Home The inhibitory effects of lobaplatin, or in combination with gemcitabine on triple-negative breast cancer cells in vitro and in vivo
Article Open Access

The inhibitory effects of lobaplatin, or in combination with gemcitabine on triple-negative breast cancer cells in vitro and in vivo

  • Chengyan Jiang , Ye Zhang , Xiaoyu Xu , Shanshan Su , Huafeng Pan EMAIL logo and Aiqin Jiang ORCID logo EMAIL logo
Published/Copyright: February 10, 2023

Abstract

Objectives

To study the therapeutic effects of lobaplatin in combination with conventional chemotherapy drugs on triple-negative breast cancer (TNBC) cells.

Methods

We used the CCK-8 assay, flow cytometry, western blotting, and immunofluorescence staining methods to detect the effects of lobaplatin or in combination with gemcitabine on the survival, apoptosis, and cell cycle progression of TNBC cells. A cell-derived xenograft mouse model was used to verify the antitumor effects of lobaplatin alone or in combination with gemcitabine.

Results

Lobaplatin significantly inhibited MDA-MB-468 cell growth in vitro, either alone or in combination with gemcitabine. Lobaplatin arrested the cell cycle at the S phase, induced nuclear cell damage, and promoted apoptosis. Also, the percentage of apoptotic cells was greatly increased when lobaplatin was combined with gemcitabine. Cleaved Caspase-3 and Poly (ADP-Ribose) Polymerase-1 (PARP-1) fragments indicated that lobaplatin promoted apoptosis through the classical pathway. Lobaplatin effectively inhibited the growth of tumors in vivo. Compared with the vehicle group (567.6 ± 126.2 mm3), the tumor volume of the lobaplatin group (302.7 ± 131.6 mm3) was significantly reduced (p<0.01). The combination of lobaplatin and gemcitabine (207.7 ± 83.94 mm3) was a little better than lobaplatin alone in the inhibition of the transplanted tumor (p>0.05).

Conclusions

Lobaplatin alone or in combination with gemcitabine had significant inhibitory effects on MDA-MB-468 cells in vitro. Lobaplatin also significantly inhibited the growth of nude mice xenografts. The synergistic effect between lobaplatin and gemcitabine in vivo was minimal, perhaps due to the low dose of gemcitabine used.

Introduction

According to the data reported by the International Agency for Research on Cancer in 2020, the incidence of breast cancer ranked first worldwide [1], among which, the most malignant subtype is TNBC, which accounts for 10–15% [2]. Compared with other BC subtypes, TNBC is usually accompanied by distant metastasis and poor prognosis. TNBC lacks relevant receptors, such as estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), and is not sensitive to endocrine therapy or HER2 treatment [3]. Although there are already many new therapies for improving the survival rate of BC patients, traditional chemotherapy remains the primary systemic treatment for metastatic TNBC [4]. Conventional chemotherapy regimens for TNBC, such as DNA damaging agents (platinum), DNA synthesis inhibitors (gemcitabine, fluorouracil, etc.), anthracyclines (doxorubicin or epirubicin), and microtubule inhibitors (taxanes, vinorelbine, etc.) [5], suffer from high recurrence rates, even if the initial response rate is high.

Lobaplatin (LBP) is a third-generation platinum anticancer drug. Similar to other platinum drugs, it mainly hinders the DNA replication of tumor cells by cross-linking with the tumor cell DNA and thereby interfering with cell cycle progression [6]. However, LBP has better water solubility, a broader anti-tumor spectrum, stronger anti-tumor activity, and lower toxicity or side effects than other platinum drugs. It has no cross-resistance with other platinum drugs [7]. Thus, it has better clinical applicability than other platinum drugs. According to the instruction, LBP has been approved for the treatment of breast cancer in the clinic [8]. However, systematic studies of LBP in the treatment of TNBC are still rare [9]. The treatment effect of LBP on TNBC should be further explored.

Currently, combining therapy has become a key point to improving agent effectiveness on TNBC [3]. The main aims of synergistic therapy are to achieve a better effect, lower the dose, minimize or delay the induction of drug resistance, and reduce toxicity. Gemcitabine (GEM), a difluorinated pyrimidine analog of deoxycytidine that inhibits the elongation of replicating DNA strands [10], has been widely used for the treatment of cancers and can be used alone or in combination with other drugs, including cisplatin and carboplatin [11], [12], [13]. However, no basic research has been performed on its combination with LBP for the treatment of TNBC. Thus, in this study, the effects of LBP alone or in combination with GEM in TNBC cell lines were explored. This study aims to develop new effective therapeutic strategies for TNBC.

Materials and methods

Cell culture and reagents

Human TNBC cells MDA-MB-231 and MDA-MB-468 (Shanghai Cell Bank at Chinese Academy of Sciences) are both basal subtypes. MDA-MB-231 cells were derived from the pleural effusion of a 51-year-old Caucasian female patient with metastatic breast adenocarcinoma, and MDA-MB-468 cells were isolated from the pleural fluid of metastatic cancer in a 51-year-old black woman with breast cancer [14], [15], [16]. They were cultured in Leibovitz’s L15 medium (BasalMedia, Shanghai, China) supplemented with 10% fetal bovine serum (FBS) (Wisent, USA), 100 U/mL penicillin, and 100 μg/mL streptomycin at 37 °C in a 100% air atmosphere. GEM (Hansen, Jiangsu, China), doxorubicin hydrochloride (HISUN, Zhejiang, China), and epirubicin hydrochloride (pudepharma, Shanxi, China) were dissolved in 0.9% NaCl, while Cisplatin (Hansen, Jiangsu, China), LBP (Hainan Changan International Pharmaceutical Co.Ltd, China), Carboplatin, and Oxaliplatin were dissolved in the complete medium and stored at – 80 °C.

Cell viability

Cellular viability assay was performed by CCK-8 (Dojindo, Japan) assay. Exponentially growing cells were seeded into 96-well plates and treated with the above anticancer drugs alone or in combination on the second day. After 48 h, 10 µL of CCK-8 was added to each well and incubated at 37 °C. The optical density of cells was determined by a multifunctional microwell detector (Bio-Tek, USA) at 450 nm and expressed as absorbance values. Relative to blank-well values, values in experimental wells and control wells were normalized.

