Open Access

A novel approach for transforming breast cancer stem cells into endothelial cells

  • Authors:
    • Qi-Qi Mao
    • Xiao-Chun Ji
    • Jia-Nan Zhang
    • Wei-Feng Teng
    • Shao-Cheng Zhou
  • View Affiliations

  • Published online on: December 21, 2023     https://doi.org/10.3892/etm.2023.12362
  • Article Number: 74
  • Copyright: © Mao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tumor vascular endothelial cells play a pivotal in the tumor microenvironment, influencing the proliferation, invasion, and metastasis of tumor progression. The present study investigated a novel method for inducing the transformation of breast cancer stem cells into endothelial cells, providing a cellular model investigating anti‑angiogenic mechanisms in vitro. The breast cancer cell line MCF‑7 was used, and the expression of CD133 was initially detected using flow cytometry. CD133+ breast cancer cells were purified using immunomagnetic bead sorting technology, yielding an MCF‑7CD133+ subpopulation. The proliferation ability of these cells was assessed using an MTT assay, while their microsphere formation ability was evaluated using a microsphere formation assay. Post‑transformation in an optimized endothelial cell culture medium, expression of endothelial cell markers CD31 and CD105 were detected using flow cytometry. Endothelial cell tube formation assays and DiI‑labeled acetylated low‑density lipoprotein (DiI‑Ac‑LDL) assays were employed to analyze the endothelial cell function of the MCF‑7CD133+ cells. MDM2/CEN12 gene amplification was detected through fluorescence in situ hybridization (FISH). The MCF‑7 breast cancer cell line exhibited 1.7±0.3% trace cells expressing the stem cell surface marker CD133. After anti‑CD133 immunomagnetic bead sorting, MCF‑7CD133+ and MCF‑7CD133‑ subpopulation cells were obtained, with CD133 expression rates of 85.6±2.8 and 0.18±0.08%, respectively. MTT assay results demonstrated that, after 7 days, the proliferation rate of MCF‑7CD133+ cells was significantly higher compared with MCF‑7CD133‑ cells. MCF‑7CD133+ subpopulation cells displayed strong stem cell characteristics, growing in suspension in serum‑free media and forming tumor cell spheres. In contrast, MCF‑7CD133‑ cells failed to form microspheres. After culturing cells in endothelial cell differentiation and maintenance media, the percentage of MCF‑7CD133+ cells before and after endothelial cell culture was 0.3±0.16 and 81.4±8.37% for CD31+ cells and 0.2±0.08 and 83.8±7.24% for CD105+ cells, respectively. Vascular‑like structure formation and Ac‑LDL phagocytosis with red fluorescence in the tube formation assays confirmed endothelial cell function in the MCF‑7CD133+ cells. FISH was used to verify MDM2/CEN12 gene amplification in the induced MCF‑7CD133+ cells, indicating tumor cell characteristics. The modified endothelial cell transformation medium effectively induced differentiated tumor stem cells to express vascular endothelial cell markers and exhibit endothelial functions, ideal for in vitro anti‑angiogenesis research.

Introduction

Breast cancer represents a leading cause of cancer-associated mortality in women globally. Despite notable advancements in the early detection and treatment of breast cancer, including improved surgical techniques, chemotherapy, radiation therapy and targeted biological treatments, a subset of patients still experience recurrence and/or metastasis, resulting in the failure of conventional therapeutic strategies (1). Thus, there is an urgent need to understand the etiology of breast cancer and identify innovative therapeutic approaches to address this critical health concern.

The complex nature of breast cancer, characterized by its heterogeneity in molecular profiles, pathological features and response to treatment, underscores the necessity of a deeper understanding (2). Research is increasingly focusing on the genetic and molecular underpinnings of breast cancer, exploring the role of genetic mutations, epigenetic alterations and the tumor microenvironment in cancer progression and resistance to treatment (3). Furthermore, the emerging field of cancer stem cell biology is shedding light on a subset of cells within tumors that possess the ability to self-renew, differentiate and potentially drive tumor growth and metastasis.

In 1983, Mackillop et al (4) first proposed the tumor stem cell hypothesis, suggesting that tumors contain a small subpopulation of cells with stem cell-like properties. The significance of the tumor stem cell hypothesis lies in its implications for cancer treatment and resistance. Cancer stem cells (CSCs) have been implicated in the resilience of malignant tumors to chemotherapeutic agents and radiation therapy (5). Their stem-like properties allow them to survive traditional therapies, such as chemotherapy and radiotherapy, that primarily target rapidly dividing cells. CSCs thrive in hypoxic environments and exhibit high expression levels of free-radical scavenging mechanisms. This adaptive response results in the decreased intracellular accumulation of reactive oxygen species following exposure to radiation, which consequently gives rise to the development of a radioresistant phenotype (6). This survival advantage of CSCs is thought to contribute to the post-treatment recurrence and metastasis of tumors, as these residual CSCs can regenerate the tumor mass and facilitate its spread to distant sites (7-9). CD133, a 5-transmembrane (5_TM) glycoprotein, is a stem cell surface marker widely used as a biomarker in various solid tumors including brain (10), lung (11), gastric (12) and ovarian cancer (13).

