Next Article in Journal
MsHDZ23, a Novel Miscanthus HD-ZIP Transcription Factor, Participates in Tolerance to Multiple Abiotic Stresses
Previous Article in Journal
Mitochondrial Ribosomal Protein MRPS15 Is a Component of Cytosolic Ribosomes and Regulates Translation in Stressed Cardiomyocytes
Previous Article in Special Issue
Leucine Zipper Downregulated in Cancer-1 Interacts with Clathrin Adaptors to Control Epidermal Growth Factor Receptor (EGFR) Internalization and Gefitinib Response in EGFR-Mutated Non-Small Cell Lung Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Efficacy of Cold Atmospheric Plasma vs. Chemotherapy in Triple-Negative Breast Cancer: A Systematic Review

by
Catarina Almeida-Ferreira
1,2,3,4,
Carlos Miguel Marto
1,2,3,5,6,7,8,
Chrislaura Carmo
1,2,9,
Joana Almeida-Ferreira
10,
Cristina Frutuoso
1,2,11,
Maria João Carvalho
1,2,3,11,12,
Maria Filomena Botelho
1,2,3,5,† and
Mafalda Laranjo
1,2,3,5,*,†
1
Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
2
Institute of Biophysics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
3
Center for Innovative Biomedicine and Biotechnology (CIBB), 3000-548 Coimbra, Portugal
4
Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
5
Clinical Academic Center of Coimbra (CACC), University of Coimbra, 3000-354 Coimbra, Portugal
6
Institute of Integrated Clinical Practice, Faculty of Medicine, University of Coimbra, 3000-354 Coimbra, Portugal
7
Laboratory for Evidence-Based Sciences and Precision Dentistry, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
8
Institute of Experimental Pathology, Faculty of Medicine, University of Coimbra, 3000-354 Coimbra, Portugal
9
Coimbra Chemistry Center (CQC), Department of Chemistry, Faculty of Sciences and Technology, University of Coimbra, 3004-535 Coimbra, Portugal
10
University Hospital Center of Central Lisbon, 1150-199 Lisbon, Portugal
11
Gynecology Service, Coimbra Hospital and University Centre, Coimbra Health Local Unit, 3004-561 Coimbra, Portugal
12
Universitary Clinic of Gynecology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2024, 25(6), 3254; https://doi.org/10.3390/ijms25063254
Submission received: 16 January 2024 / Revised: 5 March 2024 / Accepted: 6 March 2024 / Published: 13 March 2024
(This article belongs to the Special Issue Molecular Advances in New Combination Therapies for Cancer)

Abstract

:
Breast cancer is a growing disease, with a high worldwide incidence and mortality rate among women. Among the various types, the treatment of triple-negative breast cancer (TNBC) remains a challenge. Considering the recent advances in cold atmospheric plasma (CAP) cancer research, our goal was to evaluate efficacy data from studies based on chemotherapy and CAP in TNBC cell lines and animal models. A search of the literature was carried out in the PubMed, Web of Science, Cochrane Library, and Embase databases. Of the 10,999 studies, there were fifty-four in vitro studies, three in vivo studies, and two in vitro and in vivo studies included. MDA-MB-231 cells were the most used. MTT, MTS, SRB, annexin-V/propidium iodide, trypan blue, and clonogenic assay were performed to assess efficacy in vitro, increasing the reliability and comprehensiveness of the data. There was found to be a decrease in cell proliferation after both chemotherapy and CAP; however, different protocol settings, including an extensive range of drug doses and CAP exposure times, were reported. For both therapies, a considerable reduction in tumor volume was observed in vivo compared with that of the untreated group. The treatment of TNBC cell lines with CAP proved successful, with apoptosis emerging as the predominant type of cellular death. This systematic review presents a comprehensive overview of the treatment landscape in chemotherapy and CAP regarding their efficacy in TNBC cell lines.

1. Introduction

Cancer is a growing disease worldwide. In 2020, breast cancer (BC) had highest incidence among cancers in women and was responsible for the largest number of deaths across all age groups [1]. The expressions of estrogen receptor-, progesterone receptor-, and human epidermal growth factor receptor-related protein are primary determinants of BC biology. This profile, which is associated with various high-throughput techniques, is used for BC stratification, prognosis, and treatment [2]. Triple-negative breast cancer (TNBC) represents approximately 15–20% of all breast cancer molecular subtypes [3]. It is characterized by the absence of these three types of receptors [4] and tends to have a worse prognosis [5]. Accurate molecular classification of TNBC is crucial for risk stratification [6,7]. Six TNBC molecular subtypes have been proposed, each one with its own features and responses to standard treatment: two different basal-like types (basal-like 1 and basal-like 2), immunomodulatory, mesenchymal, mesenchymal stem-like, and luminal androgen receptors [8,9]. These updates have allowed for personalized treatment with enhanced specificity. Currently, TNBC therapy responds to conventional chemotherapy and monoclonal antibodies, for example, pembrolizumab and avelumab, in the presence of specific biological markers such as programmed death-ligand 1 [10,11]. According to the European Society for Medical Oncology (ESMO) and American Society for Clinical Oncology (ASCO) guidelines for the treatment of TNBC, (neo)adjuvant chemotherapy drugs are used in almost all cases [11,12]. There are different regimens in use, including doxorubicin or epirubicin in combination with cyclophosphamide and paclitaxel or docetaxel in combination with carboplatin. If residual disease or the presence of the BRCA gene mutations is positive, capecitabine or olaparib is also included as an option [11,12]. Unfortunately, TNBC treatment continues to be a challenge, and new approaches are still needed. Some patients present insufficient response, others develop resistance, and treatments are frequently associated with adverse effects [13].
The plasma state, also known as the fourth state of matter, has enough energy to ionize a significant amount of positive and negative particles [14]. There are two types of plasma, namely thermal and non-thermal; the latter is frequently referred to as “cold atmospheric plasma” (CAP). CAP is characterized as non-thermal because the heavy particles are at room temperature. Several methods for producing it have been described [15]. CAP has recently been studied as a potential cancer treatment, with evidence obtained in several malign neoplasms both in vitro and in vivo [16,17,18,19,20]. Some authors have demonstrated CAP selectivity to tumoral cells compared to non-malignant counterparts, highlighting its potential in cancer treatment [21,22,23]. Some mechanisms have been proposed to explain the effects of CAP, namely properties of ultraviolet radiation [24], electric fields that may affect cellular permeabilization by increasing calcium permeability [25], and reactive species that may alter the intracellular redox state, triggering critical cellular responses [20]. Thus, it is crucial to understand the potential of CAP when compared to chemotherapy drugs. Based on the population, intervention, comparison, outcome, and study design (PICOS) criteria, this study aimed to systematically review the literature to determine whether CAP can be as effective as chemotherapy in the treatment of TNBC. Since CAP therapy is not yet a clinically approved treatment, TNBC cell lines and animal models were chosen to compare the cytotoxic effects of CAP with chemotherapeutic agents selected according to ESMO and ASCO guidelines [11,12]. Specifically, doxorubicin, epirubicin, cyclophosphamide, paclitaxel, docetaxel, carboplatin, capecitabine, and olaparib were considered for this study. These chemotherapeutic drugs are the most commonly used in clinical practice. Therefore, the main aim of this systematic review was to answer the PICO question: Is cold atmospheric plasma as effective as chemotherapy in the treatment of triple-negative breast cancer?

2. Methods

This systematic review was developed and reported following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines [26], and its protocol was registered in the International Prospective Register of Systematic Reviews—PROSPERO—with the number CRD42023414394. The research question was built according to the PICO methodology, as described in Table 1.

2.1. Search Strategy

The literature search was performed in four databases, namely Medline (through PubMed), Web of Science (all databases), Embase, and Cochrane Library. The search formulas used for each database are presented in the Supplemental Materials. No restriction on publication date was applied, and the English, Portuguese, Spanish, and French language filters were used. The search was completed on 13 February 2023. A manual search of the reference lists of relevant studies was performed to find additional potentially relevant studies. The search results were imported to the reference management program Mendeley Reference Manager© v2.80.1 (Mendeley Ltd., London, UK), and duplicate results were removed.

2.2. Inclusion and Exclusion Criteria

Two independent reviewers critically assessed the eligibility of studies for inclusion, first by title and abstract and later by evaluating the full text. In case of uncertainty or discrepancies regarding eligibility, a third reviewer was consulted, and a decision was made by consensus. In the eligibility phase, only in vitro and in vivo studies were considered, according to the following inclusion criteria: (1) cell lines and animal models of TNBC; (2) CAP treatment; (3) treatment with chemotherapy drugs selected according to the most recent European and American Society for Medical Oncology Clinical Practice Guidelines (ESMO and ASCO 2021), specifically doxorubicin or epirubicin or cyclophosphamide or paclitaxel or docetaxel or carboplatin or capecitabine or olaparib, and (4) papers whose main goal was to study the cell viability/proliferation of both treatments alone or in combination or in vivo tumor regression, measured as volume or histopathological changes. The exclusion criteria were as follows: (1) other study types, (2) new drugs, (3) resistant cell lines, (4) delivery methods or other formulations, (5) patient samples, (6) non-approved pharmacological combinations, (7) no report of cell viability/proliferation or tumor regression, (8) other experimental models, and (9) other types of studies whose main aim was not to evaluate the efficacy of treatments.

2.3. Data Extraction

For studies that met the inclusion criteria, the following information was collected: (1) authors and year of publication; (2) study model (i.e., triple-negative cell line(s) or animal model(s)), (3) chemotherapy treatment (i.e., chemotherapy agent, concentration drug, periodicity of drug administration and/or how it was carried out), (4) CAP treatment (i.e., plasma source, features of machine as voltage, and exposure times), (5) combination treatments with chemotherapy and CAP, (6) methods for cytotoxicity assay and/or histopathological assessment, and (7) cell viability/proliferation and in vivo tumor regression measurements.

2.4. Quality Assessment

The risk of bias of the in vitro studies was evaluated with Toxicological Data Reliability Assessment Tool (ToxRTool), which provides guidance on assessing the consistency and quality of toxicologic data [27]. The methodological quality of in vivo studies was checked by assessing the risk of bias with the Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE) risk-of-bias tool [28]. Two independent authors evaluated the quality assessment methodology of eligible studies included in this systematic review.

3. Results

3.1. Study Selection

A total of 19,364 studies were obtained for analysis, with 4614 articles from PubMed (4507 regarding chemotherapy drugs plus 107 on CAP treatment), 6308 articles from Web of Science (6016 regarding chemotherapy drugs plus 292 on CAP treatment), 7308 articles from Embase (7219 regarding chemotherapy drugs plus 89 on CAP treatment), and 1134 from Cochrane Library (1107 regarding chemotherapy drugs plus 27 on CAP treatment). Before the screening, duplicate articles (8365) were removed using Web Manager (Clarivate™), leading to 10,999 records. Of these, 10,927 were excluded based on title and abstract analysis, resulting in 72 studies for full-text reading. Despite attempts to obtain all full-text records, only 66 were available to assess eligibility. Of these, seven articles were excluded for not meeting the inclusion criteria (two did not examine any drugs included in our review, one performed a mathematical analysis not encompassed in the inclusion criteria, and four did not evaluate cell viability/proliferation as defined in the PICO strategy). Thus, 59 articles were included in this systematic review for full-text reading and analysis, with publication dates from 1986 to 2023.
The PRISMA flow diagram, which summarizes the study selection performed in this systematic review, is shown in Figure 1.

3.2. Studies’ Characteristics

Table 2 summarizes the main results observed in the studies included. In this systematic review, 59 studies were considered. Of these, 54 were in vitro studies, 3 were in vivo, and 2 articles included in vitro and in vivo experiments. The outcomes regarding the different types of studies are reported separately. We found articles on all chemotherapy drugs considered, as well as CAP treatment. Concerning CAP therapy, we identified two distinct methods: directly administered CAP and plasma-activated media (PAM), an indirect approach consisting of previously exposed solutions. Moreover, a table quantifying the percentage of inhibition induced by the therapies administered was compiled and included in the Supplemental Materials (Table S1).

3.2.1. In Vitro Studies

The TNBC cell line MDA-MB-231 was employed in most of the research. The BT-20, BT-549, CAL51, CAL148, DU4475, HCC1143, HCC1395, HCC1806, HCC1937, HCC28, HCC38, HCC70, Hs578T, MDA-MB-157, MDA-MB-436, MDA-MB-453, MDA-MB-468, MFM223, MUM51, SUM52, SUM102, SUM149, SUM159, and SUM185 cell lines were also used. MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide], MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium], sulforhodamine B (SRB) assay, annexin-V/propidium iodide (PI) for flow cytometry (FC), trypan blue, and clonogenic assay were performed as described in Table 2. Detailed values can be consulted in Table S1.