I n h i b i t i o n r a t e = ( a b s o r b a n c e v a l u e o f c o n t r o l g r o u p a b s o r b a n c e v a l u e o f e x p e r i m e n t a l g r o u p ) a b s o r b a n c e v a l u e o f c o n t r o l g r o u p × 100 %

Synergy assay

The combination index (CI) was used to evaluate the synergy of LBP and other chemotherapy drugs. This method was originally described by Chou and Talalay [17, 18]. To calculate CI values, the software CompuSyn was used, taking the entire shape of the growth inhibition curve into account for calculating whether a combination is synergistic, additive, or antagonistic [19, 20]. CI<0.90 indicates synergy, 0.91≤CI<1.10 indicates additive and CI≥1.11 indicates the antagonism of the two-drug combination.

Cellular apoptosis analysis

TNBC cells (2–3 × 105 cells) were seeded in 12-well plates overnight and treated with LBP, GEM, or LBP plus GEM for 48 h. Samples were prepared using Annexin V-FITC Apoptosis Detection Kit (Nanjing Fcmacs Biotechnology, China) according to the manufacturer’s instructions, then detected using a BD ACCURI C6 flow cytometer (BD Biosciences, USA) and analyzed by FlowJo 10.4 software (FlowJo, USA). Early apoptotic cells were Annexin-V+/PI- and late apoptotic cells were Annexin-V+/PI+.

Bliss independent test was used to detect whether LBP and GEM synergistically promote apoptosis. Suppose the apoptosis rate of LBP and GEM alone at different concentrations was Wa and Wb. Wab is calculated by the Bliss formula (Wab=Wa + Wb – WaWb). The observed apoptosis rate of LBP plus GEM combination (Wab, o) is compared with Wab. If Wab, o>Wab, the combination treatment is thought to be Synergy; if Wab, o=Wab, the combination treatment is Independent; and if Wab, o<Wab, the combination is Antagonism [21], [22], [23].

Cell cycle progression analysis

TNBC cells (2–3 × 105 cells) were seeded in 12-well plates overnight and then, the next night, treated for 24 or 48 h. Cells were harvested and resuspended in ice-cold 70% ethanol and fixed overnight at –20 °C. After washing with PBS, 20 µg/mL−1 RnaseA solution (TaKaRa, Japan) was added and incubated at 37 °C for 30 min. Then add PI staining solution (KeyGEN BioTECH, Jiangsu, China), incubate for 20 min in the dark, use a flow cytometer to detect immediately, and analyze the cell cycle distribution by FlowJo 10.4 software.

Immunofluorescence staining

MDA-MB-468 cells (6 × 105 cells) were seeded in 6-well plates containing sterile coverslips. After treatment, cells were washed with PBS, fixed in 4% paraformaldehyde for 10 min, and permeabilized with 0.1% Triton X-100/PBS for 15 min. Block with 5% bovine serum albumin (BSA) (BioFroxx, Germany) for 1 h and incubate with anti-γ-H2A.X antibody (CST, USA) diluted (1:400) in blocking buffer for 2 h at room temperature. Then incubate goat anti-rabbit IgG-iFluor 488 fluorescent secondary antibodies (1:1,000, Fcmacs) followed by DAPI staining solution (Beyotime, Shanghai, China) in the dark. Fluorescence was viewed with an Olympus Fluoview FV3000 confocal microscope system (Olympus, Center Valley, PA) using a 60 × objective and the results were semi-quantitatively analyzed with Image software.

Western blotting

Cell protein was extracted by radioimmunoprecipitation assay (RIPA) lysis buffer (Beyotime, Shanghai, China). 40 µg of total protein was electrophoresed at constant pressure (80–100 V) on a 10% SDS-polyacrylamide gel, and then the proteins were transferred to 0.2 µm PVDF membrane (Bio-Rad, Hercules) by electroporation at 350 mA. The membrane was blocked with blocking buffer for 1 h, then incubated with the specific primary antibody (1:1,000) overnight at 4 °C, and horseradish peroxidase (HRP)-linked anti-mouse/rabbit secondary antibody (1:2,000) for 1 h. Specific first antibodies: anti-GAPDH (60004-1-lg) and anti-dCK (17758-1-AP) antibodies were bought from proteintech (China), anti-cleaved caspase 3 (#9661), and anti-PARP (#9532) antibodies were bought from Cell Signaling Technology (Danvers, MA, USA). Horseradish peroxidase (HRP)-linked anti-mouse and anti-rabbit secondary antibody were bought from Fcmacs and Beyotime, respectively. Immunoreactive bands were imaged in the minichemTM chemiluminescence imaging system (SAGECREATION, China) and the intensities of bands were quantified by ImageJ software.

TNBC xenograft mouse model

All mouse experiments were performed with the approval of the Institutional Animal Care and Use Committee of Nanjing University. Forty SPF-grade female athymic BALB/c nude mice of four-week-old were purchased from GemPharmatech (Jiangsu, China) and stabilized for one week. MDA-MB-468 (1 × 107 cells) was inoculated into the right armpit of the mice. Record body weight and tumor size (V=length×width2×½) every 2 days, when the tumor grew to 80–100 mm3, the mice were randomly divided into four groups (n=4) and injected (i. p.) with 0.9% NaCl (Control), 20 mg/kg GEM followed by 0.9% NaCl (GEM), 0.9% NaCl followed by 6 mg/kg LBP (LBP), or GEM followed by LBP (LBP + GEM) once a week for 6 treatment cycles. After anesthesia, the periocular blood was collected for routine blood tests (mindray BC-5,500, Shenzhen, China), then the mice were sacrificed, and tumor tissues were removed, photographed, and weighed. The relative tumor volume (RTV)=Vt/V0, where Vt is the volume on each measurement and V0 is the volume on initial treatment. T/C(%)=(Mean RTV of the treated group)/(Mean RTV of the control group) × 100%, which is expressed for the therapeutic effect of a given compound [24, 25].

Statistical analysis

All data were obtained from at least three independent experiments and expressed as mean ± standard deviation (mean ± SD). Prism 7.0 software was mainly used for statistical analysis and graphing. Difference comparison was determined via Student’s t-test or analysis of variance (ANOVA) followed by Tukey’s test using SPSS software (IBM, Armonk, NY, USA). p<0.05 was considered the minimal level of significance.