The traditional theory of tumor angiogenesis states that tumor neovascularization primarily stems from two processes: Angiogenesis and vasculogenesis (14,15). Angiogenesis involves the sprouting of new blood vessels from pre-existing ones. In the context of tumors, angiogenic factors are released by cancer cells, which then stimulate the neighboring vascular endothelial cells to proliferate and form new vessel branches (16). Vascular endothelial growth factor (VEGF) is one of the most potent inducers of angiogenesis (17). In cancer, VEGF is produced and secreted by tumor cells, which is associated with tumor progression, invasiveness, metastasis and tumor recurrence (18). Fibroblast growth factor-2 (FGF2) exerts its effects on endothelial cells via a paracrine signaling after being released by tumor cells (19) Unlike angiogenesis, vasculogenesis refers to the formation of new blood vessels from endothelial progenitor cells (EPCs) that originate in the bone marrow. These EPCs are mobilized to the tumor site, where they differentiate into endothelial cells and contribute to the neovascular network (20).

In the absence of vascular endothelial cells, tumor stem cells within the tumor tissues can differentiate into vascular endothelial cells, promoting tumor angiogenesis (21,22). This process, a form of neovascularization distinct from traditional angiogenesis and vasculogenesis, involves the direct contribution of tumor stem cells to the tumor vasculature. These cells undergo endothelial differentiation, integrating into the developing vascular structure and thereby supporting the angiogenic process (23). Several studies have highlighted the link between tumor cell proliferation, invasion, metastasis, and angiogenesis (24-27). However, the role of tumor stem cells in inducing the formation of tumor blood vessels is not fully understood.

The present study utilized CD133 as a stem cell surface marker to isolate and purify tumor stem cells from breast cancer cell lines. By enhancing the differentiation of CD133+ breast CSCs into vascular endothelial cells in vitro, this study aimed to establish a basis for studying anti-angiogenic mechanisms.

Materials and methods

Cells and cell culture

MCF-7 (cat. no. SCSP-531) were purchased from the Cell Bank of the Chinese Academy of Sciences and HUVECs (cat. no. iCell-h110) were purchased from Cellverse Bioscience Technology Co., Ltd. The MCF-7 breast cancer cell line was cultured in DMEM supplemented with 10% FBS, 100 µg/ml streptomycin, and 100 U/ml penicillin. HUVECs were maintained in an ECM endothelial cell culture medium supplemented with 5% FBS, 100 µg/ml streptomycin, and 100 U/ml penicillin. HUVECs are known to exhibit senescence after several passages, thus they were revived at cell passage 2 and consistently used at low passages, typically below passage 5. This practice was essential to maintain their physiological relevance and ensure the consistency of the results. Both cell types were incubated at 37˚C in a 5% CO2 incubator. MCF-7 cells exhibiting adherent growth were deemed suitable for experimental use when adherent growth exceeded 80%. Before passaging or cryopreservation, cells were digested with trypsin.

Flow cytometry

Cells were digested using 0.25% trypsin, and digestion was halted by adding culture media. Subsequently, cells were washed with 0.01M PBS, resuspended, and centrifuged at 800 x g for 5 min at room temperature to remove the supernatant. The cells were prepared at a concentration of 1x106 cells in 1 ml of 0.01 M PBS. Subsequently, 100 µl cell suspension was added into 5 ml flow tubes with phycoerythrin-labeled CD133 (1:20; cat. no. 12-1338-42; Thermo Fisher Scientific, Inc.), CD31 (1:40; cat. no. 12-0319-42; Thermo Fisher Scientific, Inc.), or CD105 antibodies (1:40; cat. no. MHCD10504; Thermo Fisher Scientific, Inc.). The mixture was thoroughly mixed and incubated at 4˚C in the dark for 10 min. Following incubation, cells were washed, resuspended, centrifuged at 800 x g for 5 min at 4˚C, and the supernatant was removed. Finally, cells were resuspended in 500 µl 0.01 M PBS, flow data were collected using BD FACSCanto II (Becton, Dickinson and Company) and analyzed using FLOWJO version 7.6.2 (flowjo.com/).

Isolation of CD133-positive cells

After culturing MCF-7 cells, they were digested and washed. Subsequently, cells were resuspended in 300 µl sorting buffer and combined with 100 µl anti-CD133 immunomagnetic beads (cat. no. 130-097-049; Miltenyi Biotec GmbH). The mixture was incubated at 4˚C in the dark for 30 min. After incubation, cells were washed with 0.01 M PBS and centrifuged at 800 x g for 10 min at 4˚C. The supernatant was discarded, and the cell pellet was resuspended in a sorting buffer. A pre-rinsed separation column with 500 µl sorting buffer was then loaded with the cell suspension and washed thrice with 0.01 M PBS. CD133+ cells were eluted by washing the column with 1 ml sorting buffer using a syringe pump.