Chemotherapy

Paclitaxel, docetaxel, doxorubicin, olaparib, cyclophosphamide, and carboplatin concentrations ranged from 0.1 nM to 10,000 nM, from 0.1 nM to 500 nM, from 0.1 µM to 100 µM, from 0.001 µM to 250 µM, from 0.01 µM to 1 µM, and from 1 µM to 20 µM, respectively. The concentrations of capecitabine and epirubicin were not reported for in vitro studies [40,66]. Survival, assessed via the formation of colonies after 14 days, was inhibited by paclitaxel concentrations higher than 5 nM [83]. Cell viability was reduced to 80% with the exposure to 6 µM of doxorubicin for 24 hours [30]. The half-maximal inhibitory concentration (IC50) of doxorubicin was 0.3 µM in MDA-MB-231 and BT-20 cells [47,49]; however, other studies reported different values regarding MDA-MB-231 cells (1 µM [67], 6602 nM [64], 888.75 ± 65.26 nM [73], 45–50 µM and 5–10 µM [74]). The concentration of docetaxel that reduced cell proliferation by 75% in MDA-MB-231 cells was 2 nM when a 24-hour incubation was performed [42]. Cyclophosphamide induced a decrease in cell proliferation at 1 μM. Olaparib demonstrated an IC50 > 100 µM and an IC50 = 18 µM in MDA-MB-231 and MDA-MB-468 cells, respectively [37]. However, a different study in MDA-MB-231 and MDA-MB-468 cells reported values of IC50 = 13.5 µM and IC50 = 5.2 µM, respectively [45]. Concentrations from 1 µM to 10 µM (olaparib) were insufficient to induce significant alterations in the cell viability after 72 hours [78]. Carboplatin showed an IC50 = 10 µM after 24 h, and 1, 4, 8, 10, and 20 µM significantly reduced cell proliferation (MTT assay) [60]. The IC50 of capecitabine in MDA-MB-231 cells was 5150 µM and 2790 µM after incubations of 24 and 72 hours [66]. Epirubicin, in combination with other drugs [40,50], namely paclitaxel, demonstrated an antagonistic effect [50]. Doses of carboplatin lower than 10 µM, and paclitaxel showed additive interactions in MDA-MB-231 cells [50]. Despite the vast number of strategies used for chemotherapy studies, doxorubicin, paclitaxel, and docetaxel seemed to be the most promising drugs, exhibiting greater cell viability reduction at lower concentrations. Paclitaxel was capable of inhibiting 50% of cell viability at lower concentrations (0.07 nM–SRB assay) in contrast to olaparib (13.5 µM–MTT assay), with all IC50 values for the MDA-MB-231 cell line being compared.

CAP Treatment

This section describes the analysis of 19 studies. Regarding PAM treatment, the solutions used were millipore water, cell culture medium, and Ringer’s solution. A decrease of cell proliferation to 20% (p = 0.001) was observed in a volume of 150 µL and 200 µL of PAM (millipore water-based) [72]. The cell viability was reduced to 0.41 and 0.46 in MDA-MB-231 and MDA-MB-468 cells, respectively, compared to control cells after 5 minutes of PAM treatment (medium-based) [79]. The viable cells significantly decreased from 80.50 ± 1.59% to 65.00 ± 3.39% after 120 seconds of CAP treatment in HCC1806 cells [20], while another study demonstrated a reduction of more than 50% in the MDA-MB-231 cell line [52]. Chen et al. reported a reduction of cell viability of approximately 27.4% and 14.7%, respectively, with argon or helium gas flow. CAP decreased cell viability in a dose-dependent manner with statistical significance, as described in Ma et al. [55]. The viability of the MDA-MB-231 cell line decreased to under 40% or 50% after 5 minutes of CAP or PAM exposure, respectively [44]. Furthermore, apoptosis seemed to be the most prevalent type of cell death [20,55,65]. In general, CAP and PAM demonstrated an effective anti-tumoral effect in short exposure times, such as 60 and 120 seconds; however, some authors tested longer exposure times, mainly in PAM treatment.

CAP Treatment and Chemotherapy

Interestingly, CAP treatment and chemotherapy drugs, specifically paclitaxel and olaparib, were combined in two studies [19,51]. Olaparib showed a tendency to improve the efficacy of CAP in all cell lines [51]. Moreover, the chemosensitivity to paclitaxel (0.01 µM) was improved after 15 seconds of CAP exposure. According to clonogenic assay, the combined treatment decreased the number of colonies to a number similar to or even smaller than that specific to PAM or paclitaxel [19].

3.2.2. In Vivo Studies

To obtain animal models, the strains CB-17 of severe combined immunodeficiency mice [56,61,70] and Balb/c were used [79,84] and were inoculated with the TNBC cell line MDA-MB-231.

Chemotherapy

As described by Man et al., cyclophosphamide was administered following two regimens. The group continuously administered 25 mg/kg of cyclophosphamide (low concentration) via drinking water showed an initial reduction of tumor size with no weight loss for 50 days. However, the group of mice treated in cycles of 6 days with 450 mg/kg/cycle (150 mg/kg/injection every other day) demonstrated severe weight loss and death one week after starting the therapy [56]. Munõz et al., demonstrated that 20 mg/kg/day via drinking water starting on the 14th day reduced the volume of tumor compared to the control group. The percentage of necrosis increased from the control group (78%) to the treated group (85%), and no weight loss or other signs of toxicity were observed [61]. Another study demonstrated that cyclophosphamide (20 mg/kg/day) added to capecitabine (100 mg/kg) significantly increased survival compared to the control [70].

CAP Treatment

CAP treatment was administered directly into animal tumors, and PAM injection of PBS showed significant inhibition of tumor growth (p = 0.044 and p = 0.017, respectively). In the comparison of both approaches, the survival of mice in the CAP treatment group was significantly higher than that in the PAM group (p = 4.9 × 10−4). In addition, all control mice died within 27 days, while all mice from the CAP group survived until the end of the experiment (30 days) [84]. Another study showed that the tumor volume was inhibited by PAM injection, and tumor weight dropped considerably after treatment (from 4.053 g to 0.787 g, p = 4.69 × 10−4). No visible adverse effects were observed [79].

3.3. Quality Assessment

In terms of quality assessment, most studies presented an unclear bias, determined by the SYRCLE tool regarding in vivo studies, as described in Figure 2 and detailed in Supplemental Table S2. According to these bias tool guidelines, only three parameters were correctly reported for all studies, specifically, incomplete outcome data, selective outcome reporting, and other sources of bias. Moreover, 20% of articles presented no data on random outcome assessment, as shown in Figure 2. For other studies, the ToxRTool protocol was used, and articles were categorized based on this score. We observed scores between 11 and 14 as the most prevalent, where the studies were considered reliable with restrictions. Although five articles were classified as not reliable, eleven studies were classified under the criterion of reliability without restrictions. The risk assessment for individual study bias is represented in Figure 3 and detailed in Supplemental Table S3.

4. Discussion

The clinical complexity of treating TNBC often requires a tailored approach due to its aggressive nature and poor prognosis compared to other molecular subtypes [85,86,87]. Chemotherapy is a well-established treatment indicated in clinical practice regarding TNBC [10,11]; however, it has associated adverse effects. CAP has been investigated across a vast array of medical fields, specifically in dental medicine, regeneration of tissues, and tumor therapy, without causing significant harm to healthy cells, as well as having antimicrobial effects and an impact on stem cells and nitric oxide levels [88]. It has been explored as a new emerging medical approach for several types of cancer, including TNBC, with promising results in terms of cell death [20,44,75,77]. As CAP is not a clinically approved treatment, being under preclinical research, a comparison was conducted to determine the efficacy of both treatments without associated interventions.
The chemotherapeutic drugs explored—paclitaxel, docetaxel, cyclophosphamide, doxorubicin, olaparib, carboplatin, and capecitabine—were selected based on ESMO and ASCO guidelines [11,12], ensuring a broad and complete assessment of cellular effects [52,79]. Currently, chemotherapy drugs are used in clinical practice and are heavily used as a positive control in most experiments [89,90]. For all treatments, the outcomes depended on the protocol definitions, including the dose of chemotherapy drugs administered and the time of exposure to CAP. Several assays, such as MTT, MTS, SRB, annexin-V/propidium iodide, trypan blue, and clonogenic assay, were performed to prove the in vitro efficacy of CAP and chemotherapy drugs.
We found a wide range of chemotherapy concentrations tested in in vitro assays. The papers showed a reduction in cell proliferation, which is supported by the mechanisms of action of the drugs on tumor cells [91,92,93,94,95]. There were additive interactions between paclitaxel and carboplatin in vitro [50]. In fact, in patients whose TNBC disease progressed after taxane administration, carboplatin was one of the recommended chemotherapy agents [11,12]. According to the guidelines, taxane-anthracycline-based combinations are options for treatment [11,12]. The results obtained corroborated our expectations about the efficacy of chemotherapy in TNBC cell lines. However, the evidence available was often insufficient to statistically compare several studies since the data were not described quantitatively, and the various strategies do not allow for a comparative evaluation.
CAP therapy is an emerging therapeutic approach in cancer although the mechanism of action remains unclear. Recent studies have shown its effects on cell proliferation in several types of cancer, leading to cell death [35,44,96,97,98]. Here, we examine two different strategies concerning the application of CAP in TNBC cell lines, namely CAP and PAM, which used different exposure times and solutions. Both strategies are also reported in preclinical studies regarding other types of cancer [17,18,19,44,99,100,101]. Different types of equipment that are able to generate CAP are described. Some authors used a flow of gases such as argon or helium [34,44,53,62,77], and the frequency and high voltage were also dependent on the equipment of each research group. Generally, the exposure times were 60 seconds or 120 seconds [19,20,21,33,35,44,51,52,53,55,71,80,81]. The reduction of cell viability in TNBC cells was time dependent in all studies. Apoptosis seemed to be the predominant type of cell death [19,20]. Studies on different types of cancer demonstrated a close correlation with the abovementioned, supporting the anti-tumoral potential of CAP as a promising strategy. A vast number of cell lines representing melanoma, brain tumor and leukemia, cervical, breast, colorectal, gastric, lung, ovarian, head and neck, and pancreatic cancers have already been used for CAP studies, demonstrating its anti-proliferative effect [88]. Moreover, some authors reported the selectivity of CAP to tumor cell lines [22,102,103]. The efficacy of olaparib or paclitaxel combined with CAP tended to improve the cytotoxicity of treatment in five distinct TNBC cell lines [51]. PAM improved the chemosensitivity to the lowest concentration of paclitaxel and reduced the metabolic activity compared to isolated therapies [19].
Translating cell line models into animal models is a crucial stage in assessing the efficacy of therapy in a biologically complex organism. In the studies, the methods used were tumor growth or volume, animal weight, or survival monitoring. Cyclophosphamide was able to reduce the volume of tumor with no weight loss and no signs of toxicity [61]. The combination of cyclophosphamide and capecitabine increased survival [70]. Similarly, CAP and PAM inhibited tumor growth significantly, and no noticeable adverse effects were observed [52,84]. CAP was applied locally to the tumor in both plasma approaches. Although we only analyzed two papers with CAP treatment in vivo, the results proved the efficacy in TNBC. In vivo studies with different types of cancer have been performed in recent years [104,105]. Nevertheless, further studies should include cell lines representative of other TNBC molecular subtypes and consider other animal models to encompass as many of the characteristics observed in clinical practice as possible. Unfortunately, in vivo studies regarding CAP and chemotherapy combination are unavailable. In the future, to maximize the anti-tumor potential of CAP, combining it with chemotherapy drugs should also be studied in vivo, promoting a potential coordinated translation to the clinical setting. Furthermore, patient-derived xenograft models could be an enriched research option for evaluating CAP efficacy in human BC tissue samples.
As regards CAP, it could be a promising option for clinical practice. From the results obtained, we hypothesize a reduction of the side effects associated with chemotherapy with a lowering of the concentrations of the drugs administered.
One of the limitations observed was the heterogeneity between the studies due to methodological approaches, namely exposure, intervals and doses of drugs, voltages of equipment, times of exposure, and evaluation times. Moreover, the lack of important information, such as concentrations and detailed and quantitative results, impeded meta-analysis studies. In addition, we observed differences between the number of studies for each drug, which means that the sample included in some groups was reduced, limiting the conclusions. The bias assessment, based on a set of well-defined guidelines, proved the difficulty in obtaining all the information necessary to proceed to the planned statistical study, as illustrated in Figure 2 and Figure 3. CAP studies provided a more detailed description of the conditions of treatment used. Table 3 summarizes key aspects of this systematic review, including the main considerations of the study and the next steps. In the future, based on our results, we suggest creating a list of standard methodologies to address the limitations found. Moreover, correlating studies with human tissue samples of TNBC after CAP treatment with clinical reports of patients will enhance the potential translational value of therapy. Nevertheless, combining chemotherapy and CAP or PAM in vivo appears to be a strategic option.