Results

Inhibitory effect of LBP on TNBC cells

Platinum drugs are common clinical chemotherapeutic drugs. To compare the inhibitory effect of three generations of platinum drugs on TNBC cells, we treated MDA-MB-468 and MDA-MB-231 cells with cisplatin, carboplatin, oxaliplatin, and LBP. The inhibitory effect of platinum drugs on cell proliferation was determined by CCK-8 assays. The 50% inhibitory concentration (IC50) of cisplatin, carboplatin, oxaliplatin, and LBP on MDA-MB-468 cells was 6.71, 103.8, 35.48, and 7.45 µM, respectively, while the inhibitory rate of the four drugs at 25 µM was about 89.5 ± 0.2%, 16.0 ± 1.4%, 41.2 ± 1.5%, and 92.9 ± 0.1%, respectively (Figure 1A). The IC50 of cisplatin, carboplatin, oxaliplatin and LBP on MDA-MB-231 cells was 22.9, 54.37, 47.86, and 22.38 µM, respectively, while the inhibitory rate of 25 µM cisplatin, oxaliplatin, and LBP on MDA-MB-231 cells was about 51.3 ± 5.5%, 42.0 ± 1.1%, and 50.3 ± 1.3%, respectively. 25 µM carboplatin had almost no inhibitory effect on MDA-MB-231 cells (Figure 1A). Thus, we concluded that LBP exhibited a similar inhibitory effect to cisplatin, but was superior to that of carboplatin and oxaliplatin in both cell lines.

Figure 1: 
The inhibitory effects of LBP on TNBC cells. (A) The inhibition curves of three generations of platinum drugs and the bar graphs corresponding to the inhibition rates of the four drugs at 25 µM for 48 h. (B) Apoptosis detected by flow cytometry for 48 h. (C) Distribution of different phrases in the cell cycle detected by flow cytometry for 24 h. Data are represented as the mean ± SD. *p<0.05, **p<0.01, ***p<0.001.
Figure 1:

The inhibitory effects of LBP on TNBC cells. (A) The inhibition curves of three generations of platinum drugs and the bar graphs corresponding to the inhibition rates of the four drugs at 25 µM for 48 h. (B) Apoptosis detected by flow cytometry for 48 h. (C) Distribution of different phrases in the cell cycle detected by flow cytometry for 24 h. Data are represented as the mean ± SD. *p<0.05, **p<0.01, ***p<0.001.

Next, we observed the apoptotic promotion of LBP on the two cell lines using flow cytometry. LBP significantly promoted the apoptosis of MDA-MB-468 cells in a time- and dose-dependent manner (Figure 1B). Consistent with the CCK8 results, LBP promoted the apoptosis of MDA-MB-468 cells more strongly than of MDA-MB-231 cells. For example, 12.5 µM LBP induced apoptosis in 53.93 ± 4.18% of MDA-MB-468 cells, which was higher than the apoptosis rate of MDA-MB-231 cells (44.36 ± 1.88%) at 50 µM LBP. The cell cycle test showed that the percentage of the two cell lines in S phase increased greatly after being treated with LBP, from 21.13 ± 0.55% (the control) to 46.07 ± 0.4% (5 µM LBP) for MDA-MB-468 cells, and from 37.73 ± 0.81% (the control) to 69.47 ± 1.19% (25 µM LBP) for MDA-MB-231 cells (Figure 1C), indicating that DNA synthesis was blocked by LBP. The effect of cell cycle arresting by LBP on MDA-MB-468 cells was also greater than on MDA-MB-231 cells. Thus, we concluded that MDA-MB-468 cells were more sensitive to LBP than MDA-MB-231 cells.

Synergistic inhibition efficacy of LBP and GEM on TNBC cells

The combination of anti-cancer drugs can improve treatment effectiveness and reduce drug resistance. Therefore, we combined LBP with the common anticancer chemotherapy drugs, doxorubicin, epirubicin, docetaxel, and GEM to explore which drug performed the best synergistic inhibition efficacy with LBP. We first measured the inhibitory effect on cell growth of these drugs and plotted a curve to calculate the 25, 50, and 75% inhibitory concentrations (IC25, IC50, and IC75) using Prism 7.0 software (Figure 2, Tables 1 and 2). Then, we combined LBP with different drugs at the IC25, IC50, and IC75, and calculated CI values using CompuSyn software to detect whether there was a synergistic effect. The CI values are shown in Tables 3 and 4. A CI value less than 0.9 indicated a synergistic effect between the two drugs. The smaller the CI value, the more significant the synergistic effect was. LBP plus GEM had a remarkable synergistic effect on MDA-MB-468 and MDA-MB-231 cells and the CI value was significantly lower than that of other drug combinations. When 15 µM of LBP plus 0.38 µM GEM (IC25) were added to MDA-MB-468 cells, the CI value was ∼0.33, which was the lowest CI value (Table 3), thus, indicating the strongest synergy. Moreover, when LBP was combined with 0.25 µM GEM, its anti-proliferation effect was greatly improved. The IC50 reduced by ∼10-fold from 7.68 ± 1.84 µM (LBP alone) to 0.92 ± 1.51 µM (LBP + GEM) (Figure 3A).

Figure 2: 
The inhibition curves of doxorubicin, epirubicin, docetaxel, and GEM for 48 h.
Figure 2:

The inhibition curves of doxorubicin, epirubicin, docetaxel, and GEM for 48 h.

Table 1:

IC25, IC50, and IC75 of chemotherapy drugs on MDA-MB-468.

Treatment IC25/µM IC50/µM IC75/µM
Doxorubicin 0.051 0.22 0.83
Epirubicin 0.063 0.30 1.36
Docetaxel 0.0013 0.0025 0.016
GEM 0.38 1.81 10.7
Table 2:

IC25, IC50, and IC75 of chemotherapy drugs on MDA-MB-231.

Treatment IC25/µm IC50/µm IC75/µm
Doxorubicin 0.078 0.27 1.13
Epirubicin 0.16 0.59 2.56
Docetaxel 0.005 0.1 12.5
GEM 0.22 4.5
Table 3:

CI values of LBP combined with other chemotherapy drugs on MDA-MB-468.

Chemotherapy drug LBP, µM
10 12.5 15
IC25 Doxorubicin 0.75 0.88 1
Epirubicin 0.85 >1 >1
Docetaxel 0.85 0.76 0.85
GEM 0.72 0.56 0.33
IC50 Doxorubicin 0.75 0.86 0.96
Epirubicin 0.68 0.77 0.86
Docetaxel 0.94 0.75 0.82
GEM 0.68 0.54 0.38
IC75 Doxorubicin 0.69 0.74 0.84
Epirubicin 0.75 0.88 0.95
Docetaxel >1 0.83 0.87
GEM 0.79 0.65 0.44
Table 4:

CI values of LBP combined with other chemotherapy drugs on MDA-MB-231.