MTT assay

The MCF-7 cells were divided into two subpopulations, MCF-7CD133+ and MCF-7CD133-, and seeded into 96-well plates. Each well was filled with 0.2 ml serum-free media supplemented with 20 ng/ml basic fibroblast growth factor (bFGF; cat. no. AF-100-18B-500UG; Thermo Fisher Scientific, Inc.) and 20 ng/ml epidermal growth factor (EGF; cat. no. AF-100-15-500UG; Thermo Fisher Scientific, Inc.). The cells were cultured for 7 days, with the addition of 20 µl MTT reagent to each well daily. After 4 h of further incubation, the medium was removed and replaced with 150 µl DMSO. The absorbance at 490 nm was measured for each well to determine cell growth rates.

Spheroid formation assay

The MCF-7CD133+ and MCF-7CD133- subpopulations were seeded at a density of 1,000 cells/ml in 12-well low-adhesion plates, each well contained 1 ml spheroid formation medium. This medium consisted of DMEM/F12 supplemented with 1x B27, 20 ng/ml bFGF, 20 ng/ml EGF, 5 g/ml insulin, and 1% penicillin-streptomycin. Spheroid formation was observed under a light microscope (x100 magnification) after 3 days.

Endothelial cell differentiation culture

The MCF-7CD133+ and MCF-7CD133- subpopulations were cultured in stem cell maintenance medium, which was prepared by supplementing StemScale™ PSC medium (Gibco; Thermo Fisher Scientific, Inc.) with bFGF (20 ng/ml), EGF (20 ng/ml), and BMP4 (25 ng/ml; cat. no. 795604; BioLegend, Inc.). The cells were incubated for 2 days in low-adhesion dishes, and the media was replaced every 2 days. Subsequently, the cells were transferred to endothelial cell differentiation medium, which was composed of ECM medium (ScienCell) supplemented with VEGF (50 ng/ml; cat. no. AF-100-20-500UG; Thermo Fisher Scientific, Inc.) and bFGF (20 ng/ml). Cells were maintained in this differentiation culture media for 6 days, and the media was changed every 2 days.

Endothelial cell tube formation assay

Matrigel matrix gel was thawed at 4˚C and left undisturbed for 1 day. To initiate the experiment, refrigerated pipette tips and µ-Slide angiogenesis culture plates were used. A total of 10 µl Matrigel was added to each well of the µ-Slide plate. The µ-Slide plate was placed in an appropriately sized culture dish containing water-saturated absorbent paper to prevent moisture evaporation. The culture dishes were incubated for 30 min, allowing the gel to solidify. Cell suspensions (2x105 cells/ml) were prepared after cell digestion, and 50 µl of this suspension was added to the µ-Slide plate. The plates were covered and returned to the incubator for continuous culture. The cells were monitored using an optical microscope (x200 magnification) every 4-6 h.

DiI-labeled acetylated low-density lipoprotein (DiI-Ac-LDL) uptake assay

HUVECs, MCF-7CD133+, or MCF-7CD133- induced endothelial cells were seeded in 24-well plates and cultured for 48 h. Subsequently, the culture media was removed and cells were incubated in serum-free ECM endothelial cell medium for 3 h. DiI-Ac-LDL was prepared in serum-free EGM medium at a concentration of 10 µg/ml, added to cells, and incubated at 37˚C for 4 h. Following incubation, the media was discarded, and cells were washed three times in 0.01M PBS to eliminate any unbound DiI-Ac-LDL. Finally, supplemented culture medium was added and the cells were examined under a fluorescence microscope (x400 magnification).

MDM2/CEN12 fluorescent in situ hybridization (FISH) fluorescent probe detection

Cell preparation involved washing HUVEC, MCF-7CD133+, and MCF-7CD133--induced endothelial cells with 0.01M PBS, dropping them onto a glass slide, denaturing at 73˚C for 2 min, hybridizing with a MDM2/CEN12 probe (cat. no. FG0020; Abnova) at 38˚C for 16 h, and washing with 0.3% NP-40/SSC and 0.1% NP-40/SSC. The slides were sequentially dehydrated in 70, 90 and 100% ethanol for 2 min each. Finally, the cell nuclei were counterstained for 5 min at room temperature with DAPI and observed under a fluorescence microscope (x1,000 magnification).