5. Conclusions

Chemotherapy agents effectively reduced cell proliferation in most TNBC cell lines, depending on a wide range of concentrations and experimenting conditions. CAP treatment successfully treated TNBC cell lines, with apoptosis being the most prevalent type of cell death. Paclitaxel or olaparib combined with CAP in vitro should be further investigated. Our results suggest that other combinations should be considered and evaluated. In the in vivo studies, the selection of different chemotherapeutic regimens influenced the obtained results, with both CAP and PAM effectively reducing tumor volume without visible side effects. However, the lack of information on both treatments and the diversity of experimental conditions underscore the need for more research. Despite the weaknesses observed, our results indicate that CAP is as effective as chemotherapy in TNBC. Innovative studies should be performed to increase the knowledge of CAP treatment and enhance future medical options on TNBC.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ijms25063254/s1.

Author Contributions

Conceptualization, C.A.-F., C.M.M., C.C. and J.A.-F.; methodology, C.A.-F., C.M.M., J.A.-F. and C.C.; software, C.A.-F. and C.M.M.; validation, C.M.M. and M.L.; formal analysis, C.M.M., M.L., C.F. and M.J.C.; investigation, C.A.-F., C.M.M., C.C. and J.A.-F.; resources, C.M.M.; data curation, C.A.-F., C.C. and J.A.-F.; writing—original draft preparation, C.A.-F.; writing—review and editing, C.M.M., M.L., C.F., M.J.C. and M.F.B.; visualization, C.M.M., M.J.C., C.F., M.L. and M.F.B.; supervision, M.L. and M.F.B.; project administration, M.L. and M.F.B.; funding acquisition, C.A.-F. All authors have read and agreed to the published version of the manuscript.