Chemotherapy drug LBP, µM
30 50 75
IC25 Doxorubicin 0.82 0.94 0.88
Epirubicin 0.64 0.44 0.23
Docetaxel 0.87 0.88 0.97
GEM 0.76 0.71
IC50 Doxorubicin 0.8 0.81 0.82
Epirubicin 0.45 0.25 0.23
Docetaxel 0.87 0.87 0.96
GEM 0.2 0.31
IC75 Doxorubicin 0.6 0.66 0.82
Epirubicin 0.21 0.24 0.26
Docetaxel 0.87 >1 >1
Figure 3: 
The synergistic effects of LBP and GEM on MDA-MB-468 cells for 48 h. (A) The IC50 of LBP alone or in combination with 0.25 µM GEM. (B) Cell morphology changes observed using light microscopy with a 10 × objective. (C) The proportion of apoptosis determined by flow cytometry. (D) Distribution of different phrases in the cell cycle detected by flow cytometry. The data are represented as the mean ± SD. *p<0.05, **p<0.01, ***p<0.001, nsp>0.05.
Figure 3:

The synergistic effects of LBP and GEM on MDA-MB-468 cells for 48 h. (A) The IC50 of LBP alone or in combination with 0.25 µM GEM. (B) Cell morphology changes observed using light microscopy with a 10 × objective. (C) The proportion of apoptosis determined by flow cytometry. (D) Distribution of different phrases in the cell cycle detected by flow cytometry. The data are represented as the mean ± SD. *p<0.05, **p<0.01, ***p<0.001, nsp>0.05.

In the following study, concentrations of 2.5 or 5 µM LBP combined with 0.25 µM GEM were used to investigate the synergistic effects. Under the microscope, we observed that the cell morphology was damaged more significantly by the LBP and GEM combination, compared with that of LBP alone (Figure 3B). In addition, LBP + GEM effectively increased apoptosis in a dose- and time-dependent manner (Figure 3C). The apoptosis rate of LBP + GEM at (2.5 + 0.25) µM and (5 + 0.25) µM for 48 h was 22.04 ± 1.93% and 48.37 ± 5.2%, respectively, and for 72 h was 37.93 ± 0.59% and 80.2 ± 6.72%, respectively, which was higher than cells treated with LBP or GEM alone. 2.5 or 5 µM LBP or 0.25 µM GEM alone for 48 h was 6.21 ± 1.40%, 11.23 ± 5.65%, and 13.73 ± 1.29%, respectively, and for 72 h was 11.66 ± 1.24%, 51.63 ± 9.27% and 9.57 ± 0.71%, respectively. Using the Bliss Independence Model, we observed that LBP and GEM had an excellent synergistic effect to promote apoptosis. The apoptosis rate predicted by the model for 2.5 and 5 µM LBP plus GEM for 48 h was 19.09 ± 1.48% and 23.41 ± 4.34%, respectively, and for 72 h was 20.11 ± 1.11% and 56.26 ± 7.26%, which were significantly lower than the corresponding observed values (p<0.05). However, LBP in combination with 0.25 µM GEM did not enhance its arresting effect on the cell cycle (Figure 3D).

Effects of LBP and GEM on TNBC cell death

Levels of γ-H2AX produced by the phosphorylation of H2AX can reflect the degree of DNA damage. To observe DNA damage induced by GEM, LBP, or combinations thereof, the fluorescence intensity of MDA-MB-468 cells labeled with FITC-γ-H2A antibodies was detected using confocal laser scanning microscopy (Figure 4A, B). The results revealed that the fluorescence of cells treated with 0.25 µM GEM alone was only marginally more intense than the control. The fluorescence intensity of cells treated with 2.5 or 5 µM LBP was much higher than that of the control. However, the difference between treatments of LBP alone and in combination with GEM was not observed. Those results indicated that LBP could cause nuclear damage to MDA-MB-468 cells, but no synergistic effect with GEM was observed at GEM concentrations of up to 0.25 µM.

Figure 4: 
Effects of LBP and GEM on MDA-MB-468 cell death. (A) Image of the nucleus obtained using a confocal microscope with a 60 × objective for 24 h. FITC-γ-H2A antibodies were used to label DNA damage, while DAPI was used to label nuclei. (B) The mean fluorescence intensity of γ-H2AX for each group analyzed by ImageJ software. (C) Detection of cleaved PARP and Caspase3 by western blotting in TNBC cells treated with LBP alone. (D) Detection of cleaved PARP and Caspase3 in MDA-MB-468 cells treated with LBP plus GEM. (E) Detection of dCK by western blotting in MDA-MB-468 cells treated with GEM, LBP, or in combination. The data are represented as the mean ± SD. *p<0.05, **p<0.01, ***p<0.001, nsp>0.05.
Figure 4:

Effects of LBP and GEM on MDA-MB-468 cell death. (A) Image of the nucleus obtained using a confocal microscope with a 60 × objective for 24 h. FITC-γ-H2A antibodies were used to label DNA damage, while DAPI was used to label nuclei. (B) The mean fluorescence intensity of γ-H2AX for each group analyzed by ImageJ software. (C) Detection of cleaved PARP and Caspase3 by western blotting in TNBC cells treated with LBP alone. (D) Detection of cleaved PARP and Caspase3 in MDA-MB-468 cells treated with LBP plus GEM. (E) Detection of dCK by western blotting in MDA-MB-468 cells treated with GEM, LBP, or in combination. The data are represented as the mean ± SD. *p<0.05, **p<0.01, ***p<0.001, nsp>0.05.