Statistical analysis

Data were analyzed using SPSS version 18.0 (SPSS Inc.), all data are presented as the mean ± SD. Differences between groups were compared using an unpaired Student's t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

CD133 expression levels in MCF-7 pre- and post-immunomagnetic bead separation

Flow cytometry was used to assess CD133 expression in the MCF-7 breast cancer cells, and it was found that only 1.7±0.3% of the cells exhibited CD133 expression (Fig. 1A). Post anti-CD133 immunomagnetic bead sorting, two subpopulations, MCF-7CD133+ and MCF-7CD133-, were isolated. Flow cytometry was used to measure the proportions of CD133+ cells in these subpopulations, revealing expression rates of 85.6±2.8% for MCF-7CD133+ and 0.18±0.08% for MCF-7CD133- (Fig. 1B).

MCF-7 CD133+ cells exhibit increased in vitro proliferation and spheroid formation capabilities compared with the MCF-7CD133- cells

Compared with the MCF-7CD133- cells, the MCF-7CD133+ cells exhibited increased in vitro proliferation capacity, most notably on day 7 (Fig. 2A). The spheroid formation assay highlighted the cancer stem cell characteristics of both MCF-7CD133+ and MCF-7CD133- cells. After 3 days in low-attachment culture plates, MCF-7CD133+ cells displayed increased differentiation and growth capacity, resulting in a significantly higher number of spheroids compared with the MCF-7CD133- (Fig. 2B).

Assessment of endothelial cell marker expression pre- and post-endothelial cell induction culture

MCF-7CD133+ and MCF-7CD133- cells were cultured in stem cell maintenance media for 4 days, followed by 6 days in endothelial cell induction media, to induce tumor stem cells towards endothelial differentiation (Fig. 3A). Flow cytometry was used to assess the expression levels of endothelial cell surface markers CD31 and CD105 before and after induction. In the MCF-7CD133+ cells, the CD31+ proportions were 0.3±0.16% pre-induction and 81.4±8.37% post-induction, and the CD105+ proportions were 0.2±0.08% pre-induction and 83.8±7.24% post-induction (Fig. 3B). In MCF-7CD133- cells, the CD31+ proportions were 0.23±0.12% pre-induction and 3.95±2.1% post-induction, and the CD105+ proportions were 0.26±0.04% pre-induction and 6.3±2.6% post-induction (Fig. 3C).

Evaluation of endothelial cell function and gene amplification

In the endothelial cell tube formation assay, both the positive control HUVECs and MCF-7CD133+ cells formed lumen-like structures. In contrast, the MCF-7CD133- cells did not form these structures (Fig. 4A). In the endothelial cell uptake assay, both positive control HUVECs and MCF-7CD133+ cells emitted red fluorescence after staining with DiI-Ac-LDL (Fig. 4B). FISH experiments indicated that after induction, MCF-7CD133+ cells still exhibited amplification of the MDM2/CEN12 gene in the cell chromosomes (Fig. 4C).

Discussion

Breast cancer, a heterogeneous malignant tumor, is influenced by various risk factors including diet, environment, genetics, and epigenetics. Current data suggest that the 5-year survival rates for stage II and III breast cancer patients are 75 and 61%, respectively. However, 20-30% of cases still exhibit recurrence and/or metastasis (28). Therefore, finding effective strategies to prevent the recurrence and metastasis of breast cancer is crucial.

Contemporary theories suggest the presence of tumor stem cells in tumor patients, cells with characteristics akin to embryonic stem cells. These cells have unlimited proliferation, self-renewal, and multilineage differentiation capabilities. Additionally, they exhibit chemoresistance and radioresistance, contributing to recurrence and metastasis despite comprehensive anti-tumor therapies (29-31). CD133, also known as Prominin-1, a member of the Prominin family, is a five-transmembrane domain glycoprotein predominantly located on cell membrane surface protrusions and is recognized as a key biomarker for CSCs (32,33). In the present study, we initially identified a minor population of CD133-expressing cells within the MCF-7 breast cancer cell line. Utilizing anti-CD133 immunomagnetic bead separation, MCF-7CD133+ cells with a high CD133 positivity rate of approximately 85% were isolated, as determined by flow cytometry. It is important to note that the proportion of CSCs within cancer cell lines can indeed change over time and with continuous passaging. Thus, the proportion of stem cells and cancer cells within the cell lines across different passages was monitored to assess this variability. In comparison with MCF-7CD133- cells, the MCF-7CD133+ cells exhibited a significantly enhanced proliferative capacity. An actively proliferating subset of CSCs may play a crucial role in the growth and progression of tumors. These cells can give rise to more differentiated tumor cells while maintaining the CSC population, allowing the tumor to grow and potentially spread. When cultured in serum-free media enriched with growth factors, MCF-7CD133+ cells rapidly formed spheroids, exhibiting growth in suspension, and robust proliferation, while MCF-7CD133- cells showed a significantly reduced capacity for spheroid formation. These findings indicate that the MCF-7CD133+ cells were enriched in stem cells.