Funding

The authors disclosed receipt of the following financial support for the research, authorship, and publication of this work. FCT (Foundation for Science and Technology) supports the Center for Innovative Biomedicine and Biotechnology (CIBB) through the Strategic Projects UIDB/04539/2020 (https://doi.org/10.54499/UIDB/04539/2020) and UIDP/04539/2020 (https://doi.org/10.54499/UIDP/04539/2020) and the Associated Laboratory funding LA/P/0058/2020 (https://doi.org/10.54499/LA/P/0058/2020). This work was supported by the Project CARBONCT, 2022.03596.PTDC (https://doi.org/10.54499/2022.03596.PTDC). A PhD fellowship granted by FCT supports Catarina Almeida-Ferreira (2022.12228.BD).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Rakha, E.A.; Green, A.R. Molecular classification of breast cancer: What the pathologist needs to know. Pathology 2017, 49, 111–119. [Google Scholar] [CrossRef]
  3. Palomba, G.; Budroni, M.; Olmeo, N.; Atzori, F.; Ionta, M.T.; Pisano, M.; Tanda, F.; Cossu, A.; Palmieri, G. Triple-negative breast cancer frequency and type of BRCA mutation: Clues from Sardinia. Oncol. Lett. 2014, 7, 948–952. [Google Scholar] [CrossRef]
  4. Provenzano, E.; Ulaner, G.A.; Chin, S.-F. Molecular Classification of Breast Cancer. PET Clin. 2018, 13, 325–338. [Google Scholar] [CrossRef]
  5. Cosar, R.; Sut, N.; Ozen, A.; Tastekin, E.; Topaloglu, S.; Cicin, I.; Nurlu, D.; Ozler, T.; Demir, S.; Yıldız, G.; et al. Breast Cancer Subtypes and Prognosis: Answers to Subgroup Classification Questions, Identifying the Worst Subgroup in Our Single-Center Series. Breast Cancer Targets Ther. 2022, 14, 259–280. [Google Scholar] [CrossRef]
  6. Lu, B.; Natarajan, E.; Balaji Raghavendran, H.R.; Markandan, U.D. Molecular Classification, Treatment, and Genetic Biomarkers in Triple-Negative Breast Cancer: A Review. Technol. Cancer Res. Treat. 2023, 22, 15330338221145246. [Google Scholar] [CrossRef]
  7. Sakach, E.; O’Regan, R.; Meisel, J.; Li, X. Molecular Classification of Triple Negative Breast Cancer and the Emergence of Targeted Therapies. Clin. Breast Cancer 2021, 21, 509–520. [Google Scholar] [CrossRef]
  8. Lehmann, B.D.; Jovanović, B.; Chen, X.; Estrada, M.V.; Johnson, K.N.; Shyr, Y.; Moses, H.L.; Sanders, M.E.; Pietenpol, J.A. Refinement of Triple-Negative Breast Cancer Molecular Subtypes: Implications for Neoadjuvant Chemotherapy Selection. PLoS ONE 2016, 11, e0157368. [Google Scholar] [CrossRef]
  9. Lehmann, B.D.; Bauer, J.A.; Chen, X.; Sanders, M.E.; Chakravarthy, A.B.; Shyr, Y.; Pietenpol, J.A. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J. Clin. Investig. 2011, 121, 2750. [Google Scholar] [CrossRef]
  10. Harbeck, N.; Penault-Llorca, F.; Cortes, J.; Gnant, M.; Houssami, N.; Poortmans, P.; Ruddy, K.; Tsang, J.; Cardoso, F. Breast cancer. Nat. Rev. Dis. Primers 2019, 5, 66. [Google Scholar] [CrossRef]
  11. Gennari, A.; André, F.; Barrios, C.H.; Cortés, J.; de Azambuja, E.; DeMichele, A.; Dent, R.; Fenlon, D.; Gligorov, J.; Hurvitz, S.A.; et al. ESMO Clinical Practice Guideline for the diagnosis, staging and treatment of patients with metastatic breast cancer. Ann. Oncol. 2021, 32, 1475–1495. [Google Scholar] [CrossRef]
  12. Korde, L.A.; Somerfield, M.R.; Carey, L.A.; Crews, J.R.; Denduluri, N.; Hwang, E.S.; Khan, S.A.; Loibl, S.; Morris, E.A.; Perez, A.; et al. Neoadjuvant Chemotherapy, Endocrine Therapy, and Targeted Therapy for Breast Cancer: ASCO Guideline. J. Clin. Oncol. 2020, 39, 1485–1505. [Google Scholar] [CrossRef]
  13. Lainetti, P.d.F.; Leis-Filho, A.F.; Laufer-Amorim, R.; Battazza, A.; Fonseca-Alves, C.E. Mechanisms of Resistance to Chemotherapy in Breast Cancer and Possible Targets in Drug Delivery Systems. Pharmaceutics 2020, 12, 1193. [Google Scholar] [CrossRef]
  14. Izadjoo, M.; Zack, S.; Kim, H.; Skiba, J. Medical applications of cold atmospheric plasma: State of the science. J. Wound. Care 2018, 27 (Suppl. S9), S4–S10. [Google Scholar] [CrossRef]
  15. Hoffmann, C.; Berganza, C.; Zhang, J. Cold Atmospheric Plasma: Methods of production and application in dentistry and oncology. Med. Gas Res. 2013, 3, 21. [Google Scholar] [CrossRef]
  16. Ikeda, J.; Tanaka, H.; Ishikawa, K.; Sakakita, H.; Ikehara, Y.; Hori, M. Plasma-activated medium (PAM) kills human cancer-initiating cells. Pathol. Int. 2018, 68, 23–30. [Google Scholar] [CrossRef]
  17. Yan, D.; Sherman, J.H.; Keidar, M. The Application of the Cold Atmospheric Plasma-Activated Solutions in Cancer Treatment. Anticancer Agents Med. Chem. 2018, 18, 769–775. [Google Scholar] [CrossRef]
  18. Ishaq, M.; Kumar, S.; Varinli, H.; Han, Z.J.; Rider, A.E.; Evans, M.D.M.; Murphy, A.B.; Ostrikov, K. Atmospheric gas plasma-induced ROS production activates TNF-ASK1 pathway for the induction of melanoma cancer cell apoptosis. Mol. Biol. Cell 2014, 25, 1523–1531. [Google Scholar] [CrossRef]
  19. Mihai, C.-T.; Mihaila, I.; Pasare, M.A.; Pintilie, R.M.; Ciorpac, M.; Topala, I. Cold Atmospheric Plasma-Activated Media Improve Paclitaxel Efficacy on Breast Cancer Cells in a Combined Treatment Model. Curr. Issues Mol. Biol. 2022, 44, 1995–2014. [Google Scholar] [CrossRef]
  20. Almeida-Ferreira, C.; Silva-Teixeira, R.; Gonçalves, A.C.; Marto, C.M.; Sarmento-Ribeiro, A.B.; Caramelo, F.; Botelho, M.F.; Laranjo, M. Cold Atmospheric Plasma Apoptotic and Oxidative Effects on MCF7 and HCC1806 Human Breast Cancer Cells. Int. J. Mol. Sci. 2022, 23, 1698. [Google Scholar] [CrossRef]
  21. Wang, M.; Holmes, B.; Cheng, X.; Zhu, W.; Keidar, M.; Zhang, L.G. Cold Atmospheric Plasma for Selectively Ablating Metastatic Breast Cancer Cells. PLoS ONE 2013, 8, e73741. [Google Scholar] [CrossRef]
  22. Silva-Teixeira, R.; Laranjo, M.; Lopes, B.; Almeida-Ferreira, C.; Gonçalves, A.C.; Rodrigues, T.; Matafome, P.; Sarmento-Ribeiro, A.B.; Caramelo, F.; Botelho, M.F. Plasma activated media and direct exposition can selectively ablate retinoblastoma cells. Free Radic. Biol. Med. 2021, 171, 302–313. [Google Scholar] [CrossRef]
  23. Guerrero-Preston, R.; Ogawa, T.; Uemura, M.; Shumulinsky, G.; Valle, B.L.; Pirini, F.; Ravi, R.; Sidransky, D.; Keidar, M.; Trink, B. Cold atmospheric plasma treatment selectively targets head and neck squamous cell carcinoma cells. Int. J. Mol. Med. 2014, 34, 941–946. [Google Scholar] [CrossRef]
  24. Keidar, M.; Shashurin, A.; Volotskova, O.; Ann Stepp, M.; Srinivasan, P.; Sandler, A.; Trink, B. Cold atmospheric plasma in cancer therapy. Phys. Plasmas 2013, 20, 057101. [Google Scholar] [CrossRef]
  25. Schneider, C.; Gebhardt, L.; Arndt, S.; Karrer, S.; Zimmermann, J.L.; Fischer, M.J.M.; Bosserhoff, A.-K. Cold atmospheric plasma causes a calcium influx in melanoma cells triggering CAP-induced senescence. Sci. Rep. 2018, 8, 10048. [Google Scholar] [CrossRef]
  26. Liberati, A.; Altman, D.G.; Tetzlaff, J.; Mulrow, C.; Gotzsche, P.C.; Ioannidis, J.P.A.; Clarke, M.; Devereaux, P.J.; Kleijnen, J.; Moher, D. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: Explanation and elaboration. BMJ 2009, 339, b2700. [Google Scholar] [CrossRef]
  27. Schneider, K.; Schwarz, M.; Burkholder, I.; Kopp-Schneider, A.; Edler, L.; Kinsner-Ovaskainen, A.; Hartung, T.; Hoffmann, S. “ToxRTool”, a new tool to assess the reliability of toxicological data. Toxicol. Lett. 2009, 189, 138–144. [Google Scholar] [CrossRef]
  28. Hooijmans, C.R.; Rovers, M.M.; de Vries, R.B.; Leenaars, M.; Ritskes-Hoitinga, M.; Langendam, M.W. SYRCLE’s risk of bias tool for animal studies. BMC Med. Res. Methodol. 2014, 14, 43. [Google Scholar] [CrossRef] [PubMed]
  29. Adachi, Y.; Yoshimura, M.; Nishida, K.; Usuki, H.; Shibata, K.; Hattori, M.; Kondo, N.; Yatabe, Y.; Iwata, H.; Kikumori, T.; et al. Acute phase dynamics of circulating tumor cells after paclitaxel and doxorubicin chemotherapy in breast cancer mouse models. Breast Cancer Res. Treat. 2018, 167, 439–450. [Google Scholar] [CrossRef]
  30. Arun, R.; Dhivya, S.; Abraham, S.K.; Premkumar, K. Low-dose chemotherapeutic drugs induce reactive oxygen species and initiate apoptosis-mediated genomic instability. Toxicol. Res. 2016, 5, 547–556. [Google Scholar] [CrossRef]
  31. Bernhardt, G.; Reile, H.; Birnbiick, H.; Sprufl, T.; Schiinenberger, H. Standardized kinetic microassay to quantify differential chemosensitivity on the basis of proliferative activity. J. Cancer Res. Clin. Oncol. 1992, 118, 35–43. [Google Scholar] [CrossRef]
  32. Blois, J.; Smith, A.; Josephson, L. The slow cell death response when screening chemotherapeutic agents. Cancer Chemother. Pharmacol. 2011, 68, 795–803. [Google Scholar] [CrossRef]
  33. Chen, Z.; Lin, L.; Zheng, Q.; Sherman, J.H.; Canady, J.; Trink, B.; Keidar, M. Micro-Sized Cold Atmospheric Plasma Source for Brain and Breast Cancer Treatment. Plasma Med. 2018, 8, 203–215. [Google Scholar] [CrossRef]
  34. Chen, Z.; Cheng, X.; Lin, L.; Keidar, M. Cold atmospheric plasma discharged in water and its potential use in cancer therapy. J. Phys. D Appl. Phys. 2017, 50, 015208. [Google Scholar] [CrossRef]
  35. Chen, Z.; Zhang, S.; Levchenko, I.; Beilis, I.I.; Keidar, M. In vitro Demonstration of Cancer Inhibiting Properties from Stratified Self-Organized Plasma-Liquid Interface. Sci. Rep. 2017, 7, 12163. [Google Scholar] [CrossRef] [PubMed]
  36. Cheng, X.; Murthy, S.R.K.; Zhuang, T.; Ly, L.; Jones, O.; Basadonna, G.; Keidar, M.; Kanaan, Y.; Canady, J. Canady helios cold plasma induces breast cancer cell death by oxidation of histone mRNA. Int. J. Mol. Sci. 2021, 22, 9578. [Google Scholar] [CrossRef]
  37. Chuang, H.C.; Kapuriya, N.; Kulp, S.K.; Chen, C.S.; Shapiro, C.L. Differential anti-proliferative activities of poly(ADP-ribose) polymerase (PARP) inhibitors in triple-negative breast cancer cells. Breast Cancer Res. Treat. 2012, 134, 649–659. [Google Scholar] [CrossRef]
  38. Di, X.; Shiu, R.P.; Newsham, I.F.; Gewirtz, D.A. Apoptosis, autophagy, accelerated senescence and reactive oxygen in the response of human breast tumor cells to Adriamycin. Biochem. Pharmacol. 2009, 77, 1139–1150. [Google Scholar] [CrossRef]
  39. Frankfurt, O.S.; Krishan, A. Apoptosis-based drug screening and detection of selective toxicity to cancer cells. Anti-Cancer Drugs 2003, 14, 555–561. [Google Scholar] [CrossRef] [PubMed]
  40. Halfter, K.; Hoffmann, O.; Ditsch, N.; Ahne, M.; Arnold, F.; Paepke, S.; Grab, D.; Bauerfeind, I.; Mayer, B. Testing chemotherapy efficacy in HER2 negative breast cancer using patient-derived spheroids. J. Transl. Med. 2016, 14, 112. [Google Scholar] [CrossRef]
  41. Hassan, S.; Esch, A.; Liby, T.; Gray, J.W.; Heiser, L.M. Pathway-enriched gene signature associated with 53BP1 response to PARP inhibition in triple-negative breast cancer. Mol. Cancer Ther. 2017, 16, 2892–2901. [Google Scholar] [CrossRef]
  42. Hernández-Vargas, H.; Palacios, J.; Moreno-Bueno, G. Molecular profiling of docetaxel cytotoxicity in breast cancer cells: Uncoupling of aberrant mitosis and apoptosis. Oncogene 2007, 26, 2902–2913. [Google Scholar] [CrossRef]
  43. Izbicka, E.; Campos, D.; Carrizales, G.; Tolcher, A. Biomarkers for sensitivity to docetaxel and paclitaxel in human tumor cell lines in vitro. Cancer Genom. Proteom. 2005, 2, 219–226. [Google Scholar]
  44. Jezeh, M.A.; Tayebi, T.; Khani, M.R.; Niknejad, H.; Shokri, B. Direct cold atmospheric plasma and plasma-activated medium effects on breast and cervix cancer cells. Plasma Process. Polym. 2020, 17, 1900241. [Google Scholar] [CrossRef]
  45. Keung, M.Y.; Wu, Y.; Badar, F.; Vadgama, J.V. Response of breast cancer cells to PARP inhibitors is independent of BRCA status. J. Clin. Med. 2020, 9, 940. [Google Scholar] [CrossRef] [PubMed]