To investigate the mechanism of LBP on cell death, the apoptotic proteins were detected by western blotting. After MDA-MB-468 cells were treated with LBP alone at 12.5 or 25 µM, cleavage-Caspase3 and cleavage-PARP were visible and in a dose-and time-dependent manner while that was not observed in MDA-MB-231 cells treated with 50 or 75 µM LBP (Figure 4C). Thus, LBP alone promoted the apoptosis of MDA-MB-468 cells through the classical apoptosis pathway. While adding 0.25 µM GEM to LBP, the level of cleavage-Caspase3 and cleavage-PARP was not elevated (Figure 4D), indicating that 0.25 µM GEM enhanced the apoptosis-promoting effect of LBP on MDA-MB-468 cells probably not through the classical apoptotic pathway. Deoxycytidine kinase (dCK) can convert GEM to GEM monophosphate for subsequent active metabolites, which are incorporated into DNA and block DNA synthesis. dCK deficiency is frequently described as one of the important causes of tumor resistance to GEM in vitro or in vivo [10]. Besides, cisplatin was found to be able to increase dCK activity in HCT116 cells [26]. Therefore, we hypothesized whether LBP also improved the activity of dCK to make GEM play a better role in promoting MDA-MB-468 cell death so that they showed synergistic in promoting cell death. Using western blotting, we found that dCK protein levels of MDA-MB-468 cells could be upregulated by LBP treatment (Figure 4E), which might explain why there was such a synergy between LBP and GEM.

Effects of LBP and GEM in TNBC xenograft mouse models

The xenograft mouse model of MDA-MB-468 was established to test the in vivo anti-tumor effect of LBP alone or in combination with GEM. When the tumor grew to 80–100 mm3, the mice were randomly divided into four groups (n=4), the Control group, the GEM (20 mg/kg) group, the LBP (6 mg/kg) group, and the LBP + GEM (6 mg/kg + 20 mg/kg) group, of which the regimen-administration schedule is as the below (Figure 5A). Because the MDA-MB-468 model exhibited weak tumorigenesis and slow growth, the treatment lasted ∼6 weeks. We observed that mice in three drug-treated groups lost weight to varying degrees following each dose, but regained the lost weight within several days (Figure 5B). At the end of the treatment, significant differences in tumor size were observed between the control group (567.6 ± 126.2 mm3) and LBP group (302.7 ± 131.6 mm3) (p<0.05), which indicated that 6 mg/kg LBP could effectively inhibit tumor growth in vivo. GEM treatment alone (529.5 ± 97.7 mm3) had a weak inhibitory effect on the growth of tumors in vivo compared with the control group (p>0.05). When LBP was combined with GEM, the synergistic effect was slight and not statistically significant (302.7 ± 131.6 mm3 in the LBP group vs. 207.7 ± 83.94 mm3 in the LBP + GEM group; p>0.05). Similar results were also observed for tumor weight (Figure 5C–E). Thus, we concluded that LBP alone had an excellent ability to inhibit the growth of MDA-MB-468 cells in vivo, but the effect was not significantly enhanced when combined with GEM. Routine blood tests showed that there was no significant difference between the treated groups and the control group (Table 5), indicating that the dosing schedule was tolerable and safe for mice.

Figure 5: 
Effects of LBP and GEM in TNBC xenograft mouse models. (A) The regimen-administration schedule: BALB/c tumor-bearing nude mice were randomly divided into four groups (n=4), the control group, the GEM (20 mg/kg) group, the LBP (6 mg/kg) group, and the LBP + GEM (6 mg/kg + 20 mg/kg) group. Each group was injected once a week for 6 treatment cycles. (B) The curves of body weights during administration. (C) The curves of tumor growth between groups. (D) Image of tumors following therapy. (E) Comparisons of tumor weights between groups. The data are represented as the mean ± SD. *p<0.05, **p<0.01, ***p<0.001, nsp>0.05.
Figure 5:

Effects of LBP and GEM in TNBC xenograft mouse models. (A) The regimen-administration schedule: BALB/c tumor-bearing nude mice were randomly divided into four groups (n=4), the control group, the GEM (20 mg/kg) group, the LBP (6 mg/kg) group, and the LBP + GEM (6 mg/kg + 20 mg/kg) group. Each group was injected once a week for 6 treatment cycles. (B) The curves of body weights during administration. (C) The curves of tumor growth between groups. (D) Image of tumors following therapy. (E) Comparisons of tumor weights between groups. The data are represented as the mean ± SD. *p<0.05, **p<0.01, ***p<0.001, nsp>0.05.

Table 5:

Blood routine indexes of mice in different experimental groups.

Parameter Con GEM LBP LBP GEM
WBC (109/L) 2.228 ± 1.898 1.498 ± 0.4251 3.308 ± 2.454 1.575 ± 0.6219
RBC (1012/L) 10.3 ± 1.218 10.04 ± 1.15 9.335 ± 1.545 8.655 ± 1.092
HGB, g/L 161.3 ± 16.28 154.8 ± 13.6 146.8 ± 9.878 141.8 ± 12.53
HCT, % 49.55 ± 5.076 49 ± 4.276 46.83 ± 3.948 44 ± 3.947
MCV, fL 48.23 ± 2.339 48.9 ± 1.744 50.58 ± 4.358 51.03 ± 2.665
MCH, pg 15.7 ± 0.8756 15.45 ± 0.4655 15.93 ± 1.632 16.48 ± 0.789
MCHC (g/L) 325.3 ± 7.588 315.8 ± 4.349 314.3 ± 8.098 322.8 ± 8.539
PLT (109/L) 437.3 ± 446.2 715.8 ± 333.4 798.8 ± 436.9 596.3 ± 190.8
MPV, fL 6.075 ± 0.8846 5.825 ± 0.5058 5.975 ± 0.45 5.8 ± 0.216
PDW 16.33 ± 0.3775 16.28 ± 0.15 16.23 ± 0.3862 16.25 ± 0.05774
  1. Values are described as mean ± SD. Comparison of treated groups and control group: ap<0.05, bp<0.01, cp<0.001, nsp>0.05 (no footnote indicates no significance).

Discussion

TNBC is a highly malignant tumor that readily recurs and metastasizes. Chemotherapy is currently the only systemic treatment option for TNBC. Taxane and anthracycline-based regimens represent the mainstay of TNBC treatments, while in neoadjuvant and metastatic settings, platinum-based chemotherapy has shown promising results [27]. The role of platinum in the treatment of TNBC has been extensively studied [28], [29], [30], [31], and numerous studies, including large randomized clinical trials, have extensively evaluated the benefit of adding cisplatin or carboplatin to standard anthracycline and taxane neoadjuvant chemotherapies (NACT) [4]. However, the first-generation platinum drug, cisplatin, has serious side effects and frequently affects patients’ quality of life, thus, limiting its applicability in clinical settings.

As one of the third-generation platinum antineoplastic drugs, LBP was developed by ASTA (Germany) and has been approved for clinical use in a variety of cancers in China [24]. Some studies indicated that the effect of LBP-based regimens in metastatic breast cancer was better than that of cisplatin after anthracycline and taxane therapy [32]. In this study, we systematically studied the effect of LBP on TNBCs.

