, in turn helping tumors to evade immune surveillance. Additionally, more studies are required to understand how PD-L1 is regulated and whether eFT-508 regulates tumor immunity through PD-L1.
eFT-508 is known to affect tumor growth. Studies have found that eFT-508 reduces the development and progression of liver cancer in myc-driven mouse model [17]. However, the therapeutic effect of eFT-508 on other tumors still needs further exploration. Cancer cells have evolved various cellular mechanisms for stress-adaptive translation [16, 17, 4042]; hence, therapeutic targeting of translation has evolved as a selective yet efficacious regimen [16]. PD-L1 inhibitors are partially effective, and PD-L1 transcripts are independent of the overall survival of patients with BC. Therefore, eFT-508 contributed to the partial effectiveness of PD1/PD-L1 inhibitors. Targeting these specialized adaptive translation mechanisms might dramatically improve the therapeutic index.

Conclusions

Whether eFT-508 regulated the expression of additional ligands of immune checkpoint receptors was unclear. EFT-508 seems to cause global translation inhibition. Hence, it was unclear whether the action of eFT-508 on PD-L1 expression was a direct effect or mediated indirectly by other factor(s). Furthermore, dose- and duration-based effect on toxicity and durability of antitumor response of eFT-508 in the in vivo models of TNBC remains to be determined. A phase 2, open-label study (ClinicalTrials.gov Identifier: NCT03616834) is currently being run by Effector Therapeutics to evaluate the safety profile, pharmacokinetics, and durability of response in patients who were already on anti-PD-1/anti-PD-L1 monotherapy but had progressive disease or had undergone 12 weeks of treatment with no evidence of complete or partial response. Based on our findings in the current study that eFT-508 could inhibit PD-L1 expression in TNBC and similar evidence in liver cancer [17], we speculated that the ongoing clinical trial could establish clinical synergism. Finally, whether eFT-508 had a similar effect on disease outcome in other classes of BC or if it was specific to TNBC needs further investigation.

Acknowledgements

Not applicable.