  46. Kibria, G.; Hatakeyama, H.; Akiyama, K.; Hida, K.; Harashima, H. Comparative Study of the Sensitivities of Cancer Cells to Doxorubicin, and Relationships between the Effect of the Drug-Efflux Pump P-gp. Biol. Pharm. Bull. 2014, 37, 1926–1935. [Google Scholar] [CrossRef]
  47. Kim, R.; Tanabe, K.; Emi, M.; Uchida, Y.; Toge, T. Death receptor-dependent and -independent pathways in anticancer drug-induced apoptosis of breast cancer cells. Oncol. Rep. 2003, 10, 1925–1930. [Google Scholar] [CrossRef]
  48. Koechli, O.R.; Sevin, B.-U.; Perras, J.R.; Chao Chou, T.; Angioli, R.; Steren, A.; Untch, M.; Averette, H.E. Characteristics of the combination paclitaxel plus doxorubicin in breast cancer cell lines analyzed with the ATP-Cell Viability Assay. Breast Cancer Res. Treat. 1993, 28, 21–27. [Google Scholar] [CrossRef]
  49. Koechli, O.R.; Sevin, B.-U.; Perras, J.P.; Angioli, R.; Untch, M.; Steren, A.; Ramachandran, C.; Averette, H.E. Comparative chemosensitivity profiles in three human breast cancer cell lines with the ATP-cell viability assay. Oncology 1994, 51, 552–558. [Google Scholar] [CrossRef]
  50. Konecny, G.; Untch, M.; Slamon, D.; Beryt, M.; Kahlert, S.; Felber, M.; Langer, E.; Lude, S.; Hepp, H.; Pegram, M. Drug interactions and cytotoxic effects of paclitaxel in combination with carboplatin, epirubicin, gemcitabine or vinorelbine in breast cancer cell lines and tumor samples. Breast Cancer Res. Treat. 2001, 67, 223–233. [Google Scholar] [CrossRef]
  51. Lafontaine, J.; Boisvert, J.-S.; Glory, A.; Coulombe, S.; Wong, P. Synergy between non-thermal plasma with radiation therapy and olaparib in a panel of breast cancer cell lines. Cancers 2020, 12, 348. [Google Scholar] [CrossRef]
  52. Liu, Y.; Tan, S.; Zhang, H.; Kong, X.; Ding, L.; Shen, J.; Lan, Y.; Cheng, C.; Zhu, T.; Xia, W. Selective effects of non-thermal atmospheric plasma on triple-negative breast normal and carcinoma cells through different cell signaling pathways. Sci. Rep. 2017, 7, 7980. [Google Scholar] [CrossRef]
  53. Ly, L.; Cheng, X.; Murthy, S.R.K.; Zhuang, T.; Jones, O.Z.; Basadonna, G.; Keidar, M.; Canady, J. Canady cold plasma conversion system treatment: An effective inhibitor of cell viability in breast cancer molecular subtypes. Clin. Plasma Med. 2020, 19–20, 100109. [Google Scholar] [CrossRef]
  54. Ma, J.; Zhang, Q.; Huang, Y.; Xu, Z. Study on inhibitory effect of paclitaxel on MEK and ERK protein overexpression and activation in different breast cancer cell lines. Int. J. Clin. Exp. Med. 2017, 10, 2986–2991. [Google Scholar]
  55. Ma, J.; Yu, K.N.; Zhang, H.; Nie, L.; Cheng, C.; Cui, S.; Chen, G.; Han, W. Non-Thermal Plasma Induces Apoptosis Accompanied by Protective Autophagy via Activating JNK/Sestrin2 Pathway. J. Phys. D Appl. Phys. 2020, 53, 465201. [Google Scholar] [CrossRef]
  56. Man, S.; Bocci, G.; Francia, G.; Green, S.; Jothy, S.; Hanahan, D.; Bohlen, P.; Hicklin, D.J.; Bergers, G.; Kerbel, R.S. Antitumor effects in mice of low-dose (metronomic) cyclophosphamide administered continuously through the drinking water. Cancer Res. 2002, 62, 2731–2735. [Google Scholar] [PubMed]
  57. Mccloskey, D.E.; Kaufmann, S.H.; Prestigiacomo, L.J.; Davidson, N.E. Paclitaxel Induces Programmed Cell Death in MDAMB-468 Human Breast Cancer Cells. Clin. Cancer Res. 1996, 2, 847–854. [Google Scholar] [PubMed]
  58. Merrill, N.M.; Lachacz, E.J.; Vandecan, N.M.; Ulintz, P.J.; Bao, L.; Lloyd, J.P.; Yates, J.A.; Morikawa, A.; Merajver, S.D.; Soellner, M.B. Molecular determinants of drug response in TNBC cell lines. Breast Cancer Res. Treat. 2020, 179, 337–347. [Google Scholar] [CrossRef]
  59. Morse, D.L.; Gray, H.; Payne, C.M.; Gillies, R.J. Docetaxel induces cell death through mitotic catastrophe in human breast cancer cells. Mol. Cancer Ther. 2005, 4, 1495–1504. [Google Scholar] [CrossRef]
  60. Moschetta-Pinheiro, M.G.; Colombo, J.; Tuckumantel, M.S.; Rebolho, G.K.; Zuccari, D.A.P.C. Treatment of Triple Negative Cell Lines with Olaparib to Block DNA Repair. Anticancer Agents Med. Chem. 2022, 22, 2036–2045. [Google Scholar] [CrossRef]
  61. Muñoz, R.; Hileeto, D.; Cruz-Muñoz, W.; Wood, G.A.; Xu, P.; Man, S.; Viloria-Petit, A.; Kerbel, R.S. Suppressive impact of metronomic chemotherapy using UFT and/or cyclophosphamide on mediators of breast cancer dissemination and invasion. PLoS ONE 2019, 14, e0222580. [Google Scholar] [CrossRef]
  62. Ninomiya, K.; Ishijima, T.; Imamura, M.; Yamahara, T.; Enomoto, H.; Takahashi, K.; Tanaka, Y.; Uesugi, Y.; Shimizu, N. Evaluation of extra- and intracellular OH radical generation, cancer cell injury, and apoptosis induced by a non-thermal atmospheric-pressure plasma jet. J. Phys. D Appl. Phys. 2013, 46, 425401. [Google Scholar] [CrossRef]
  63. Norris, R.E.; Adamson, P.C.; Nguyen, V.T.; Fox, E. Preclinical evaluation of the PARP inhibitor, olaparib, in combination with cytotoxic chemotherapy in pediatric solid tumors. Pediatr. Blood Cancer 2014, 61, 145–150. [Google Scholar] [CrossRef]
  64. Oncul, S.; Ercan, A. Discrimination of the Effects of Doxorubicin on Two Different Breast Cancer Cell Lines on Account of Multidrug Resistance and Apoptosis. Indian J. Pharm. Sci. 2017, 79, 599–607. [Google Scholar] [CrossRef]
  65. Park, S.B.; Kim, B.; Bae, H.; Lee, H.; Lee, S.; Choi, E.H.; Kim, S.J. Differential epigenetic effects of atmospheric cold plasma on MCF-7 and MDA-MB-231 breast cancer cells. PLoS ONE 2015, 10, e0129931. [Google Scholar] [CrossRef] [PubMed]
  66. Parrella, A.; Lavorgna, M.; Criscuolo, E.; Russo, C.; Isidori, M. Estrogenic activity and cytotoxicity of six anticancer drugs detected in water systems. Sci. Total Environ. 2014, 485–486, 216–222. [Google Scholar] [CrossRef] [PubMed]
  67. Pilco-Ferreto, N.; Calaf, G.M. Influence of doxorubicin on apoptosis and oxidative stress in breast cancer cell lines. Int. J. Oncol. 2016, 49, 753–762. [Google Scholar] [CrossRef]
  68. Risinger, A.L.; Dybdal-Hargreaves, N.F.; Mooberry, S.L. Breast cancer cell lines exhibit differential sensitivities to microtubule-targeting drugs independent of doubling time. Anticancer Res. 2015, 35, 5845–5850. [Google Scholar] [PubMed]
  69. Sauter, C.; Cogoli, M.; Arrenbrecht, S. Interactions of Cytotoxic and Other Drugs: Rapid Cell Culture Assay. Oncology 1986, 43, 46–49. [Google Scholar] [CrossRef]
  70. Shaked, Y.; Pham, E.; Hariharan, S.; Magidey, K.; Beyar-Katz, O.; Xu, P.; Man, S.; Wu, F.T.H.; Miller, V.; Andrews, D.; et al. Evidence implicating immunological host effects in the efficacy of metronomic low-dose chemotherapy. Cancer Res. 2016, 76, 5983–5993. [Google Scholar] [CrossRef]
  71. Stope, M.B.; Benouahi, R.; Sander, C.; Haralambiev, L.; Nitsch, A.; Egger, E.; Mustea, A. Protherapeutic effects and inactivation of mammary carcinoma cells by a medical argon plasma device. Anticancer Res. 2020, 40, 6205–6212. [Google Scholar] [CrossRef]
  72. Subramanian, P.S.G.; Jain, A.; Shivapuji, A.M.; Sundaresan, N.R.; Dasappa, S.; Rao, L. Plasma-activated water from a dielectric barrier discharge plasma source for the selective treatment of cancer cells. Plasma Process. Polym. 2020, 17, 1900260. [Google Scholar] [CrossRef]
  73. Taherian, A.; Mazoochi, T. Different expression of extracellular signal-regulated kinases (ERK) 1/2 and phospho-Erk proteins in MBA-MB-231 and MCF-7 cells after chemotherapy with doxorubicin or docetaxel. Iran. J. Basic Med. Sci. 2012, 15, 669–677. [Google Scholar] [PubMed]
  74. Tassone, P.; Tagliaferri, P.; Perricelli, A.; Blotta, S.; Quaresima, B.; Martelli, M.L.; Goel, A.; Barbieri, V.; Costanzo, F.; Boland, C.R.; et al. BRCA1 expression modulates chemosensitivity of BRCA1-defective HCC1937 human breast cancer cells. Br. J. Cancer 2003, 88, 1285–1291. [Google Scholar] [CrossRef] [PubMed]
  75. Terefinko, D.; Dzimitrowicz, A.; Bielawska-Pohl, A.; Klimczak, A.; Pohl, P.; Jamroz, P. The influence of cold atmospheric pressure plasma-treated media on the cell viability, motility, and induction of apoptosis in human non-metastatic (Mcf7) and metastatic (mda-mb-231) breast cancer cell lines. Int. J. Mol. Sci. 2021, 22, 3855. [Google Scholar] [CrossRef]
  76. Wali, V.B.; Langdon, C.G.; Held, M.A.; Platt, J.T.; Patwardhan, G.A.; Safonov, A.; Aktas, B.; Pusztai, L.; Stern, D.F.; Hatzis, C. Systematic drug screening identifies tractable targeted combination therapies in triple-negative breast cancer. Cancer Res. 2017, 77, 566–578. [Google Scholar] [CrossRef]
  77. Wang, P.; Zhou, R.; Thomas, P.; Zhao, L.; Zhou, R.; Mandal, S.; Jolly, M.K.; Richard, D.J.; Rehm, B.H.A.; Ostrikov, K.; et al. Epithelial-to-mesenchymal transition enhances cancer cell sensitivity to cytotoxic effects of zcold atmospheric plasmas in breast and bladder cancer systems. Cancers 2021, 13, 2889. [Google Scholar] [CrossRef]
  78. Węsierska-Gądek, J.; Mauritz, M.; Mitulovic, G.; Cupo, M. Differential Potential of Pharmacological PARP Inhibitors for Inhibiting Cell Proliferation and Inducing Apoptosis in Human Breast Cancer Cells. J. Cell Biochem. 2015, 116, 2824–2839. [Google Scholar] [CrossRef]
  79. Xiang, L.; Xu, X.; Zhang, S.; Cai, D.; Dai, X. Cold atmospheric plasma conveys selectivity on triple negative breast cancer cells both in vitro and in vivo. Free Radic. Biol. Med. 2018, 124, 205–213. [Google Scholar] [CrossRef]
  80. Yan, D.; Talbot, A.; Nourmohammadi, N.; Cheng, X.; Canady, J.; Sherman, J.; Keidar, M. Principles of using Cold Atmospheric Plasma Stimulated Media for Cancer Treatment. Sci. Rep. 2015, 5, 18339. [Google Scholar] [CrossRef]
  81. Yan, D.; Cui, H.; Zhu, W.; Nourmohammadi, N.; Milberg, J.; Zhang, L.G.; Sherman, J.H.; Keidar, M. The Specific Vulnerabilities of Cancer Cells to the Cold Atmospheric Plasma-Stimulated Solutions. Sci. Rep. 2017, 7, 4479. [Google Scholar] [CrossRef] [PubMed]
  82. Yaourtis, A.M.; Levina, A.; Lay, P.A. Tumour cell heterogeneity in triple-negative breast cancer cells affects response to cisplatin, but not doxorubicin. J. Inorg. Biochem. 2023, 239, 112082. [Google Scholar] [CrossRef] [PubMed]
  83. Zasadil, L.M.; Andersen, K.A.; Yeum, D.; Rocque, G.B.; Wilke, L.G.; Tevaarwerk, A.J.; Raines, R.T.; Burkard, M.E.; Weaver, B.A. Cytotoxicity of Paclitaxel in Breast Cancer Is due to Chromosome Missegregation on Multipolar Spindles. Sci. Transl. Med. 2014, 6, 229ra43. [Google Scholar] [CrossRef]
  84. Zhou, X.; Cai, D.; Xiao, S.; Ning, M.; Zhou, R.; Zhang, S.; Chen, X.; Ostrikov, K.; Dai, X. Invivopen: A novel plasma source for in vivo cancer treatment. J. Cancer 2020, 11, 2273–2282. [Google Scholar] [CrossRef] [PubMed]
  85. Bosch, A.; Eroles, P.; Zaragoza, R.; Viña, J.R.; Lluch, A. Triple-negative breast cancer: Molecular features, pathogenesis, treatment and current lines of research. Cancer Treat. Rev. 2010, 36, 206–215. [Google Scholar] [CrossRef] [PubMed]
  86. Gregório, A.C.; Lacerda, M.; Figueiredo, P.; Simões, S.; Dias, S.; Moreira, J.N. Therapeutic Implications of the Molecular and Immune Landscape of Triple-Negative Breast Cancer. Pathol. Oncol. Res. 2017, 24, 701–716. [Google Scholar] [CrossRef]
  87. Parsons, H.A.; Beaver, J.A.; Cimino-Mathews, A.; Ali, S.M.; Axilbund, J.; Chu, D.; Connolly, R.M.; Cochran, R.L.; Croessmann, S.; Clark, T.A.; et al. Individualized Molecular Analyses Guide Efforts (IMAGE): A prospective study of molecular profiling of tissue and blood in metastatic triple-negative breast cancer. Clin. Cancer Res. 2017, 23, 379–386. [Google Scholar] [CrossRef] [PubMed]
  88. Braný, D.; Dvorská, D.; Halašová, E.; Škovierová, H. Molecular Sciences Cold Atmospheric Plasma: A Powerful Tool for Modern Medicine. Int. J. Mol. Sci. 2020, 21, 2932. [Google Scholar] [CrossRef] [PubMed]
  89. Li, Y.; Hou, H.; Zhang, P.; Zhang, Z. Co-delivery of doxorubicin and paclitaxel by reduction/pH dual responsive nanocarriers for osteosarcoma therapy. Drug Deliv. 2020, 27, 1044–1053. [Google Scholar] [CrossRef]