We compared the inhibitory effect of cisplatin, carboplatin, oxaliplatin, and LBP in MDA-MB-231 and MDA-MB-468 cells. The inhibitory effect of LBP against TNBC cells was comparable to that of cisplatin but stronger than that of carboplatin and oxaliplatin. The antitumor mechanism of LBP results from the formation of DNA-drug adducts, mainly as GG and AG intra-strand cross-links [33]. In 1995, Eliopoulos reported that LBP may also influence the expression of the c-Myc gene, which is involved in oncogenesis, apoptosis, and cell proliferation [34]. In the current study, LBP could arrest MDA-MB-231 and MDA-MB-468 cells in S phase and promote apoptosis in a dose- and time-dependent manner. In addition, MDA-MB-468 cells were more sensitive to LBP than MDA-MB-231 cells. For example, 5 µM LBP promoted the apoptosis of MDA-MB-468 cells by 40%, while 25 µM LBP was required to achieve the same effect in MDA-MB-231 cells. The cleaved PARP and Caspase3 were observed after treating MDA-MB-468 cells with 12.5 µM LBP, whereas such bands were not observed at LBP concentrations up to 75 µM in MDA-MB-231 cells. MDA-MB-231 and MDA-MB-468 cells were both derived from the pleural effusion of the patient and are basal subtypes, but there were differences in the genetic backgrounds. Some studies have reported that growth factor receptor (EGFR) levels in MDA-MB-468 were higher than that in MDA-MB-231, which resulted in their different sensitivity to different drugs [35], [36], [37]. What’s more, the difference in PARP-1 and AKT expression between two cell lines was also demonstrated to be associated with sensitivity to cisplatin treatment [38, 39]. Perhaps these genetic differences were the factors that MDA-MB-468 cells were more sensitive to LBP than MDA-MB-231 cells. Thus, more attention was given to MDA-MB-468 cells in this study.

The use of combination therapy is a common method to overcome drug resistance and reduce side effects during tumor treatment. To determine if any such treatment regimens could enhance the effect of LBP against TNBC, four common chemotherapy drugs, including doxorubicin, epirubicin, docetaxel, and GEM were combined with LBP at different concentrations and used to treat the above cell lines. GEM and LBP exhibited a good synergistic effect in vitro. In MDA-MB-468 cells, GEM alone exhibited the weakest inhibitory effect on cell proliferation, however, the CI value for the combination of GEM and LBP was the lowest, indicating that the synergistic effect was the best. Adding 0.25 µM GEM reduced the IC50 of LBP by 10 times. In addition, the number of apoptotic cells increased significantly.

GEM is an analogue of cytidine and a prodrug that requires cellular uptake and intracellular phosphorylation. After being transported into cells, GEM is phosphorylated by deoxycytidine kinase (dCK) to form GEM monophosphate (dFdCMP), which is then converted to GEM di-and triphosphate (dFdCDP and dFdCTP, respectively). dFdCTP is an inhibitor of DNA polymerase and is also incorporated into DNA, which leads to the termination of DNA chain elongation. In general, the phosphorylation of GEM by dCK is the rate-limiting step for further phosphorylation to active metabolites. dCK deficiency is frequently described as one of the important causes of tumor resistance to GEM in vitro or in vivo [10]. In addition, substantial research has proved that DNA-damaging agents can increase dCK mRNA expression or promote dCK activity [40]. In 2010, METHAROM also found that cisplatin, but not oxaliplatin, can increase dCK activity in HCT116 cells [26]. We hypothesized that LBP perhaps induced dCK levels increase in MDA-MB-468 cells, which promoted the conversion of more GEM to dFdCTP. Western blot results showed that LBP could indeed improve dCK protein levels.

The inhibitory effect of LBP or combined with GEM on tumor growth in the MDA-MB-468 xenograft model was explored. We found that LBP could significantly inhibit TNBC cell growth in vivo, but its effect was slightly enhanced by GEM at the dose of 20 mg/kg. We hypothesized that this dose of GEM was perhaps not sufficient to bring about the synergistic effect with LBP, because the usual dose of GEM in the many xenograft models was 60 mg/kg [41], [42], [43]. Therefore, the optimal combination of LBP and GEM doses should be explored further in future studies.


Corresponding author: Huafeng Pan, General Surgery Department of Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, P. R. China, E-mail: ; and Aiqin Jiang, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, P. R. China, E-mail:

Funding source: Science and Education Integration Project by the Innovation and Entrepreneurship Office of Nanjing university

Award Identifier / Grant number: 0214-1480608207

Acknowledgments

Thanks to Associate Professor Aiqin Jiang for her support with this project.

  1. Research funding: This study was supported by the Science and Education Integration Project by the Innovation and Entrepreneurship Office of Nanjing university (0214–1480608207).

  2. Author contributions: Chengyan Jiang designed and performed the experiments, analyzed data, and drafted the manuscript. Ye Zhang, Xiaoyu Xu, and Shanshan Su performed a part of the cell experiments. Huafeng Pan provided the key experimental materials and analyzed data, and Aiqin Jiang guided the projects and wrote the paper. All authors read and approved the final paper.

  3. Competing interests: The authors declare no potential conflicts of interest.

  4. Informed consent: Informed consent was obtained from all individuals included in this study.

  5. Ethical approval: All mouse experiments were performed with the approval of the Institutional Animal Care and Use Committee of Nanjing University.