Tumor activities such as proliferation, invasion, and metastasis are closely associated with angiogenesis; however, the relationship between tumor stem cells and tumor vascular formation remains unclear and necessitates further investigation. In the present study, MCF-7CD133+ cells were isolated using immunomagnetic bead separation, and these cells were subjected to endothelial cell-inducing and maintenance media to transform the cells. Subsequently, the MCF-7CD133+ subpopulation cells formed luminal-like structures in the Matrigel matrix, akin to those formed by HUVECs (the positive control). Conversely, the MCF-7CD133- cells failed to form similar structures in endothelial cell transformation culture. In the endothelial cell phagocytosis assay, both the transformed MCF-7CD133+ cells and HUVECs internalized DiI-Ac-LDL and emitted red fluorescence, corroborating related studies (34,35). FISH was used to confirm that MCF-7CD133+ cells retained their tumor cell characteristics following endothelial cell culture conversion. These findings suggest that a distinct subpopulation of cells with stem cell-like properties, capable of differentiating into endothelial cells, exists in breast cancer.

The complex molecular mechanisms driving the differentiation of CSCs into vascular endothelial cells remain elusive. Previous research has shown that in hypoxic conditions, CSCs secrete VEGF, a powerful factor stimulating their transformation into endothelial cells, thus enhancing their propensity to differentiate under oxygen-deprived conditions (36). Alvero et al (37) found that CSCs from ovarian cancer possess the ability to differentiate into progenitors of vascular endothelial cells and form vascular-like structures in xenograft tumor inhibition models. In the present study, by enriching the culture medium with stimulatory factors such as bFGF, EGF, BMP4, and VEGF, transformation and maintenance cultivation of MCF-7CD133+ and MCF-7CD133- cells was successfully achieved. Subsequent endothelial cell tube formation and phagocytosis experiments suggested that the transformed MCF-7CD133+ cells exhibited endothelial cell functionality. These findings support the notion that CSCs can transform into vascular endothelial cells.

The tumor microenvironment plays a pivotal role in the onset and progression of malignant tumors (38). Modulating the tumor microenvironment can mitigate or inhibit tumor growth (39). Currently, modulating changes in the tumor microenvironment is challenging, but indirectly delaying or suppressing the formation of the tumor microenvironment by adjusting the functions of relevant cells within it, may serve as a treatment method for malignant tumors. Recent research has suggested that targeting tumor vascular endothelial cells is a promising direction for anti-tumor drug development. Anti-angiogenic drugs target various aspects of the angiogenic process. This includes inhibiting growth factors such as VEGF and its receptors (VEGFR), which are key drivers in the formation of new blood vessels. Drugs such as Bevacizumab (Avastin), an antibody that binds to VEGF, prevents it from activating VEGFR on endothelial cells. Tyrosine kinase inhibitors such as Sunitinib target VEGFR directly. The partial failure of anti-VEGF strategies in controlling cancer can be attributed to two major factors. First, the precise molecular mechanisms of cancer neo-angiogenesis are incompletely understood. Additionally, the abrogation of blood supply also restricts drug delivery to the tumor, reducing its effectiveness and promoting drug resistance (40). Correspondingly, a paradox in using anti-angiogenic drugs has emerged from recent findings. By inhibiting new blood vessel formation, anti-angiogenic drugs can increase the level of hypoxia (oxygen deprivation) within the tumor. This hypoxic environment can lead to the selection of more aggressive tumor cells that are better adapted to survive in low oxygen conditions (41). Several studies suggest that anti-angiogenic therapy might stimulate the tumor to become more invasive (42-45). In an attempt to access more blood supply, cancer cells might begin to invade surrounding tissues or spread to other parts of the body (46-51). These suggest that strategies aimed at normalizing tumor vessels, rather than eradicating blood supply, could enhance the delivery of therapeutic agents to cancer cells, thereby improving the efficacy and limiting cancer cell spread (52).

In conclusion, the present study induced the transformation of breast CSCs, and these transformed cells provided a more representative model for studying anti-angiogenesis in vitro than HUVECs. This approach addresses the challenges of the low availability and difficulty of separation of tumor vascular endothelial cells in vivo. It also lays the groundwork for more comprehensive and targeted research for understanding and combating tumor vascularization.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by grants from the National Natural Science Foundation of Ningbo (grant nos. 202003N4022 and 2019A610313) and the Zhejiang Provincial Medical and Health Science and Technology Plan (grant nos. 2021KY312 and 2023KY1048).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

QQM and XCJ contributed to the conception and design of the research. JNZ, WFT, QQM and SCZ performed the experiments and collected and interpreted the data. The first draft of the manuscript was written by QQM. SCZ and XCJ confirm the authenticity of all the raw data. All authors have read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Arnold M, Morgan E, Rumgay H, Mafra A, Singh D, Laversanne M, Vignat J, Gralow JR, Cardoso F, Siesling S and Soerjomataram I: Current and future burden of breast cancer: Global statistics for 2020 and 2040. Breast. 66:15–23. 2022.PubMed/NCBI View Article : Google Scholar