Declarations

All animal experiments were approved by the Laboratory Animal Welfare and Ethics Committee of the Third Military Medical University.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61(2):69–90.PubMedCrossRef Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61(2):69–90.PubMedCrossRef
2.
Zurück zum Zitat Li J, Zhang BN, Fan JH, Pang Y, Zhang P, Wang SL, et al. A nation-wide multicenter 10-year (1999–2008) retrospective clinical epidemiological study of female breast cancer in China. BMC Cancer. 2011;11:364.PubMedPubMedCentralCrossRef Li J, Zhang BN, Fan JH, Pang Y, Zhang P, Wang SL, et al. A nation-wide multicenter 10-year (1999–2008) retrospective clinical epidemiological study of female breast cancer in China. BMC Cancer. 2011;11:364.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat 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 : An Official J American Association for Cancer Res. 2007;13(15 Pt 1):4429–34.CrossRef 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 : An Official J American Association for Cancer Res. 2007;13(15 Pt 1):4429–34.CrossRef
4.
Zurück zum Zitat Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest. 2011;121(7):2750–67.PubMedPubMedCentralCrossRef Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest. 2011;121(7):2750–67.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Haffty BG, Yang Q, Reiss M, Kearney T, Higgins SA, Weidhaas J, et al. Locoregional relapse and distant metastasis in conservatively managed triple negative early-stage breast cancer. J Clin Oncol. 2006;24(36):5652–7.PubMedCrossRef Haffty BG, Yang Q, Reiss M, Kearney T, Higgins SA, Weidhaas J, et al. Locoregional relapse and distant metastasis in conservatively managed triple negative early-stage breast cancer. J Clin Oncol. 2006;24(36):5652–7.PubMedCrossRef
6.
Zurück zum Zitat Early Breast Cancer Trialists’ Collaborative G. Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005;365(9472):1687–717.CrossRef Early Breast Cancer Trialists’ Collaborative G. Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005;365(9472):1687–717.CrossRef
7.
Zurück zum Zitat Sayer HG, Kath R, Kliche KO, Hoffken K. Premenopausal breast cancer: chemotherapy and endocrine therapy. Drugs. 2002;62(14):2025–38.PubMedCrossRef Sayer HG, Kath R, Kliche KO, Hoffken K. Premenopausal breast cancer: chemotherapy and endocrine therapy. Drugs. 2002;62(14):2025–38.PubMedCrossRef
8.
Zurück zum Zitat Gupta GK, Collier AL, Lee D, Hoefer RA, Zheleva V, Siewertsz van Reesema LL, et al. Perspectives on triple-negative breast cancer: current treatment strategies, unmet needs, and potential targets for future therapies. Cancers (Basel). 2020;12(9):2392.CrossRef Gupta GK, Collier AL, Lee D, Hoefer RA, Zheleva V, Siewertsz van Reesema LL, et al. Perspectives on triple-negative breast cancer: current treatment strategies, unmet needs, and potential targets for future therapies. Cancers (Basel). 2020;12(9):2392.CrossRef
9.
Zurück zum Zitat Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, et al. Tumor-associated B7–H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8(8):793–800.PubMedCrossRef Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, et al. Tumor-associated B7–H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8(8):793–800.PubMedCrossRef
10.
Zurück zum Zitat Ghebeh H, Mohammed S, Al-Omair A, Qattan A, Lehe C, Al-Qudaihi G, et al. The B7–H1 (PD-L1) T lymphocyte-inhibitory molecule is expressed in breast cancer patients with infiltrating ductal carcinoma: correlation with important high-risk prognostic factors. Neoplasia (New York, NY). 2006;8(3):190–8.CrossRef Ghebeh H, Mohammed S, Al-Omair A, Qattan A, Lehe C, Al-Qudaihi G, et al. The B7–H1 (PD-L1) T lymphocyte-inhibitory molecule is expressed in breast cancer patients with infiltrating ductal carcinoma: correlation with important high-risk prognostic factors. Neoplasia (New York, NY). 2006;8(3):190–8.CrossRef
11.
Zurück zum Zitat Purwanto I, Heriyanto DS, Ghozali A, Widodo I, Dwiprahasto I, Aryandono T, et al. Overexpression of programmed death-ligand 1 receptor mRNA as an independent negative prognostic factor for triple negative breast cancer. World J Oncol. 2020;11(5):216–22.PubMedPubMedCentralCrossRef Purwanto I, Heriyanto DS, Ghozali A, Widodo I, Dwiprahasto I, Aryandono T, et al. Overexpression of programmed death-ligand 1 receptor mRNA as an independent negative prognostic factor for triple negative breast cancer. World J Oncol. 2020;11(5):216–22.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Pastushenko I, Blanpain C. EMT transition states during tumor progression and metastasis. Trends Cell Biol. 2019;29(3):212–26.PubMedCrossRef Pastushenko I, Blanpain C. EMT transition states during tumor progression and metastasis. Trends Cell Biol. 2019;29(3):212–26.PubMedCrossRef
14.
Zurück zum Zitat Ghigo A, Li M, Hirsch E. New signal transduction paradigms in anthracycline-induced cardiotoxicity. Biochimica et biophysica acta. 2016;1863:1916–25.PubMedCrossRef Ghigo A, Li M, Hirsch E. New signal transduction paradigms in anthracycline-induced cardiotoxicity. Biochimica et biophysica acta. 2016;1863:1916–25.PubMedCrossRef