  90. Li, H.; Xu, W.; Li, F.; Zeng, R.; Zhang, X.; Wang, X.; Zhao, S.; Weng, J.; Li, Z.; Sun, L. Amplification of anticancer efficacy by co-delivery of doxorubicin and lonidamine with extracellular vesicles. Drug Deliv. 2022, 29, 192–202. [Google Scholar] [CrossRef]
  91. Pazdur, R.; Kudelka, A.P.; Kavanagh, J.J.; Cohen, P.R.; Raber, M.N. The taxoids: Paclitaxel (Taxol®) and docetaxel (Taxotere®). Cancer Treat. Rev. 1993, 19, 351–386. [Google Scholar] [CrossRef]
  92. Goulooze, S.C.; Cohen, A.F.; Rissmann, R. Olaparib. Br. J. Clin. Pharmacol. 2016, 81, 171–173. [Google Scholar] [CrossRef]
  93. de Sousa, G.F.; Wlodarczyk, S.R.; Monteiro, G. Carboplatin: Molecular mechanisms of action associated with chemoresistance. Braz. J. Pharm. Sci. 2014, 50, 693–701. [Google Scholar] [CrossRef]
  94. Mills, K.A.; Chess-Williams, R.; McDermott, C. Novel insights into the mechanism of cyclophosphamide-induced bladder toxicity: Chloroacetaldehyde’s contribution to urothelial dysfunction in vitro. Arch. Toxicol. 2019, 93, 3291–3303. [Google Scholar] [CrossRef]
  95. Walko, C.M.; Lindley, C. Capecitabine: A review. Clin. Ther. 2005, 27, 23–44. [Google Scholar] [CrossRef] [PubMed]
  96. Yan, X.; Xiong, Z.; Zou, F.; Zhao, S.; Lu, X.; Yang, G.; He, G.; Ostrikov, K.K. Plasma-Induced Death of HepG2 Cancer Cells: Intracellular Effects of Reactive Species. In Plasma Processes and Polymers; Wiley: New York, NY, USA, 2012; Volume 9, pp. 59–66. [Google Scholar]
  97. Kumara, M.H.S.R.; Piao, M.J.; Kang, K.A.; Ryu, Y.S.; Park, J.E.; Shilnikova, K.; Jo, J.O.; Mok, Y.S.; Shin, J.H.; Park, Y.; et al. Non-thermal gas plasma-induced endoplasmic reticulum stress mediates apoptosis in human colon cancer cells. Oncol. Rep. 2016, 36, 2268–2274. [Google Scholar] [CrossRef] [PubMed]
  98. Yoshikawa, N.; Nakamura, K.; Kajiyama, H. Current understanding of plasma-activated solutions for potential cancer therapy. Free Radic. Res. 2023, 57, 69–80. [Google Scholar] [CrossRef] [PubMed]
  99. Ahn, H.J.; Kim, K.I.; Hoan, N.N.; Kim, C.H.; Moon, E.; Choi, K.S.; Yang, S.S.; Lee, J.S. Targeting cancer cells with reactive oxygen and nitrogen species generated by atmospheric-pressure air plasma. PLoS ONE 2014, 9, e86173. [Google Scholar] [CrossRef]
  100. Almeida-Ferreira, C.; Silva-Teixeira, R.; Laranjo, M.; Almeida, N.; Brites, G.; Dias-Ferreira, J.; Marques, I.; Neves, R.; Serambeque, B.; Teixo, R.; et al. Open-Air Cold Plasma Device Leads to Selective Tumor Cell Cytotoxicity. Appl. Sci. 2021, 11, 4171. [Google Scholar] [CrossRef]
  101. Tavares-da-Silva, E.; Pereira, E.; Pires, A.S.; Neves, A.R.; Braz-Guilherme, C.; Marques, I.A.; Abrantes, A.M.; Gonçalves, A.C.; Caramelo, F.; Silva-Teixeira, R.; et al. Cold Atmospheric Plasma, a Novel Approach against Bladder Cancer, with Higher Sensitivity for the High-Grade Cell Line. Biology 2021, 10, 41. [Google Scholar] [CrossRef]
  102. Hamouda, I.; Labay, C.; Cvelbar, U.; Ginebra, M.-P.; Canal, C. Selectivity of direct plasma treatment and plasma-conditioned media in bone cancer cell lines. Sci. Rep. 2021, 11, 17521. [Google Scholar] [CrossRef] [PubMed]
  103. Canal, C.; Fontelo, R.; Hamouda, I.; Guillem-Marti, J.; Cvelbar, U.; Ginebra, M.-P. Plasma-induced selectivity in bone cancer cells death. Free Radic. Biol. Med. 2017, 110, 72–80. [Google Scholar] [CrossRef] [PubMed]
  104. Vaquero, J.; Judée, F.; Vallette, M.; Decauchy, H.; Arbelaiz, A.; Aoudjehane, L.; Scatton, O.; Gonzalez-Sanchez, E.; Merabtene, F.; Augustin, J.; et al. Cold-atmospheric plasma induces tumor cell death in preclinical in vivo and in vitro models of human cholangiocarcinoma. Cancers 2020, 12, 1280. [Google Scholar] [CrossRef] [PubMed]
  105. Jung, J.M.; Yoon, H.K.; Kim, S.Y.; Yun, M.R.; Kim, G.H.; Lee, W.J.; Lee, M.W.; Chang, S.E.; Won, C.H. Anticancer Effect of Cold Atmospheric Plasma in Syngeneic Mouse Models of Melanoma and Colon Cancer. Molecules 2023, 28, 4171. [Google Scholar] [CrossRef]
Figure 1. PRISMA flow diagram summarizing study selection in this systematic review.
Figure 1. PRISMA flow diagram summarizing study selection in this systematic review.
Ijms 25 03254 g001
Figure 2. Summary of the quality assessment of the in vivo studies included in the systematic review completed with the SYRCLE tool.
Figure 2. Summary of the quality assessment of the in vivo studies included in the systematic review completed with the SYRCLE tool.
Ijms 25 03254 g002
Figure 3. Score of the quality assessment of the in vitro studies included in the systematic review completed with the ToxRTool [19,20,21,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,57,58,59,60,61,62,63,64,65,66,67,68,69,71,72,73,74,75,76,77,78,79,80,81,82,83]. Articles with scores <11 were represented in red, scores between 11–14 were represented in yellow, and scores between 15–18 were represented in green.
Figure 3. Score of the quality assessment of the in vitro studies included in the systematic review completed with the ToxRTool [19,20,21,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,57,58,59,60,61,62,63,64,65,66,67,68,69,71,72,73,74,75,76,77,78,79,80,81,82,83]. Articles with scores <11 were represented in red, scores between 11–14 were represented in yellow, and scores between 15–18 were represented in green.
Ijms 25 03254 g003
Table 1. Population, intervention, comparison, and outcome (PICO) research strategy used in this systematic review.
Table 1. Population, intervention, comparison, and outcome (PICO) research strategy used in this systematic review.
ParameterDescription
Population (P)Triple-negative breast cancer cell lines and animal models
Intervention (I)Cold atmospheric plasma
Comparison (C)Doxorubicin or epirubicin or cyclophosphamide or paclitaxel or docetaxel or carboplatin or capecitabine or olaparib
Outcome (O)In vitro studies: cell viability/proliferation
In vivo studies: tumor volume and histopathological changes
Table 2. Studies’ details and outcomes.
Table 2. Studies’ details and outcomes.
Author
(Year)
Type of StudyExperimental
Model
Treatment
Characteristics
MethodsMain Results
Adachi et al. (2018)
[29]
In vitroMDA-MB-231PTX (1, 10, 100 nM, and 1 µM) Incubation: 48 h
PTX (100 nM) or DOX (10 µg/mL) for FC
Incubation: 24 h
Viable cells counted with a hemocytometer; Annexin V/PI (FC)The growth of cells in vitro was significantly inhibited by increasing doses of PTX (p < 0.01). Cells treated with PTX (100 nM) for 24 h showed a slight increase in early apoptotic cells. DOX markedly inhibited cells in a dose-dependent manner (p < 0.01). Apoptosis analysis confirmed an apparent increase in early plus late apoptotic cells 24 h after DOX3 treatment.
Almeida-Ferreira et al. (2022)
[20]
In vitroHCC1806CAP. Plasma device: 4 kV, pulses of 1 kHz through a sterilized needle 0.9 mm in radius and 40 mm in length
Exposure time: 60 and 120 s
Incubation: 24 h
Annexin V/PI (FC)The proportion of viable cells significantly decreased from 80.50 ± 1.59% to 64.67 ± 2.16% (p = 0.0008) after 60 s of exposure and to 65.00 ± 3.39% (p = 0.01) after 120 s.
Arun et al. (2016)
[30]
In vitroMDA-MB-231DOX (1, 2, 3, 4, 5, and 6 µM)
Incubation: 12 and 24 h
MTT assayDOX (1 μM) resulted in 90% of cell viability, and DOX (6 μM) resulted in 80% at 12 h. Cell viability was 65% after 24 h when low doses were used.
Bernhardt et al. (1992)
[31]
In vitroMDA-MB-231DOX (1 µM)
Incubation: 0–333 h
Crystal violet assayThe drug was cytotoxic (data were not shown).
Blois et al. (2011)
[32]
In vitroMDA-MB-231PTX (100 mM)
Incubation: 48 h
SRB assayIC50 (nM): 0.07
Chen et al. (2018)
[33]
In vitroMDA-MB-231CAP. Plasma device: 8 kV, 16 kHz. He gas was injected into the quartz tube with a 0.2 L/min flow rate. Micro-sized CAP with stainless-steel tubes 20 mm and 60 mm in length.
Exposure time: 5, 10, 30, 60, and 120 s
Incubation: 24 and 48 h
MTT assayThe cell viability dropped with increasing treatment time for both 20 mm and 60 mm µCAP treatment. For each exposure time, the cell viability of breast cancer cells was lower for the 20 mm CAP length than for the 60 mm CAP length.
Chen et al. (2017)
[34]
In vitroMDA-MB-231PAM. Plasma device: immersed in DI water. The electrodes were connected to a secondary high-voltage transformer (2–5 kV, 30 kHz). Ar, He, and N2 were used, and the flow rate was maintained at about 0.3 min−1.
Exposure gas: Ar, He, and N2
Solutions: DI water and DMEM
Incubation: 24 and 48 h
MTT assayAfter 24 h, cell viability decreased by approximately 27.4% and 14.7% when treated with Ar and He plasma, respectively. Only a slight decrease in cell viability was observed in the case of DI water and N2 plasma solution.
After 48 h, viability decreased approximately 73.1%, 22.8%, 14.1%, and 13.5% when cells were treated with Ar plasma solution, He plasma solution, N2 plasma solution, and DI water, respectively. Thus, the most potent effect was observed in the case of Ar plasma, while the smallest was in N2 plasma.
Chen et al. (2017)
[35]
In vitroMDA-MB-231PAM. Plasma device: 1–3012 V, 40 mA activated with low or high current
Solutions: DI water and DMEM
Exposure time: 12, 24, 36, 48, and 60 s
Incubation: 24 and 48 h
MTT assayThe viability of cells incubated in low-current PAM was generally lower than that of cells incubated in high = current PAM. This was observed both after 24 h (36 s, p > 0.05; 48 s, p < 0.05; 60 s, p < 0.001) and 48 h (36 s, p < 0.01; 48 s, p > 0.05; 60 s, p < 0.05). The cell viability of cells incubated for 48 h at low current steadily decreased with treatment duration, while the cell viability at high current initially decreased and then increased slightly.
Cheng et al. (2021)
[36]
In vitroMDA-MB-231CAP. Plasma device: Canady Helios Cold Plasma™. He flow rate at 3 L/min; 4 kV, 300 kHz, and 40 W
Power settings: 80P (15.7 W), 100P (22.3 W) and 120P (28.7 W)
Exposure time: 3, 5, and 6 min
Incubation: 6, 24, and 48 h
Annexin V/PI (FC)Exposure for 3, 5, or 6 min at 80P or 120P reduced the live cells after 24 and 48 h of incubation.
Chuang et al. (2012)
[37]
In vitroMDA-MB-231, MDA-MB-468, CAL51Olaparib (20, 40, 60, 80, or 100 µM)
Incubation: 72 h
MTT assay;
Clonogenic assay (14–21 days)
IC50 (µM) by MTT
MDA-MB-231: >100; MDA-MB-468: 18; CAL51: 9.5
IC50 (µM) by clonogenic assay
MDA-MB-231: 4.5; MDA-MB-468: 0.2; CAL51: 0.4
Di et al. (2009)
[38]
In vitroMDA-MB-231DOXClonogenic assay (14 days)The mean number of colonies was 2.7 ± 0.9% compared to the control.
Frankfurt & Krishan (2003)
[39]
In vitroMDA-MB-468DOX or PTX
Incubation: 48 h
MTT assay
SRB assay
IC50 (µM) by MTT and SRB assays, respectively.
DOX: 0.05; 0.1
PTX: 0.01; 0.01
Halfter et al. (2016)
[40]
In vitroHCC1143 and HCC1937 spheroidsSingle or combined regimens of CAR, CCP, DOC, EPI, and PTX
Incubation: 96 h
ATP assayMetabolic activity (HCC1143 and HCC1937, respectively)
CAR: 104.71 ± 26.68; 80.95 ± 0.67
DOC: 121.36 ± 45.60; 101.38 ± 2.69
PTX: 81.97 ± 42.22; 93.37 ± 1.71
EPI and CCP: 86.07 ± 24.99; 85.12 ± 5.26
EPI and CCP and DOC: 117.00 ± 23.75; 87.57 ± 5.03
EPI and CCP and PTX: 91.59 ± 33.35; 83.74 ± 10.44
DOC and DOX and CCP: 97.56 ± 6.57; 75.06 ± 2.80
Hassan et al. (2017)
[41]
In vitroMDA-MB-436, MDA-MB-231, MDA-MB-453, MDA-MB-468, HCC1143, HCC1937, HCC1806, HCC1395Olaparib (0.25 nmol/L to 100 mmol/L)
Incubation: 9 days, drugs plus media changed after 4–5 days
Chemosensitivity assayIC50 differed in all cell lines, ranging from 0.003 to 3.8 mmol/L.
Hernández-Vargas et al. (2007)
[42]
In vitroMDA-MB-231DOC (from 0 to 500 nM)
Incubation: 24–96 h
Crystal violet assay;
Annexin-V-FITC Apoptosis detection kit (FC)
Cells were sensitive to nM concentrations of DOC. There was a growth inhibition at concentrations lower than 10 nM.