2 

Rivenbark AG, O'Connor SM and Coleman WB: Molecular and cellular heterogeneity in breast cancer. Am J Pathol. 183:1113–1124. 2013.PubMed/NCBI View Article : Google Scholar

3 

Szczepanek J, Skorupa M, Jarkiewicz-Tretyn J, Cybulski C and Tretyn A: Harnessing epigenetics for breast cancer therapy: The role of DNA methylation, histone modifications, and microRNA. Int J Mol Sci. 24(7235)2023.PubMed/NCBI View Article : Google Scholar

4 

Mackillop WJ, Ciampi A, Till JE and Buick RN: A stem cell model of human tumor growth: implications for tumor cell clonogenic assays. J Natl Cancer Inst. 70:9–16. 1983.PubMed/NCBI

5 

Llaguno SA and Parada LF: Cancer stem cells in gliomas: Evolving concepts and therapeutic implications. Curr Opin Neurol. 34:868–874. 2021.PubMed/NCBI View Article : Google Scholar

6 

Diehn M, Cho RW, Lobo NA, Kalisky T, Dorie MJ, Kulp AN, Qian D, Lam JS, Ailles LE, Wong MZ, et al: Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature. 458:780–783. 2009.PubMed/NCBI View Article : Google Scholar

7 

Hu Y and Fu L: Targeting cancer stem cells: A new therapy to cure cancer patients. Am J Cancer Res. 2:340–356. 2012.PubMed/NCBI

8 

Loureiro R, Mesquita KA, Oliveira PJ and Vega-Naredo I: Mitochondria in cancer stem cells: A target for therapy. Recent Pat Endocr Metab Immune Drug Discov. 7:102–114. 2013.PubMed/NCBI View Article : Google Scholar

9 

Phi LH, Sari IN, Yang YG, Lee SH, Jun NY, Kim KS, Lee YK and Kwon HY: Cancer stem cells (CSCs) in drug resistance and their therapeutic implications in cancer treatment. Stem Cells Int. 28(5416923)2018.PubMed/NCBI View Article : Google Scholar

10 

Griguer CE, Oliva CR, Gobin E, Marcorelles P, Benos DJ, Lancaster JR and Gillespie GY: CD133 is a marker of bioenergetic stress in human glioma. PLoS One. 3(e3655)2008.PubMed/NCBI View Article : Google Scholar

11 

Kunihiro AG, Sarrett SM, Lastwika KJ, Solan JL, Pisarenko T, Keinänen O, Rodriguez O, Taverne LR, Fitzpatrick AL, Li CI, et al: CD133 as a biomarker for an autoantibody-to-immunoPET paradigm for the early detection of small cell lung cancer. J Nucl Med. 63:1701–1707. 2022.PubMed/NCBI View Article : Google Scholar

12 

Howard R, Diffalha SA, Pimiento J, Mejia J, Enderling H, Giuliano A and Coppola D: CD133 expression as a helicobacter pylori-independent biomarker of gastric cancer progression. Anticancer Res. 38:4443–4448. 2018.PubMed/NCBI View Article : Google Scholar

13 

Ikram D, Masadah R, Nelwan BJ, Zainuddin AA, Ghaznawie M and Wahid S: CD133 act as an essential marker in ovarian carcinogenesis. Asian Pac J Cancer Prev. 22:3525–3531. 2021.PubMed/NCBI View Article : Google Scholar

14 

Lugano R, Ramachandran M and Dimberg A: Tumor angiogenesis: Causes, consequences, challenges and opportunities. Cell Mol Life Sci. 77:1745–1770. 2020.PubMed/NCBI View Article : Google Scholar

15 

Liu ZL, Chen HH, Zheng LL, Sun LP and Shi L: Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct Target Ther. 8(198)2023.PubMed/NCBI View Article : Google Scholar

16 

Viallard C and Larrivée B: Tumor angiogenesis and vascular normalization: alternative therapeutic targets. Angiogenesis. 20:409–426. 2017.PubMed/NCBI View Article : Google Scholar

17 

Ferrara N, Gerber HP and LeCouter J: The biology of VEGF and its receptors. Nat Med. 9:669–676. 2003.PubMed/NCBI View Article : Google Scholar

18 

Apte RS, Chen DS and Ferrara N: VEGF in signaling and disease: Beyond discovery and development. Cell. 176:1248–1264. 2019.PubMed/NCBI View Article : Google Scholar

19 

Turner N and Grose R: Fibroblast growth factor signaling: from development to cancer. Nat Rev Cancer. 10:116–129. 2010.PubMed/NCBI View Article : Google Scholar

20 

Johnson KE and Wilgus TA: Vascular endothelial growth factor and angiogenesis in the regulation of cutaneous wound repair. Adv Wound Care (New Rochelle). 3:647–661. 2014.PubMed/NCBI View Article : Google Scholar

21 

Phillips TM, McBride WH and Pajonk F: The response of CD24(-/low)/CD44+ breast cancer-initiating cells to radiation. J Natl Cancer Inst. 98:1777–1785. 2006.PubMed/NCBI View Article : Google Scholar