15.
Zurück zum Zitat Sallustio BC, Boddy AV. Is there scope for better individualisation of anthracycline cancer chemotherapy? Br J Clin Pharmacol. 2020;87:295.PubMedCrossRef Sallustio BC, Boddy AV. Is there scope for better individualisation of anthracycline cancer chemotherapy? Br J Clin Pharmacol. 2020;87:295.PubMedCrossRef
16.
Zurück zum Zitat Pal I, Safari M, Jovanovic M, Bates SE, Deng C. Targeting translation of mRNA as a therapeutic strategy in cancer. Curr Hematol Malig Rep. 2019;14(4):219–27.PubMedCrossRef Pal I, Safari M, Jovanovic M, Bates SE, Deng C. Targeting translation of mRNA as a therapeutic strategy in cancer. Curr Hematol Malig Rep. 2019;14(4):219–27.PubMedCrossRef
17.
Zurück zum Zitat Xu Y, Poggio M, Jin HY, Shi Z, Forester CM, Wang Y, et al. Translation control of the immune checkpoint in cancer and its therapeutic targeting. Nat Med. 2019;25(2):301–11.PubMedPubMedCentralCrossRef Xu Y, Poggio M, Jin HY, Shi Z, Forester CM, Wang Y, et al. Translation control of the immune checkpoint in cancer and its therapeutic targeting. Nat Med. 2019;25(2):301–11.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Mazumder B, Poddar D, Basu A, Kour R, Verbovetskaya V, Barik S. Extraribosomal l13a is a specific innate immune factor for antiviral defense. J Virol. 2014;88(16):9100–10.PubMedPubMedCentralCrossRef Mazumder B, Poddar D, Basu A, Kour R, Verbovetskaya V, Barik S. Extraribosomal l13a is a specific innate immune factor for antiviral defense. J Virol. 2014;88(16):9100–10.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce-Rodriguez I, Chakravarthi B, et al. UALCAN: a portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia (New York, NY). 2017;19(8):649–58.CrossRef Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce-Rodriguez I, Chakravarthi B, et al. UALCAN: a portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia (New York, NY). 2017;19(8):649–58.CrossRef
20.
Zurück zum Zitat Reiser J, Banerjee A. Effector, memory, and dysfunctional CD8(+) T cell fates in the antitumor immune response. J Immunol Res. 2016;2016:8941260.PubMedPubMedCentralCrossRef Reiser J, Banerjee A. Effector, memory, and dysfunctional CD8(+) T cell fates in the antitumor immune response. J Immunol Res. 2016;2016:8941260.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Liedtke C, Mazouni C, Hess KR, Andre F, Tordai A, Mejia JA, et al. Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J Clin Oncol. 2008;26(8):1275–81.PubMedCrossRef Liedtke C, Mazouni C, Hess KR, Andre F, Tordai A, Mejia JA, et al. Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J Clin Oncol. 2008;26(8):1275–81.PubMedCrossRef
23.
Zurück zum Zitat Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.PubMedCrossRef Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.PubMedCrossRef
24.
Zurück zum Zitat Prat A, Parker JS, Karginova O, et al. Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res. 2010;12:R68.PubMedPubMedCentralCrossRef Prat A, Parker JS, Karginova O, et al. Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res. 2010;12:R68.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Neve RM, Chin K, Fridlyand J, et al. A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell. 2006;10:515–27.PubMedPubMedCentralCrossRef Neve RM, Chin K, Fridlyand J, et al. A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell. 2006;10:515–27.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Kang Y, Siegel PM, Shu W, et al. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell. 2003;3:537–49.PubMedCrossRef Kang Y, Siegel PM, Shu W, et al. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell. 2003;3:537–49.PubMedCrossRef
28.
Zurück zum Zitat Baas M, Besançon A, Goncalves T, Valette F, Yagita H, Sawitzki B, et al. TGFβ-dependent expression of PD-1 and PD-L1 controls CD8(+) T cell anergy in transplant tolerance. eLife. 2016;5:e08133.PubMedPubMedCentralCrossRef Baas M, Besançon A, Goncalves T, Valette F, Yagita H, Sawitzki B, et al. TGFβ-dependent expression of PD-1 and PD-L1 controls CD8(+) T cell anergy in transplant tolerance. eLife. 2016;5:e08133.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Jiang X, Zhou J, Giobbie-Hurder A, Wargo J, Hodi FS. The activation of MAPK in melanoma cells resistant to BRAF inhibition promotes PD-L1 expression that is reversible by MEK and PI3K inhibition. Clin Cancer Res : An Official J of The American Association for Cancer Research. 2013;19(3):598–609.CrossRef Jiang X, Zhou J, Giobbie-Hurder A, Wargo J, Hodi FS. The activation of MAPK in melanoma cells resistant to BRAF inhibition promotes PD-L1 expression that is reversible by MEK and PI3K inhibition. Clin Cancer Res : An Official J of The American Association for Cancer Research. 2013;19(3):598–609.CrossRef
30.
Zurück zum Zitat Kataoka K, Shiraishi Y, Takeda Y, Sakata S, Matsumoto M, Nagano S, et al. Aberrant PD-L1 expression through 3’-UTR disruption in multiple cancers. Nature. 2016;534(7607):402–6.PubMedCrossRef Kataoka K, Shiraishi Y, Takeda Y, Sakata S, Matsumoto M, Nagano S, et al. Aberrant PD-L1 expression through 3’-UTR disruption in multiple cancers. Nature. 2016;534(7607):402–6.PubMedCrossRef