IC75 (nM): 2 nM
Izbicka et al. (2005)
[43]
In vitroMDA-MB-231DOC (0.1, 0.5, or 5 nM) or PTX (0.1, 1, or 5 nM)
Incubation: 72 and 120 h
MTS assayIC50 (pM)
DOC: 499 (72 h); 35 (120 h)
PTX: 933 (72 h)
PTX toxicity at day five increased about twofold in comparison with day three.
Jezeh et al. (2020)
[44]
In vitroMDA-MB-231CAP or PAM. Plasma device: 20–70 kHz and 5 kV. Gas flow: He or He + 0.5% O2
Exposure time: 1, 2, 3, 4, and 5 min
Incubation: 48 h
PAM: 200 µL of medium
MTT assayCAP: Cell viability was reduced in almost all CAP exposure. Generally, better results were obtained using He + 0.5% O2 than pure gas. The viability of MDA-MB-231 cells decreased by more than 60% after 5 min of treatment.
PAM: Similar results were observed despite no significant differences between He + 0.5% O2 and He. Cell viability decreased to about 50% after 5 min.
Keung et al. (2020)
[45]
In vitroMDA-MB-231, MDA-MB-436, MDA-MB-468, HCC1143, HCC1937, BT-549, HCC70, HCC1806Olaparib (from 0.001 to 200 μM)
Incubation: 7 days
MTT assayIC50 (µM)
MDA-MB-231: 13.5; MDA-MB-436: 4.7; MDA-MB-468: 5.2; HCC1143: 14; HCC1937: 96; BT-549: 81; HCC70: 11; HCC1806: 1.2
Kibria et al. (2014)
[46]
In vitroMDA-MB-231DOX (several concentrations)
Incubation: 24 h with medium changed after 8 h
WST-8 assayEC50: 25.72 ± 20.27 μg/mL
Kim et al. (2003)
[47]
In vitroMDA-MB-231DOX or PTX
Incubation: 48 h
MTT assayIC50 (µM)
DOX: 0.3
PTXl: 0.03
Koechli et al. (1993)
[48]
In vitroBT-20PTX (0.001; 0.002; 0.005; 0.01 PPCs) or DOX (0.1; 0.2; 0.5; 1.0 PPCs) or PTX and DOX (1:1000)ATP cell viability assayIC50 (PPCs):
PTX: 0.00163; DOX: 0.319; PTX and DOX: 0.2277. The CI values ranged from 5.4 to 0.9. At a ratio of 1:10 (PPC), the CI values ranged from 0.4 to 0.5, indicating synergism over the whole range.
Koechli et al. (1994)
[49]
In vitroBT-20DOX or PTX or CCP (0.01, 0.02, 0.05, 0.1, and 0.5 PPCATP cell viability assayIC50 (µM)
DOX: 0.32; PTX: 0.007; CCP: 5.53
Konecny et al. (2001)
[50]
In vitroMDA-MB-231PTX (0.9, 1.8, 3.6, 7.2, 14.5, 29, 58, 116 nM) and CAR (3.1, 6.2, 12.5, 25, 50, 100, 200, 400 µM) or PTX (0.4, 0.9, 1.8, 3.6, 7.2, 14.5, 29, 58, 116 nM and EPI (1.7, 3.3, 6.7, 13.4, 26.8, 53.7, 107, 215, 430 nM)Crystal violet assayCAR doses (<10 μM) showed additive interactions in combination with PTX. However, EPI and PTX demonstrated an antagonistic effect.
Lafontaine et al. (2020)
[51]
In vitroBT549, Hs578T, MDA-MB-157, MDA-MB-231, MDA-MB-468CAP. Plasma device: 10 or 35 W, He and O2 gas flow
Exposure time: 10 to 120 s
Olaparib
Concentration: 2 µM
Incubation: 2 h before CAP
Crystal violet assayCAP: Only 30 s of CAP treatment reached a more intense effect than did other application modes. The efficacy increased with treatment time.
Olaparib: Affected cell growth, especially for MDA-MB-468 (more than 60% of inhibition, p < 0.001).
CAP + olaparib: The combination improved the cytotoxic effect of CAP in all cell lines.
Liu et al. (2017)
[52]
In vitroMDA-MB-231, MDA-MB-453CAP. Plasma device: 10 kV and 5 mA
Exposure time: 60, 90, and 120 s
Incubation: 48 h
Trypan blueThere was significant reduction of cell viability after 60 s in MDA-MB-231, while MDA-MB-453 did not show significant reduction. After 120 s, CAP treatment decreased the viability to <80% MDA-MB-453 and <50% MDA-MB-231.
Ly et al. (2020)
[53]
In vitroMDA-MB-231, Hs578T, HCC1806CAP. Plasma device: Canady Helios Cold Plasma™ Scalpel, 4 kV, He flow rate at 3 L/min and power set to 80, 100, and 120 P.
Exposure time: 1, 2, 3, 4, 5, and 6 min
Incubation: 48 h
MTT assayIncreasing power and treatment duration from 80 to 120 P for 1–6 min yielded a greater viability reduction in MDA-MB-231. A 92–99% decrease in cell viability was achievable after 120 P at 5 or 6 min (p ≤ 0.05). HCC1806 showed the greatest overall CAP resistance.
Ma et al. (2017)
[54]
In vitroHCC1937, BT-549, HCC38PTXAnnexin V/PI (FC) and 7AAD kit (BD559763)PTX increased the number of apoptotic cells.
Ma et al. (2020)
[55]
In vitroMDA-MB-231CAP. Plasma device: 12 kV, 24 kHz. Power density: 0.9 W/cm2. Gas flow: He at 120 L/h
Exposure time: 30, 60, 90, and 120 s
Incubation: 24 h
Cell counting kit-8 kit and Annexin V-FITC/PI (FC)CAP significantly decreased the cell viability in a dose-dependent manner and induced apoptotic cell death.
Man et al. (2002)
[56]
In vivoMDA-MB-231; Female CB-17 SCID mice; Orthotopically implanted into the mammary fat padCCP (25 mg/kg of continuous low doses via drinking water; 450 mg/kg/cycle: 150 mg/kg/injection every other day over six days)Tumor and weight monitoringSix-day therapy cycles were similar to low-dose administration for tumor size reduction. However, the former was extremely toxic to SCID mice, resulting in severe weight loss and death of mice after the first week. No weight loss or other signs of toxicity were observed in the group of SCID mice treated via drinking water.
McCloskey et al. (1996)
[57]
In vitroMDA-MB-468PTX
Incubation: 3, 24, and 120 h
Trypan blueCells exposed for 3 h demonstrated concentration-dependent growth inhibition at ≥10 nM PTX (IC50 = 17 nM). At 24 h, growth inhibition was at 1 nM (IC50 = 2.6 nM). At 120 h, the IC50 was 1.8 nM.
Merrill et al. (2019)
[58]
In vitroMUM51, BT-20, BT-549, CAL148, CAL51, DU4475, HCC1806, HCC1937, HCC38, HCC70, Hs578T, MDA-MB-157, MDA-MB-231, MDA-MB-436, MDA-MB-453, MDA-MB-468, MFM223, SUM52, SUM102, SUM149, SUM159, SUM185PTX (from 0.6 to 10,000 nM) or DOC (from 0.1 to 1000 nM)
Incubation: 72 h
WTS-1 followed by CellTiter-Glo® assayIC50 (nM)
PTX: 110 (MUM51); 159 (BT-20); 110 (BT-549); 4 (CAL148); 310 (CAL51); 19 (DU4475); 77 (HCC1806); 130 (HCC1937); 1700 (HCC38); 3 (HCC70); 150 (Hs578T); 190 (MDA-MB-157); 200 (MDA-MB-231); 110 (MDA-MB-436); 2 (MDA-MB-453); 89 (MDA-MB-468); 4 (MFM223); 9 (SUM102); 13 (SUM149); 2 (SUM159); 10 (SUM185); 3 (SUM52).
DOC: 2 (MUM51); 2 (BT20); 1 (BT549); 2 (CAL148); 4 (CAL51); 5 (DU4475); 4 (HCC1806); 1 (HCC1937); 1 (HCC38); 1 (HCC70); 1 (Hs578T); 1 (MDA-MB-157); 2 (MDA-MB-231); 1 (MDA-MB-436); 1 (MDA-MB-453); 1 (MDA-MB-468); 740 (MFM223); 1 (SUM102); 5 (SUM149); 140 (SUM159); 2 (SUM185); 2 (SUM52).
Mihai et al. (2022)
[19]
In vitroMDA-MB-231 and MDA-MB-231 spheroidsPAM. Plasma device: 10 kV and 28 kHz
Solution: DMEM without FBS (160 µL)
Exposure time: 30 and 60 s
Incubation: 20 min and medium changed to 10% FBS DMEM
PTX (0.1 μM, 0.01 μM, and 0.001 μM) after PAM
Incubation: 24 and 48 h
MTT assay; Clonogenic assay; Spheroid areaPTX: Cell viability was reduced to 63.05% and 28.31% (0.1 μM PTX) after 24 and 48 h, respectively.
PAM: At 48 h, cell viability reduced to approximately 25%.
PTX + PAM: Cell line showed values between 33.77% and 36.28% at 24 h and 18.80% and 19.95% at 48 h.
After 15 s of PAM, the total area of spheroids significantly decreased to 23.81% compared to control (p < 0.05) and 20.95% compared to PTX treatment. Cells were susceptible to PAM and combined treatment. PAM could induce a stable cytotoxic effect and improve PTX chemosensitivity.
Morse et al. (2005)
[59]
In vitroMDA-MB-231DOC (10 nmol/L)
(1) Incubation: 48 h
(2) Incubation: 24 or 48 h or 48 h followed by 24 h in drug-free medium
(3) Incubation: 0, 2, 4, 8, 16, 24, and 48 h
(1) Trypan blue
(2) Crystal violet assay
(3) Annexin V/PI (FC)
(1) Cells had 10% lower viability.
(2) IC50 (24 h): 9.28 × 10−8 (1.63 × 10−9 to 5.28 × 10−6); IC50 (48 h): 5.12 × 10−8 (3.25 × 10−8 to 8.07 × 10−8); IC50 (48 + 24 h): 5.00 × 10−8 (3.43 × 10−8 to 7.29 × 10−8).
(3) The maximal increase of apoptosis was 0.97% (p = 0.39) at 8 h.
Moschetta-Pinheiro et al. (2022)
[60]
In vitroMDA-MB-468CAR (1, 2, 4, 8, 10, and 20 µM)
Incubation: 24 h
MTT assayIC50 (µM): 10
Results showed that within 24 h, all CAR concentrations, except for 2 μM, were able to significantly reduce cell viability when compared to control (p < 0.05).
Munõz et al. (2019)
[61]
In vitro and in vivo(1) MDA-MB-231/LM2-4 (metastatic variant)
(2) Female CB-17 SCID mice; 2 × 106 cells orthotopically implanted into the right inguinal mammary fat pad
CCP
(1) 0.01, 0.05, 0.1, and 1 μM
Incubation: 6 days
(2) 20 mg/Kg/day through the drinking water, initiated on day 14
(1) MTS assay
(2) Tumor growth and volume (mm3) and H&E
(1) 1 μM showed a marked decrease in cell proliferation.
(2) Tumor volume was reduced. The median of necrosis was 78% (70–80% range) for the control group and 85% (80–90% range) for the treated group. In the invasive tumor border, the percentage of necrosis was 16% (0–40% range) for untreated tumors and 40% (20–80% range) for the CCP group. No weight loss or other signs of toxicity were observed.
Ninomiya et al. (2013)
[62]
In vitroMDA-MB-231CAP. Plasma device: 9 kHz, He gas flow
Exposure conditions: 4, 8, 12, 16, or 18 kV for 600 s
Incubation: 24 h
Trypan blueThe half-maximal effective peak-to-peak voltage was 16.7 ± 0.3 kV. Cell viability reduced with the increase in voltage.
Norris et al. (2013)
[63]
In vitroHCC1937Olaparib (0.02–100 μM)
Incubation: 120 h
SRB assayIC50 (μM): ≈100
Oncul et al. (2017)
[64]
In vitroMDA-MB-231DOX (50, 100, 200, 400, 800, 1000, 1500, 2000, 3000, 4000, 8000 nM)
Incubation: 48 h
SRB assay; Annexin V/PI (FC)IC50 (nM): 6602
Cells underwent apoptosis in proportions of 6.75, 15, and 8.25% when treated with 50, 200, and 800 nM of the drug, respectively. Necrotic cells increased by 29% as a response to treatment of 800 nM.
Park et al. (2015)
[65]
In vitroMDA-MB-231CAP
Exposure time: 30 s, 10 times
Cell counting kit-8 and clonogenic assay; Annexin V/PI (FC)Six days after the treatment, CAP reduced the growth rate compared to control. Apoptosis increased from 7.67 to 13.8%.
Parrella et al. (2014)
[66]
In vitroMDA-MB-231CAB or DOX
Incubation: 48 and 72 h
MTT assayIC50 (µM)
CAB: 5150 (24 h) and 2790 (72 h)
DOX: 19 (24 h) and 4 (72 h)
Pilco-Ferreto & Calaf (2016)
[67]
In vitroMDA-MB-231DOX (1, 2, 4, and 8 μM)
Incubation: 24 and 48 h
MTT assayIC50 (μM): 1
The increase in DOX concentration decreased the viability in a time- and dose-dependent manner.
Risinger et al. (2015)
[68]
In vitroMDA-MB-231, MDA-MB-468, BT-549, Hs578T, HCC1937PTX or DOCSRB assayMDA-MB-468 was the most sensitive, and BT-549 and HCC1937 were the most resistant.
Sauter et al. (1986)
[69]
In vitroBT-20CCP or DOXPhase-contrast microscopyCytotoxicity effect was measured by cytopathogenic effect, with no results from CCP. It was 1.7 μM regarding DOX.
Shaked et al. (2016)
[70]
In vivoMDA-MB-231/LM2–4 (metastatic variant); Female CB-17 SCID mice; 2 × 106 cells were orthotopically implanted in the mammary fat pad of 6-week-old femalesCAB (LDM: 100 mg/kg; MTD: 400 mg/kg/day for 4 days followed by a 17-day drug-free break period) or CCP (20 mg/kg/day through the drinking water)SurvivalThere were no significant differences in the mortality between CAB LDM, CAB MTD, and control. Adding CCP to the LDM of CAB significantly increased the survival percentage compared to the control (p = 0.006).
Stope et al. (2020)
[71]
In vitroMDA-MB-231CAP
Exposure time: 5, 20, and 60 s
PAM
Exposure time: 20 and 60 s
Incubation: 4, 24, 48, 72, 96, and 120 h
Cell countingCAP: Effects were seen after 20 s of treatment and 72 h of incubation. More pronounced effects were seen after 60 s. After 60 s, a 4.5-fold growth reduction occurred from 4 to 120 h of incubation.
PAM: The results showed a slightly lower anti-proliferative potential for PAM than for CAP. From 4 to 120 h, cell growth was reduced threefold. At 48 h of incubation, the cell growth reduced significantly from the control at two conditions.
Subramanian et al. (2020)
[72]
In vitroMDA-MB-231PAM. Plasma device: 5 kV, 15 kHz, 6.8 ± 0.6 W
Solution: UP water
Exposure time: 6, 12, and 18 min
Volume: 60, 80, 100, 150, and 200 µL for 6 min