References

1. Sung, H, Ferlay, J, Siegel, RL, Laversanne, M, Soerjomataram, I, Jemal, A, et al.. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J Clin 2021;71:209–49. https://doi.org/10.3322/caac.21660.Search in Google Scholar PubMed

2. Siegel, RL, Miller, KD, Jemal, A. Cancer statistics, 2019. CA A Cancer J Clin 2019;69:7–34. https://doi.org/10.3322/caac.21551.Search in Google Scholar PubMed

3. Yin, L, Duan, JJ, Bian, XW, Yu, SC. Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res 2020;22:61. https://doi.org/10.1186/s13058-020-01296-5.Search in Google Scholar PubMed PubMed Central

4. Vagia, E, Mahalingam, D, Cristofanilli, M. The landscape of targeted therapies in TNBC. Cancers 2020;12:916. https://doi.org/10.3390/cancers12040916.Search in Google Scholar PubMed PubMed Central

5. Chang-Qing, Y, Jie, L, Shi-Qi, Z, Kun, Z, Zi-Qian, G, Ran, X, et al.. Recent treatment progress of triple negative breast cancer. Prog Biophys Mol Biol 2020;151:40–53. https://doi.org/10.1016/j.pbiomolbio.2019.11.007.Search in Google Scholar PubMed

6. Dilruba, S, Kalayda, GV. Platinum-based drugs: past, present and future. Cancer Chemother Pharmacol 2016;77:1103–24. https://doi.org/10.1007/s00280-016-2976-z.Search in Google Scholar PubMed

7. McKeage, MJ. Lobaplatin: a new antitumour platinum drug. Expet Opin Invest Drugs 2001;10:119–28. https://doi.org/10.1517/13543784.10.1.119.Search in Google Scholar PubMed

8. Tsvetkova, D, Ivanova, S. Application of approved cisplatin derivatives in combination therapy against different cancer diseases. Molecules 2022;27:2466. https://doi.org/10.3390/molecules27082466.Search in Google Scholar PubMed PubMed Central

9. Engel, JB, Martens, T, Hahne, JC, Häusler, SFM, Krockenberger, M, Segerer, S, et al.. Effects of lobaplatin as a single agent and in combination with TRAIL on the growth of triple-negative p53-mutated breast cancers in vitro. Anti Cancer Drugs 2012;23:426–36. https://doi.org/10.1097/cad.0b013e32834fb8ce.Search in Google Scholar PubMed

10. Mini, E, Nobili, S, Caciagli, B, Landini, I, Mazzei, T. Cellular pharmacology of gemcitabine. Ann Oncol 2006;17:v7–12. https://doi.org/10.1093/annonc/mdj941.Search in Google Scholar PubMed

11. Culine, S, Rebillard, X, Iborra, F, Mottet, N, Faix, A, Ayuso, D, et al.. Gemcitabine and oxaliplatin in advanced transitional cell carcinoma of the urothelium: a pilot study. Anticancer Res 2003;23:1903–6.Search in Google Scholar

12. Joshi, M, Choi, JS, Park, JW, Doh, KO. Combination of doxorubicin with gemcitabine-incorporated G-quadruplex aptamer showed synergistic and selective anticancer effect in breast cancer cells. J Microbiol Biotechnol 2019;29:1799–805. https://doi.org/10.4014/jmb.1907.07029.Search in Google Scholar PubMed

13. Matsuyama, R, Morioka, D, Mori, R, Hiratani, S, Yabushita, Y, Ota, Y, et al.. Single-institution experience with gemcitabine-cisplatin combination therapy as a second-line treatment for patients with unresectable biliary tract cancer after failure of gemcitabine-S-1 combination therapy: a prospective feasibility study. Cancer Chemother Pharmacol 2018;81:949–55. https://doi.org/10.1007/s00280-018-3566-z.Search in Google Scholar PubMed

14. Brinkley, BR, Beall, PT, Wible, LJ, Mace, ML, Turner, DS, Cailleau, RM. Variations in cell form and cytoskeleton in human breast carcinoma cells in vitro. Cancer Res 1980;40:3118–29.Search in Google Scholar

15. Cailleau, R, Young, R, Olivé, M, Reeves, WJJ. Breast tumor cell lines from pleural effusions. J Natl Cancer Inst 1974;53:661–74. https://doi.org/10.1093/jnci/53.3.661.Search in Google Scholar PubMed PubMed Central

16. Siciliano, MJ, Barker, PE, Cailleau, R. Mutually exclusive genetic signatures of human breast tumor cell lines with a common chromosomal marker. Cancer Res 1979;39:919–22.Search in Google Scholar

17. Chou, TC, Talalay, P. Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzym Regul 1984;22:27–55. https://doi.org/10.1016/0065-2571(84)90007-4.Search in Google Scholar PubMed

18. Chou, TC. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res 2010;70:440–6. https://doi.org/10.1158/0008-5472.can-09-1947.Search in Google Scholar PubMed

19. Schmidt, L, Issa, II, Haraldsdóttir, H, Hald, JL, Schmitz, A, Due, H, et al.. Hsp90 inhibition sensitizes DLBCL cells to cisplatin. Cancer Chemother Pharmacol 2022;89:431–40. https://doi.org/10.1007/s00280-022-04407-5.Search in Google Scholar PubMed PubMed Central

20. Xue, L, Chen, F, Yue, F, Camacho, L, Kothapalli, S, Wei, G, et al.. Metformin and an insulin/IGF-1 receptor inhibitor are synergistic in blocking growth of triple-negative breast cancer. Breast Cancer Res Treat 2021;185:73–84. https://doi.org/10.1007/s10549-020-05927-5.Search in Google Scholar PubMed PubMed Central

21. Demidenko, E, Miller, TW. Statistical determination of synergy based on Bliss definition of drugs independence. PLoS One 2019;14:1–22. https://doi.org/10.1371/journal.pone.0224137.Search in Google Scholar PubMed PubMed Central

22. Liu, Q, Yin, X, Languino, LR, Altieri, DC. Evaluation of drug combination effect using a Bliss independence dose-response surface model. Stat Biopharm Res 2018;10:112–22. https://doi.org/10.1080/19466315.2018.1437071.Search in Google Scholar PubMed PubMed Central

23. Bliss, CI. The toxicity of poisons applied jointly. Ann Appl Biol 1939;26:585–615. https://doi.org/10.1111/j.1744-7348.1939.tb06990.x.Search in Google Scholar

24. He, J, Zhang, H. The antitumor effect of lobaplatin against Ishikawa endometrial cancer cells in vitro and in vivo. Biomed Pharmacother 2019;114:108762. https://doi.org/10.1016/j.biopha.2019.108762.Search in Google Scholar PubMed

25. Zhang, H, Chen, R, Yang, S, Liu, W, Li, K, Zhang, H, et al.. Lobaplatin for the treatment of SK-MES-1 lung squamous cell line in vitro and in vivo. OncoTargets Ther 2016;9:4215–24. https://doi.org/10.2147/ott.s108032.Search in Google Scholar

26. Metharom, E, Galettis, P, Manners, S, Links, M. Modulation of gemcitabine accumulation by DNA-damaging agents: mechanisms and specificity in an in vitro model. Anticancer Res 2010;30:3669–73.Search in Google Scholar