22 

Puca F, Fedele M, Rasio D and Battista S: Role of diet in stem and cancer stem cells. Int J Mol Sci. 23(8108)2022.PubMed/NCBI View Article : Google Scholar

23 

Katayama Y, Uchino J, Chihara Y, Tamiya N, Kaneko Y, Yamada T and Takayama K: Tumor neovascularization and developments in therapeutics. Cancers (Basel). 11(316)2019.PubMed/NCBI View Article : Google Scholar

24 

Aomatsu N, Yashiro M, Kashiwagi S, Takashima T, Ishikawa T, Ohsawa M, Wakasa K and Hirakawa K: CD133 is a useful surrogate marker for predicting chemosensitivity to neoadjuvant chemotherapy in breast cancer. PLoS One. 7(e45865)2012.PubMed/NCBI View Article : Google Scholar

25 

Sun C, Li JM, Wang B, Shangguan JJ, Figini M, Shang N, Pan L and Zhang ZL: Tumor angiogenesis and bone metastasis-correlation in invasive breast carcinoma. J Immunol Methods. 452:46–52. 2018.PubMed/NCBI View Article : Google Scholar

26 

Lv XQ, Wang YZ, Song YM, Pang X and Li HX: Association between ALDH1+/CD133+ stem-like cells and tumor angiogenesis in invasive ductal breast carcinoma. Oncol Lett. 11:1750–1756. 2016.PubMed/NCBI View Article : Google Scholar

27 

Sudhan DR, Rabaglino MB, Wood CE and Siemann DW: Cathepsin L in tumor angiogenesis and its therapeutic intervention by the small molecule inhibitor KGP94. Clin Exp Metastasis. 33:461–73. 2016.PubMed/NCBI View Article : Google Scholar

28 

Kim W, Kim KS and Park RW: Nomogram of naive bayesian model for recurrence prediction of breast cancer. Healthc Inform Res. 22:89–94. 2016.PubMed/NCBI View Article : Google Scholar

29 

Panigoro SS, Kurnia D, Kurnia A, Haryono SJ and Albar ZA: ALDH1 cancer stem cell marker as a prognostic factor in triple-negative breast cancer. Int J Surg Oncol. 2020(7863243)2020.PubMed/NCBI View Article : Google Scholar

30 

Yu JG, Liao XH, Li YL, Lv L, Zhi XL, Yu J and Zhou P: A preliminary study of the role of extracellular-5'-nucleotidase in breast cancer stem cells and epithelial-mesenchymal transition. In Vitro Cell Dev Biol Anim. 53:132–140. 2017.PubMed/NCBI View Article : Google Scholar

31 

Yang LQ, Shi PF and Zhao GC: Targeting cancer stem cell pathways for cancer therapy. Signal Transduct Target Ther. 5(8)2020.PubMed/NCBI View Article : Google Scholar

32 

Irollo E and Pirozzi G: CD133: To be or not to be, is this the real question? Am J Transl Res. 5:563–581. 2013.PubMed/NCBI

33 

Bauer N, Fonseca AV, Florek M, Freund D, Jászai J, Bornhäuser M, Fargeas CA and Corbeil D: New insights into the cell biology of hematopoietic progenitors by studying prominin-1 (CD133). Cells Tissues Organs. 188:127–138. 2008.PubMed/NCBI View Article : Google Scholar

34 

Zou L, Liu XW, Li JJ, Li W, Zhang LL, Li J and Zhang JM: Tetramethylpyrazine enhances the antitumor effect of paclitaxel by inhibiting angiogenesis and inducing apoptosis. Front Pharmacol. 10(707)2019.PubMed/NCBI View Article : Google Scholar

35 

Ahmed N, Abubaker K, Findlay J and Quinn M: Epithelial mesenchymal transition and cancer stem cell-like phenotypes facilitate chemoresistance in recurrent ovarian cancer. Curr Cancer Drug Targets. 10:268–278. 2010.PubMed/NCBI View Article : Google Scholar

36 

Liu XM, Zhang QP, Mu YG, Zhang XH, Sai K, Pang JC, Ng HK and Chen ZP: Clinical significance of vasculogenic mimicry in human gliomas. J Neurooncol. 105:173–179. 2011.PubMed/NCBI View Article : Google Scholar

37 

Alvero AB, Chen R, Fu HH, Montagna M, Schwart PE, Rutherford T, Silasi DA, Steffensen KD, Waldstrom M, Visintin I and Mor G: Molecular phenotyping of human ovarian cancer stem cells unravels the mechanisms for repair and chemoresistance. Cell Cycle. 8:158–166. 2009.PubMed/NCBI View Article : Google Scholar

38 

Yuan Y, Jiang YC, Sun CK and Chen QM: Role of the tumor microenvironment in tumor progression and the clinical applications (review). Oncol Rep. 35:2499–2515. 2016.PubMed/NCBI View Article : Google Scholar