31.
Zurück zum Zitat Sumimoto H, Takano A, Teramoto K, Daigo Y. RAS-mitogen-activated protein kinase signal is required for enhanced PD-L1 expression in human lung cancers. PLoS ONE. 2016;11(11): e0166626.PubMedPubMedCentralCrossRef Sumimoto H, Takano A, Teramoto K, Daigo Y. RAS-mitogen-activated protein kinase signal is required for enhanced PD-L1 expression in human lung cancers. PLoS ONE. 2016;11(11): e0166626.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Chen L, Gibbons DL, Goswami S, Cortez MA, Ahn YH, Byers LA, et al. Metastasis is regulated via microRNA-200/ZEB1 axis control of tumour cell PD-L1 expression and intratumoral immunosuppression. Nat Commun. 2014;5:5241.PubMedCrossRef Chen L, Gibbons DL, Goswami S, Cortez MA, Ahn YH, Byers LA, et al. Metastasis is regulated via microRNA-200/ZEB1 axis control of tumour cell PD-L1 expression and intratumoral immunosuppression. Nat Commun. 2014;5:5241.PubMedCrossRef
33.
Zurück zum Zitat Karakhanova S, Meisel S, Ring S, Mahnke K, Enk AH. ERK/p38 MAP-kinases and PI3K are involved in the differential regulation of B7–H1 expression in DC subsets. Eur J Immunol. 2010;40(1):254–66.PubMedCrossRef Karakhanova S, Meisel S, Ring S, Mahnke K, Enk AH. ERK/p38 MAP-kinases and PI3K are involved in the differential regulation of B7–H1 expression in DC subsets. Eur J Immunol. 2010;40(1):254–66.PubMedCrossRef
34.
Zurück zum Zitat Ni XY, Sui HX, Liu Y, Ke SZ, Wang YN, Gao FG. TGF-β of lung cancer microenvironment upregulates B7H1 and GITRL expression in dendritic cells and is associated with regulatory T cell generation. Oncol Rep. 2012;28(2):615–21.PubMedCrossRef Ni XY, Sui HX, Liu Y, Ke SZ, Wang YN, Gao FG. TGF-β of lung cancer microenvironment upregulates B7H1 and GITRL expression in dendritic cells and is associated with regulatory T cell generation. Oncol Rep. 2012;28(2):615–21.PubMedCrossRef
35.
Zurück zum Zitat Song S, Yuan P, Wu H, Chen J, Fu J, Li P, et al. Dendritic cells with an increased PD-L1 by TGF-β induce T cell anergy for the cytotoxicity of hepatocellular carcinoma cells. Int Immunopharmacol. 2014;20(1):117–23.PubMedCrossRef Song S, Yuan P, Wu H, Chen J, Fu J, Li P, et al. Dendritic cells with an increased PD-L1 by TGF-β induce T cell anergy for the cytotoxicity of hepatocellular carcinoma cells. Int Immunopharmacol. 2014;20(1):117–23.PubMedCrossRef
36.
Zurück zum Zitat Xie G, Li W, Li R, Wu K, Zhao E, Zhang Y, et al. Helicobacter pylori promote B7–H1 expression by suppressing miR-152 and miR-200b in gastric cancer cells. PLoS ONE. 2017;12(1): e0168822.PubMedPubMedCentralCrossRef Xie G, Li W, Li R, Wu K, Zhao E, Zhang Y, et al. Helicobacter pylori promote B7–H1 expression by suppressing miR-152 and miR-200b in gastric cancer cells. PLoS ONE. 2017;12(1): e0168822.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Zeng K, Wang Z, Ohshima K, Liu Y, Zhang W, Wang L, et al. BRAF V600E mutation correlates with suppressive tumor immune microenvironment and reduced disease-free survival in Langerhans cell histiocytosis. Oncoimmunology. 2016;5(7): e1185582.PubMedPubMedCentralCrossRef Zeng K, Wang Z, Ohshima K, Liu Y, Zhang W, Wang L, et al. BRAF V600E mutation correlates with suppressive tumor immune microenvironment and reduced disease-free survival in Langerhans cell histiocytosis. Oncoimmunology. 2016;5(7): e1185582.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Mazanet MM, Hughes CC. B7–H1 is expressed by human endothelial cells and suppresses T cell cytokine synthesis. J Immunol (Baltimor Md: 1950). 2002;169(7):3581–8.CrossRef Mazanet MM, Hughes CC. B7–H1 is expressed by human endothelial cells and suppresses T cell cytokine synthesis. J Immunol (Baltimor Md: 1950). 2002;169(7):3581–8.CrossRef
39.
Zurück zum Zitat Mittendorf EA, Philips AV, Meric-Bernstam F, Qiao N, Wu Y, Harrington S, et al. PD-L1 expression in triple-negative breast cancer. Cancer Immunol Res. 2014;2(4):361–70.PubMedPubMedCentralCrossRef Mittendorf EA, Philips AV, Meric-Bernstam F, Qiao N, Wu Y, Harrington S, et al. PD-L1 expression in triple-negative breast cancer. Cancer Immunol Res. 2014;2(4):361–70.PubMedPubMedCentralCrossRef
40.
42.
Zurück zum Zitat Uniacke J, Holterman CE, Lachance G, Franovic A, Jacob MD, Fabian MR, et al. An oxygen-regulated switch in the protein synthesis machinery. Nature. 2012;486(7401):126–9.PubMedPubMedCentralCrossRef Uniacke J, Holterman CE, Lachance G, Franovic A, Jacob MD, Fabian MR, et al. An oxygen-regulated switch in the protein synthesis machinery. Nature. 2012;486(7401):126–9.PubMedPubMedCentralCrossRef
Metadaten
Titel
Treatment with eFT-508 increases chemosensitivity in breast cancer cells by modulating the tumor microenvironment
verfasst von
Zhao-ying Yang
Cheng-wei Jiang
Wen-long Zhang
Guang Sun
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2022
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-022-03474-9

Weitere Artikel der Ausgabe 1/2022

Journal of Translational Medicine 1/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.