27. Lebert, JM, Lester, R, Powell, E, Seal, M, McCarthy, J. Advances in the systemic treatment of triple-negative breast cancer. Curr Oncol 2018;25:S142–50. https://doi.org/10.3747/co.25.3954.Search in Google Scholar PubMed PubMed Central

28. Pandy, JGP, Balolong-Garcia, JC, Cruz-Ordinario, MVB, Que, FVF. Triple negative breast cancer and platinum-based systemic treatment: a meta-analysis and systematic review. BMC Cancer 2019;19:1065. https://doi.org/10.1186/s12885-019-6253-5.Search in Google Scholar PubMed PubMed Central

29. Poggio, F, Bruzzone, M, Ceppi, M, Pondé, NF, La Valle, G, Mastro, LD, et al.. Platinum-based neoadjuvant chemotherapy in triple-negative breast cancer: a systematic review and meta-analysis. Ann Oncol 2018;29:1497–508. https://doi.org/10.1093/annonc/mdy127.Search in Google Scholar PubMed

30. Guo, L, Zhang, L, Guan, Y, Li, Y, Zhang, C, Guo, Q. In vitro studies of H520 cell cycle and apoptosis by anlotinib combined with radiotherapy. Thorac Cancer 2021;12:593–602. https://doi.org/10.1111/1759-7714.13780.Search in Google Scholar PubMed PubMed Central

31. Yu, KD, Ye, FG, He, M, Fan, L, Ma, D, Mo, M, et al.. Effect of adjuvant paclitaxel and carboplatin on survival in women with triple-negative breast cancer: a phase 3 randomized clinical trial. JAMA Oncol 2020;6:1390–6. https://doi.org/10.1001/jamaoncol.2020.2965.Search in Google Scholar PubMed PubMed Central

32. Wang, Z, Xu, L, Wang, H, Li, Z, Lu, L, Li, X, et al.. Lobaplatin-based regimens outperform cisplatin for metastatic breast cancer after anthracyclines and taxanes treatment. Saudi J Biol Sci 2018;25:909–16. https://doi.org/10.1016/j.sjbs.2018.01.011.Search in Google Scholar PubMed PubMed Central

33. Wheate, NJ, Walker, S, Craig, GE, Oun, R. The status of platinum anticancer drugs in the clinic and in clinical trials. Dalton Trans 2010;39:8113–27. https://doi.org/10.1039/c0dt00292e.Search in Google Scholar PubMed

34. Eliopoulos, AG, Kerr, DJ, Maurer, HR, Hilgard, P, Spandidos, DA. Induction of the c-myc but not the cH-ras promoter by platinum compounds. Biochem Pharmacol 1995;50:33–8. https://doi.org/10.1016/0006-2952(95)00085-e.Search in Google Scholar PubMed

35. Cavaliere, A, Sun, S, Lee, S, Bodner, J, Li, Z, Huang, Y, et al.. Development of [(89)Zr]ZrDFO-amivantamab bispecific to EGFR and c-MET for PET imaging of triple-negative breast cancer. Eur J Nucl Med Mol Imag 2021;48:383–94. https://doi.org/10.1007/s00259-020-04978-6.Search in Google Scholar PubMed PubMed Central

36. Zhou, DD, Bai, WQ, Zhai, XT, Sun, LP, Zhen, YS, Li, ZR, et al.. Excellent effects and possible mechanisms of action of a new antibody-drug conjugate against EGFR-positive triple-negative breast cancer. Mil Med Res 2021;8:63. https://doi.org/10.1186/s40779-021-00358-9.Search in Google Scholar PubMed PubMed Central

37. Subik, K, Lee, JF, Baxter, L, Strzepek, T, Costello, D, Crowley, P, et al.. The expression patterns of ER, PR, HER2, CK5/6, EGFR, Ki-67 and AR by immunohistochemical analysis in breast cancer cell lines. Breast Cancer 2010;4:35–41. https://doi.org/10.1177/117822341000400004.Search in Google Scholar

38. Wadhwa, B, Paddar, M, Khan, S, Mir, S, Clarke, AP, Grabowska, AM, et al.. AKT isoforms have discrete expression in triple negative breast cancers and roles in cisplatin sensitivity. Oncotarget 2020;11:4178–94. https://doi.org/10.18632/oncotarget.27746.Search in Google Scholar PubMed PubMed Central

39. Gajan, A, Sarma, A, Kim, S, Gurdziel, K, Wu, GS, Shekhar, MP. Analysis of adaptive olaparib resistance effects on cisplatin sensitivity in triple negative breast cancer cells. Front Oncol 2021;11:694793. https://doi.org/10.3389/fonc.2021.694793.Search in Google Scholar PubMed PubMed Central

40. Giovannetti, E, Mey, V, Danesi, R, Basolo, F, Barachini, S, Deri, M, et al.. Interaction between gemcitabine and topotecan in human non-small-cell lung cancer cells: effects on cell survival, cell cycle and pharmacogenetic profile. Br J Cancer 2005;92:681–9. https://doi.org/10.1038/sj.bjc.6602382.Search in Google Scholar PubMed PubMed Central

41. van Moorsel, CJ, Pinedo, HM, Veerman, G, Vermorken, JB, Postmus, PE, Peters, GJ. Scheduling of gemcitabine and cisplatin in Lewis lung tumour bearing mice. Eur J Cancer 1999;35:808–14. https://doi.org/10.1016/s0959-8049(99)00004-0.Search in Google Scholar PubMed

42. Le, HK, Graham, L, Cha, E, Morales, JK, Manjili, MH, Bear, HD. Gemcitabine directly inhibits myeloid derived suppressor cells in BALB/c mice bearing 4T1 mammary carcinoma and augments expansion of T cells from tumor-bearing mice. Int Immunopharm 2009;9:900–9. https://doi.org/10.1016/j.intimp.2009.03.015.Search in Google Scholar PubMed

43. Sheybani, ND, Witter, AR, Thim, EA, Yagita, H, Bullock, TNJ, Price, RJ. Combination of thermally ablative focused ultrasound with gemcitabine controls breast cancer via adaptive immunity. J Immunother Cancer 2020;8:e001008. https://doi.org/10.1136/jitc-2020-001008.Search in Google Scholar PubMed PubMed Central

Received: 2022-11-22
Accepted: 2023-01-20
Published Online: 2023-02-10

© 2023 the author(s), published by De Gruyter, Berlin/Boston

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 21.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/oncologie-2023-0026/html?lang=en
Scroll to top button