39 

Hinshaw DC and Shevde LA: The tumor microenvironment innately modulates cancer progression. Cancer Res. 79:4557–4566. 2019.PubMed/NCBI View Article : Google Scholar

40 

Rohlenova K, Veys K, Miranda-Santos I, Bock KD and Carmeliet P: Endothelial cell metabolism in health and disease. Trends Cell Biol. 28:224–236. 2018.PubMed/NCBI View Article : Google Scholar

41 

Muz B, Puente PD, Azab F and Azab AK: The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy. Hypoxia (Auckl). 3:83–92. 2015.PubMed/NCBI View Article : Google Scholar

42 

Vasudev NS and Reynolds AR: Anti-angiogenic therapy for cancer: current progress, unresolved questions and future directions. Angiogenesis. 17:471–494. 2014.PubMed/NCBI View Article : Google Scholar

43 

Alexandre J, Batteux F, Nicco C, Chéreau C, Laurent A, Guillevin L, Weill B and Goldwasser F: . Accumulation of hydrogen peroxide is an early and crucial step for paclitaxel-induced cancer cell death both in vitro and in vivo. Int J Cancer. 119:41–48. 2006.PubMed/NCBI View Article : Google Scholar

44 

Fukui M, Yamabe N and Zhu BT: . Resveratrol attenuates the anticancer efficacy of paclitaxel in human breast cancer cells in vitro and in vivo. Eur J Cancer. 46:1882–1891. 2010.PubMed/NCBI View Article : Google Scholar

45 

Haibe Y, Kreidieh M, El Hajj H, Khalifeh I, Mukherji D, Temraz S and Shamseddine A: Resistance mechanisms to anti-angiogenic therapies in cancer. Front Oncol. 10(221)2020.PubMed/NCBI View Article : Google Scholar

46 

Ansari MJ, Bokov D, Markov A, Jalil AT, Shalaby MN, Suksatan W, Chupradit S, Al-Ghamdi HS, Shomali N, Zamani A, et al: Cancer combination therapies by angiogenesis inhibitors; a comprehensive review. Cell Commun Signal. 20(49)2022.PubMed/NCBI View Article : Google Scholar

47 

Eelen G, Treps L, Li XR and Carmeliet P: Basic and therapeutic aspects of angiogenesis updated. Circ Res. 127:310–329. 2020.PubMed/NCBI View Article : Google Scholar

48 

Lee WS, Yang H, Chon HJ and Kim C: Combination of anti-angiogenic therapy and immune checkpoint blockade normalizes vascular-immune crosstalk to potentiate cancer immunity. Exp Mol Med. 52:1475–1485. 2020.PubMed/NCBI View Article : Google Scholar

49 

Huinen ZR, Huijbers EJ, Beijnum JR, Nowak-Sliwinska P and Griffioen AW: Anti-angiogenic agents-overcoming tumour endothelial cell anergy and improving immunotherapy outcomes. Nat Rev Clin Oncol. 18:527–540. 2021.PubMed/NCBI View Article : Google Scholar

50 

Wehland M, Bauer J, Infanger M and Grimm M: Target-based anti-angiogenic therapy in breast cancer. Curr Pharm Des. 18:4244–4257. 2012.PubMed/NCBI View Article : Google Scholar

51 

Yetkin-Arik B, Kastelein AW, Klaassen I, Jansen CH, Latul YP, Vittori M, Biri A, Kahraman K, Griffioen AW, Amant F, et al: Angiogenesis in gynecological cancers and the options for anti-angiogenesis therapy. Biochim Biophys Acta Rev Cancer. 1875(188446)2021.PubMed/NCBI View Article : Google Scholar

52 

Fukumura D and Jain RK: Tumor microvasculature and microenvironment: targets for anti-angiogenesis and normalization. Microvasc Res. 74:72–84. 2007.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

February-2024
Volume 27 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Mao Q, Ji X, Zhang J, Teng W and Zhou S: A novel approach for transforming breast cancer stem cells into endothelial cells. Exp Ther Med 27: 74, 2024
APA
Mao, Q., Ji, X., Zhang, J., Teng, W., & Zhou, S. (2024). A novel approach for transforming breast cancer stem cells into endothelial cells. Experimental and Therapeutic Medicine, 27, 74. https://doi.org/10.3892/etm.2023.12362
MLA
Mao, Q., Ji, X., Zhang, J., Teng, W., Zhou, S."A novel approach for transforming breast cancer stem cells into endothelial cells". Experimental and Therapeutic Medicine 27.2 (2024): 74.
Chicago
Mao, Q., Ji, X., Zhang, J., Teng, W., Zhou, S."A novel approach for transforming breast cancer stem cells into endothelial cells". Experimental and Therapeutic Medicine 27, no. 2 (2024): 74. https://doi.org/10.3892/etm.2023.12362