MTT assayCell viability was 81% (p < 0.001), 55% (p < 0.001), and 24% (p < 0.001) after 6, 12, and 18 min, respectively, under a volume of 200 μL. A significant reduction of cell viability was observed only at higher volumes (>100 μL), with 66% (p < 0.01) and 20% (p < 0.001) at 150 and 200 μL, respectively.
Taherian et al. (2012)
[73]
In vitroMDA-MB-231DOX or DOC;
Incubation: 48 h
MTT assayIC50 (nM)
DOX: 887.75 ± 65.26; DOC: 634.58 ± 92.4
Tassone et al. (2003)
[74]
In vitroMDA-MB-231, HCC1937DOX (from 0.1 to 100 µM) or PTX (from 0.01 to 2 µM);
Incubation: 48 h
MTT assay; Annexin V/PI (FC)IC50 (µM)
DOX: 45–50 (HCC1937); 5–10 (MDA-MB-231)
PTX: 2 (HCC1937); 0.01–0.02 (MDA-MB-231)
An apoptotic effect was seen in HCC1937 cells exposed to PTX IC50.
Terefinko et al. (2021)
[75]
In vitroMDA-MB-231PAM. Plasma device: 6 kV, 66.45 kHz, He gas flow rate at 10.6 L/min
Solution: DMEM or Opti-MEM media with or without 3% FBS;
Incubation: 24 and 48 h
Exposure time: 150, 180, 210, and 240 s
Volume: 1.5 or 3 mL
MTT assay; Annexin V/PI (FC)Without FBS: No significant results in cells incubated with DMEM-activated media. However, after 48 h, Opti-MEM-activated media exhibited a great impact on the decrease in cell viability, especially in the treatment times of 180 and 240 s (** p < 0.001, *** p < 0.0004, respectively).
With FBS: DMEM-activated media did not affect cell viability. On the other hand, Opti-MEM-activated media affected cell viability after 180 and 240 s (** p < 0.0014; *** p < 0.0002, respectively) at 24 h. At 48 h, cell viability was reduced in all exposure times (* p < 0.013; ** p < 0.0014; *** p < 0.0002).
Opti-MEM-activated media exhibited the most prominent reduction of the live cell population after the one-day experiment (day 1—from 84.00% to 68.12%, **** p < 0.0001; day 2—from 84.00% to 67.86%, ** p < 0.0015).
Wali et al. (2017)
[76]
In vitroBT-20, MDA-MB-231, MDA-MB-468, BT-549, MDA-MB-436, HCC38PTX (0.03, 0.01, 0.03, 0.1, 1 µM)
Incubation: 72 h
ATP-based CellTiter-Glo® luminescent; Cell viability assayPTX reduced cell viability with increased concentrations in most cell lines. MDA-MB-231 was the less sensitive cell line. The highest concentration ≤ IC50 was 3 nM.
Wang et al. (2013)
[21]
In vitroMDA-MB-231 BRCACAP. Plasma device: 60 V/6 A, He gas flow rate of 4.6 L/min
Incubation: 24, 48, and 120 h
Exposure time: 30, 60, and 90 s
MTS assayAll CAP-treated groups showed significantly inhibited cell proliferation after 3 and 5 days (p < 0.01).
Wang et al. (2021)
[77]
In vitroMDA-MB-231, MDA-MB-468PAM. Plasma device: Ar jet, model kINPen 09, gas flow rate of 5.0 L/m
Exposure time: 10 min
PAM was diluted to different concentrations as designated by the percent remaining (e.g., 70% 10PAM refers to 70% concentration at use)
Incubation: 24 h
Cell viability assayThe cell viability of MDA-MB-468 and MDA-MB-231 cells subjected to 100% PAM was 40.29 ± 6% and 16.02 ± 5.02%, respectively. Treatment did not influence the attached cell numbers of MDA-MB-468 cells but did inhibit the MDA-MB-231 cell line, which indicated MDA-MB-231 cells were more sensitive than were the other cancer lines.
Wesierska-Gadek et al. (2015)
[78]
In vitroBT-20Olaparib (from 1 to 10 µM)
Incubation: 24, 48, and 72 h
CellTiter-Glo® cell viability assay (correlated with ATP levels)There were no significant variations in the number of viable cells or increase of apoptosis.
Xiang et al. (2018)
[79]
In vitro and in vivo(1) MDA-MB-231, MDA-MB-468
(2) Female BALB/c mice; 1 × 106
MDA-MB-231 cells were injected subcutaneously
PAM. Plasma device: 1.0 to 1.4 kV, 8.8 kHz, He gas flow was 1 L/min
Solution: 2 mL of medium;
Exposure time: 1, 2, 3, 4, and 5 min
Incubation: 24 h
(1) Cell counting kit-8; Annexin V/PI (FC)
(2) Tumor growth and weight
(1) Five-minute PAM reduced the viability to 0.41 and 0.46, respectively, for MDA-MB-231 and MDA-MB-468 cells. The relative apoptosis increased on both cell lines compared to healthy cells.
(2) Tumor growth was inhibited, and tumor weight dropped considerably after PAM treatment (from 4.053 g to 0.787 g, p = 4.69 × 10−4). No visible side effects were observed.
Yan et al. (2015)
[80]
In vitroMDA-MB-231PAM. Plasma device: 3.16 kV, 5 W, He gas flow rate was 4.7 L/min
(1) Cell concentrations: 20,000, 40,000, and 80,000 cells/mL
Exposure time: 30, 60, 90, and 120 s
(2) Well number on a plate: 6, 12, 24, and 48
(3) Volume media: 1, 2, 3, and 4 mL
Exposure time: 60 s
Incubation: 72 h
MTT assay(1) The anti-tumor capacity increased as the treatment time exposure increased and decreased as the cell seeding confluence decreased.
(2) One minute after PAM, proliferation decreased as the size of the wells decreased. The effect of treatment was reduced 2/3 in the 48-well plate compared to the 6-well plate.
(3) Relative viability significantly increased as the volume of media increased from 1 to 4 mL.
Yan et al. (2017)
[81]
In vitroMDA-MB-231PAM. Plasma device: 3.16 kV, 30 kHz, He gas flow rate was 4.7 L/min
Solution: Ringer’s solution mixed with DMEM or only medium;
Exposure time: 60 s
Incubation: 20 min, 1, 2, 3, and 4 h
MTT assayRemoving PAM 2, 3, or 4 h after the treatment did not change the effect of PAM on cell viability. When PAM was removed 1 h after treatment, the cytotoxicity was not as severe. The dilution remarkably impacted the anti-cancer capacity of the PAM solutions.
Yaourtis et al. (2023)
[82]
In vitroMDA-MB-231 (spindle and stellar phenotype)DOX (Serial dilutions, from 0 to 10 µM)
Incubation: 72 h
MTT assaySpindle phenotype: IC50 = 0.31 ± 0.05 µM, p > 0.05
Stellar phenotype: IC50 = 0.25 ± 0.05 µM, p > 0.05
Zasadil et al. (2014)
[83]
In vitroMDA-MB-231; CAL51PTX (5, 10, 20, 50, or 100 nM)
Incubation: 24,72 and 120 h
Trypan blue;
Clonogenic assay
Low nanomolar concentrations of PTX caused a decrease in live cell numbers over 120 h. Colony formation was substantially inhibited at concentrations ≥5 nM after 14 days.
Zhou et al. (2020)
[84]
In vivoFemale BALB/c mice; 3 × 106 MDA-MB-231 cells were injected subcutaneously in the right forelimbsCAP or PAM. Plasma device: 5 kV, 8.8 kHz; He gas flow was 0.2 L/min
CAP
Exposure time: 5 min
PAM injection treatment
Solution: 2 mL of PBS
Exposure time: 10 min
It was subcutaneously administered into two slots of the tumor in each mouse at 100 µL/slot.
Treatments were repeated every 72 h until death or the end of study (30 days).
Tumor monitoringAll mice in the control group died within 27 days, and all mice from the CAP direct group survived to the last day. The 30-day survival of mice in the CAP group was significantly higher than that of the PAM group (p = 4.9 × 10−4). Both treatments significantly inhibited tumor growth (CAP: p = 0.044 for CAP; PAM: p = 0.017). However, the growth of the tumors in the PAM group was more suppressed than that in the CAP group.
Ar—argon; ATP—adenosine triphosphate; CAB—capecitabine; CAP—cold atmospheric plasma; CAR—carboplatin; CCP—cyclophosphamide; CI—combination index; DI—deionized; DMEM—Dulbecco’s Modified Eagle Medium; DOC—docetaxel; DOX—doxorubicin; EPI—epirubicin; FC—flow cytometry; He—helium; H&E—hematoxylin and eosin; IC50—half-maximal inhibitory concentration; IC75—concentration to inhibit 75%; LDM—low-dose metronomic; MTD—maximum tolerated dose; MTS—3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium; MTT—3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide; N2—molecular nitrogen; PAM—plasma-activated media; PI—propidium iodide; PPCs—peak plasma concentrations; PTX—paclitaxel; SCID—severe combined immunodeficiency; SRB—sulforhodamine B; WST—water-soluble tetrazolium; UP—ultrapure, *—p < 0.05; **—p < 0.01; ***—p < 0.001; ****—p < 0.0001.
Table 3. Key aspects of this systematic review.
Table 3. Key aspects of this systematic review.
Key AspectHighlights
TNBC(1) Due to its aggressive nature and poor prognosis compared to other molecular subtypes, TNBC requires a broader range of treatment options.
(2) Chemotherapy is a well-established treatment for TNBC, but it comes with associated adverse effects. The selection of chemotherapy drugs, particularly paclitaxel, docetaxel, cyclophosphamide, doxorubicin, olaparib, carboplatin, and capecitabine, was based on ESMO and ASCO guidelines.
Plasma treatmentCold plasma, investigated across various medical fields, including tumor therapy, shows promise in cancer treatment, including TNBC, with results indicating cell death.
Search(1) Initial retrieval of 19,364 studies from four databases.
(2) After screening, 59 articles were included in the systematic review. Articles were published between 1986 and 2023, with focus on the efficacy of chemotherapy drugs and CAP treatment.
In vitro studies(1) In vitro studies demonstrated a reduction in cell proliferation with various chemotherapy concentrations tested, and additive interactions were observed between paclitaxel and carboplatin.
(2) Different strategies, including CAP and PAM, exhibited a time-dependent reduction in TNBC cell viability, with apoptosis being the predominant type of cell death.
(3) Combination therapies involving cold plasma and chemotherapy drugs tended to improve cytotoxicity in TNBC cell lines.
In vivo studiesAnimal models are crucial for assessing therapy efficacy. Studies demonstrated inhibition of tumor growth with no noticeable adverse effects in animal models regarding CAP treatment.
LimitationsHeterogeneity between studies, including methodological approaches and lack of detailed information, poses limitations to meta-analysis studies and conclusive interpretations. This emphasizes the need for standardized methodologies.
Future directions(1) This systematic review underscores the need for further research using standardized methodologies to address current limitations and advance clinical translation.
(2) Studies directly comparing CAP, PAM, and standard chemotherapy regimens should be performed, including the evaluation of cell death and associated mechanisms of action.
(3) Conducting studies to unravel specific protein alterations after CAP and PAM treatment might be a strategy for establishing combinations with drugs used in clinical practice.
(4) Future studies should consider other animal models and explore combination therapies. Cold plasma therapy could potentially reduce chemotherapy-associated side effects by lowering drug concentrations, necessitating further investigation in this field.
(5) The use of patient-derived xenografts must be considered as a key approach to verifying the effects of CAP. The findings should be correlated with patients’ clinical data.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Almeida-Ferreira, C.; Marto, C.M.; Carmo, C.; Almeida-Ferreira, J.; Frutuoso, C.; Carvalho, M.J.; Botelho, M.F.; Laranjo, M. Efficacy of Cold Atmospheric Plasma vs. Chemotherapy in Triple-Negative Breast Cancer: A Systematic Review. Int. J. Mol. Sci. 2024, 25, 3254. https://doi.org/10.3390/ijms25063254

AMA Style

Almeida-Ferreira C, Marto CM, Carmo C, Almeida-Ferreira J, Frutuoso C, Carvalho MJ, Botelho MF, Laranjo M. Efficacy of Cold Atmospheric Plasma vs. Chemotherapy in Triple-Negative Breast Cancer: A Systematic Review. International Journal of Molecular Sciences. 2024; 25(6):3254. https://doi.org/10.3390/ijms25063254

Chicago/Turabian Style

Almeida-Ferreira, Catarina, Carlos Miguel Marto, Chrislaura Carmo, Joana Almeida-Ferreira, Cristina Frutuoso, Maria João Carvalho, Maria Filomena Botelho, and Mafalda Laranjo. 2024. "Efficacy of Cold Atmospheric Plasma vs. Chemotherapy in Triple-Negative Breast Cancer: A Systematic Review" International Journal of Molecular Sciences 25, no. 6: 3254. https://doi.org/10.3390/ijms25063254

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop