Evaluation of chemotherapy and P2Et extract combination in ex-vivo derived tumor mammospheres from breast cancer patients

Claudia Urueña, Tito A Sandoval, Paola Lasso, Mauricio Tawil, Alfonso Barreto, Lilian Torregrosa, Susana Fiorentino, Claudia Urueña, Tito A Sandoval, Paola Lasso, Mauricio Tawil, Alfonso Barreto, Lilian Torregrosa, Susana Fiorentino

Abstract

The main cause of death by cancer is metastasis rather than local complications of primary tumors. Recent studies suggest that breast cancer stem cells (BCSCs), retains the ability to self-renew and differentiate to repopulate the entire tumor, also, they have been associated with resistance to chemotherapy and tumor recurrence, even after tumor resection. Chemotherapy has been implicated in the induction of resistant phenotypes with highly metastatic potential. Naturally occurring compounds, especially phytochemicals such as P2Et, can target different populations of cancer cells as well as BCSC, favoring the activation of immune response via immunogenic tumor death. Here, we evaluated the presence of BCSC as well as markers related to drug resistance in tumors obtained from 78 patients who had received (or not) chemotherapy before surgery. We evaluated the ex vivo response of patient tumor-derived organoids (or mammospheres) to chemotherapy alone or in combination with P2Et. A xenotransplant model engrafted with MDA-MB-468 was used to evaluate in vivo the activity of P2Et, in this model P2Et delay tumor growth. We show that patients with luminal and TNBC, and those who received neoadjuvant therapy before surgery have a higher frequency of BCSC. Further, the treatment with P2Et in mammospheres and human breast cancer cell lines improve the in vitro tumor death and decrease its viability and proliferation together with the release of immunogenic signals. P2Et could be a good co-adjuvant in antitumor therapy in patients, retarding the tumor growth by enabling the activation of the immune response.

Conflict of interest statement

SF, CU, TS and PL are inventors of a granted patent related to P2Et. The other authors declare no competing interests.

Figures

Figure 1
Figure 1
Cancer stem cell features in breast cancer patients. The patients were classified in Healthy Donor (HD), Luminal A (LA), Luminal B (LB), Triple negative (TN) and Her-2+. (a) Sample processing workflow of breast cancer tumors. (b) Multidimensional reduction analysis (tSNE) of flow cytometry data by breast cancer sub-type, rainbow scale represents relative intensity by channel; bulk population (gray) and CD44 + CD24-EPCAM + CD49f + (light blue). (c) CD45 + tumor-infiltration lymphocytes. (d) Frequency of Aldehyde dehydrogenase 1 (ALDH1) positive cells. (e) Frequency of Lin-CD44 + CD24-CD49f + EPCAM + cells. (f) Frequency of BCRP+. (g) Frequency of Pgp+. (h) Frequency of MRP1 + cells by flow cytometry. Data are presented as violin plots and each point represents independent samples; dotted lines indicate quartiles. Multiple comparisons were calculated by one-way ANOVA with Dunnet T3 correction, and significant exact p-values are shown. *p < 0.05; **p < 0.01; ***p < 0.001, **** p < 0.0001.
Figure 2
Figure 2
Neoadjuvant chemotherapy (NAT) before surgery increase the frequency of BCSC. (a) Frequency of BCSC (Lin-/CD44 + CD24-CD49f + EPCAM+) in healthy donor (HD), breast cancer patients with or without NAT before surgery. (b) Frequency of BCSC in LB patients with or without NAT before surgery. (c) Frequency of BCSC (ALDH +) in HD, breast cancer patients with or without NAT before surgery. (d) Frequency of BCSC (ALDH +) in TN breast cancer patients who received or not NAT before surgery. (AC (Anthracyclines + Ciclophosphamide), AC + TX (Anthracyclines/Ciclophosphamide + Taxanes), TX (Taxanes). Data are presented as violin plots and each point represents an independent sample; dotted lines indicate quartiles. Multiple comparisons were calculated by one-way ANOVA with Tukey post-test analysis and unpaired t-test, significant exact p-value are shown. *p < 0.05; **p < 0.01.
Figure 3
Figure 3
Breast cancer stem cells markers (ALDH+) correlated with BCRP expression in triple negative patients. Correlation of BCRP protein and ABCG2 gene expression with BCSC markers (ALDH+ and CD24-CD44 + CD49f + EPCAM +) in breast cancer patients who received or not NAT before surgery. (a) Correlation of BCRP protein with ALDH + and CD24-CD44 + CD49f+ EPCAM + expression in all samples. (b) Correlation of ABCG2 gene expression with ALDH + and CD24-CD44 + CD49f + EPCAM + in all samples. (c) Correlation of BCRP with ALDH + and CD24-CD44 + CD49f + EPCAM + expression in breast cancer patients who received NAT before surgery. Correlation of BCRP protein with BCSC markers (ALDH+ and CD24-CD44+ CD49f + EPCAM+) expression in (d) LA only, (e) LB only and (f) TN patients who received NAT before surgery. Correlations were assessed using nonparametric Spearman correlation, determination coefficient r and p-value are shown. *p < 0.05.
Figure 4
Figure 4
P2Et extract decrease the viability of mammospheres derived from breast cancer patients after in vitro treatment. (a) Representative confocal microscopy image of the effect of P2Et extract, DX or DX + P2Et on the mammospheres viability of breast cancer patients. Luminal A (LA), Luminal B (LB), Triple negative (TN) and Her2. Calcein AM (green), Ethidium homodimer-1 (Red). (b) Cell death percentage in mammospheres by molecular subgroups of breast cancer patients after treatment. (c) Cell death percentage in all mammospheres from breast cancer patients after treatment. Data are presented as violin plots and each point represents an independent sample; dotted lines indicate quartiles. Exact p-values were calculated using One-way ANOVA with Tukey post-test analysis. (d) Spearman nonparametric correlation between percentage of cell death with ABCB1, ABCG2, ABCC1 gene in mammospheres from breast cancer patients after P2Et, DX or Ethanol (negative control) treatment, coefficient of determination r and exact p-value are shown. *p < 0.05; **p < 0.01; ***p < 0.001.
Figure 5
Figure 5
P2Et extract decrease the viability and proliferation of human triple negative breast cancer cells. (a) Frequency of BCSC ALDH1 + and Lin-CD44 + CD24-CD49f + EPCAM + cells in MCF-7, BT-549 and MDA-MB-468 breast cancer cells measured by flow cytometry. Data are presented as the mean ± SEM. (b) IC50 values of P2Et extract or Doxorubicin (DX) in MCF-7, BT-549 and MDA-MB-468 using MTT Assay. (c) Left side: MDA-MB-468 cell count by trypan blue after treatment with vehicle (Ethanol), P2Et IC50, DX IC50, or P2Et + DX IC50 for 0, 48, 72 and 96 h. Right side: MDA-MB-468 cells were stained with CFSE and treated with P2Et IC50, DX IC50, or P2Et + DX IC50 for 0, 48, 72 and 96 h. For each time the proliferation was evaluated by flow cytometry. (d) The combined inhibitory effects of P2Et (0 to 500 μg/ml) and DX (0 to 1.16 μg/ml) were tested over a range of combinations against MDA-MB-468 cells using MTT Assay (Left side); a dose–response matrix was generated and analyzed for zero-interaction potency method (ZIP), using SynergyFinder pipeline (Right side). Data are presented as the mean of three independent experiments. (e) Frequency of death (Annexin V+ , propidium iodide [PI]−) and Annexin V+ , PI+) MDA-MB-468 cells after 24 and 48 h of treatment with Etanol, P2Et (IC50 and IC50*2) or DX (IC50 and IC50*2). MDA-MB-468 cells were stained with Annexin V-Alexa Fluor 488 and PI. Data are presented as the mean value ± SEM of 4 independent experiments. (f) MDA-MB-468 cells were treated with vehicle (ethanol or DMSO), P2Et (IC50 and IC50*2) or DX (IC50 and IC50*2) for 24 h. Surface exposure of Calreticuline (CRT) was determined by flow cytometry among viable cells (Aqua negative). Data are presented as violin plots and each point represents independent samples; dotted lines indicate quartiles. (g) MDA-MB-468 cells were treated vehicle (ethanol or DMSO), P2Et (IC50 and IC50*2) or DX (IC50 and IC50*2) for 48 h. After treatment cells were stained with quinacrine (1 μM) and PI. Quinacrine low cells were determined among viable cells (PI negative cells). Data are presented as violin plots and each point represents independent samples; dotted lines indicate quartiles. Exact significant p-values are shown. *p < 0.05; **p < 0.01; ***p < 0.001, **** p < 0.0001.
Figure 6
Figure 6
P2Et extract retard initially primary tumor growth but increase survival in triple negative human breast cancer. (a) Experimental scheme of treatment. MDA-MB-468 cells were inoculated into the mouse mammary gland, 8 days later, the mice were treated with PBS (negative control) or 18.7 mg/Kg of P2Et extract twice a week until endpoint. (b) Tumor growth (mm3) of MDA-MB-468 control (PBS) mice or mice treated with P2Et extract. Multiple t-test with Holm-Sidak method correction was performed assuming significance of α = 0.05. *p < 0.05.

References

    1. Bray F, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018;68:394–424. doi: 10.3322/caac.21492.
    1. Provenzano E, Ulaner GA, Chin SF. Molecular classification of breast cancer. PET Clin. 2018;13:325–338. doi: 10.1016/j.cpet.2018.02.004.
    1. Curtis C, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486:346–352. doi: 10.1038/nature10983.
    1. Lehmann BD, et al. Refinement of triple-negative breast cancer molecular subtypes: implications for neoadjuvant chemotherapy selection. PLoS ONE. 2016;11:e0157368. doi: 10.1371/journal.pone.0157368.
    1. Litière S, et al. Breast conserving therapy versus mastectomy for stage I-II breast cancer: 20 year follow-up of the EORTC 10801 phase 3 randomised trial. Lancet Oncol. 2012;13:412–419. doi: 10.1016/S1470-2045(12)70042-6.
    1. Charafe-Jauffret E, et al. Aldehyde dehydrogenase 1-positive cancer stem cells mediate metastasis and poor clinical outcome in inflammatory breast cancer. Clin. Cancer Res. 2010;16:45–55. doi: 10.1158/1078-0432.CCR-09-1630.
    1. Butti R, Gunasekaran VP, Kumar TVS, Banerjee P, Kundu GC. Breast cancer stem cells: Biology and therapeutic implications. Int. J. Biochem. Cell. Biol. 2019;107:38–52. doi: 10.1016/j.biocel.2018.12.001.
    1. Collina F, et al. Prognostic value of cancer stem cells markers in triple-negative breast cancer. Biomed. Res. Int. 2015;2015:158682. doi: 10.1155/2015/158682.
    1. Yousefnia S, Ghaedi K, Seyed Forootan F, Nasr Esfahani MH. Characterization of the stemness potency of mammospheres isolated from the breast cancer cell lines. Tumour Biol. 2019;41:1010428319869101. doi: 10.1177/1010428319869101.
    1. Cojoc M, Mabert K, Muders MH, Dubrovska A. A role for cancer stem cells in therapy resistance: cellular and molecular mechanisms. Semin. Cancer Biol. 2015;31:16–27. doi: 10.1016/j.semcancer.2014.06.004.
    1. Gomez-Miragaya J, et al. Resistance to taxanes in triple-negative breast cancer associates with the dynamics of a CD49f+ tumor-initiating population. Stem Cell Rep. 2017;8:1392–1407. doi: 10.1016/j.stemcr.2017.03.026.
    1. Batlle E, Clevers H. Cancer stem cells revisited. Nat. Med. 2017;23:1124–1134. doi: 10.1038/nm.4409.
    1. De Angelis ML, Francescangeli F, La Torre F, Zeuner A. Stem cell plasticity and dormancy in the development of cancer therapy resistance. Front. Oncol. 2019;9:626. doi: 10.3389/fonc.2019.00626.
    1. Keklikoglou I, et al. Chemotherapy elicits pro-metastatic extracellular vesicles in breast cancer models. Nat. Cell. Biol. 2019;21:190–202. doi: 10.1038/s41556-018-0256-3.
    1. Lu H, et al. Chemotherapy-induced Ca(2+) release stimulates breast cancer stem cell enrichment. Cell Rep. 2017;18:1946–1957. doi: 10.1016/j.celrep.2017.02.001.
    1. Lu H, et al. Chemotherapy triggers HIF-1-dependent glutathione synthesis and copper chelation that induces the breast cancer stem cell phenotype. Proc. Natl. Acad. Sci. USA. 2015;112:E4600–4609. doi: 10.1073/pnas.1513433112.
    1. Kitamura T, Qian BZ, Pollard JW. Immune cell promotion of metastasis. Nat. Rev. Immunol. 2015;15:73–86. doi: 10.1038/nri3789.
    1. Hirohashi Y, et al. Immune responses to human cancer stem-like cells/cancer-initiating cells. Cancer Sci. 2016;107:12–17. doi: 10.1111/cas.12830.
    1. Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu. Rev. Immunol. 2013;31:51–72. doi: 10.1146/annurev-immunol-032712-100008.
    1. Dandawate PR, Subramaniam D, Jensen RA, Anant S. Targeting cancer stem cells and signaling pathways by phytochemicals: novel approach for breast cancer therapy. Semin. Cancer Biol. 2016;40–41:192–208. doi: 10.1016/j.semcancer.2016.09.001.
    1. Urueña C, et al. Multifunctional T lymphocytes generated after therapy with an antitumor gallotanin-rich normalized fraction are related to primary tumor size reduction in a breast cancer model. Integr. Cancer Ther. 2015;14:468–483. doi: 10.1177/1534735415596425.
    1. Gomez-Cadena A, et al. Immune-system-dependent anti-tumor activity of a plant-derived polyphenol rich fraction in a melanoma mouse model. Cell Death Dis. 2016;7:e2243. doi: 10.1038/cddis.2016.134.
    1. Sandoval TA, et al. Standardized extract from caesalpinia spinosa is cytotoxic over cancer stem cells and enhance anticancer activity of doxorubicin. Am. J. Chin. Med. 2016;44:1693–1717. doi: 10.1142/S0192415X16500956.
    1. Lasso P, et al. Breast tumor cells highly resistant to drugs are controlled only by the immune response induced in an immunocompetent mouse model. Integr. Cancer. Ther. 2019;18:1534735419848047. doi: 10.1177/1534735419848047.
    1. DeRose YS, et al. Patient-derived models of human breast cancer: protocols for in vitro and in vivo applications in tumor biology and translational medicine. Curr. Protoc. Pharmacol. 2013 doi: 10.1002/0471141755.ph1423s60.
    1. Li H, et al. Stem cell marker aldehyde dehydrogenase 1 (ALDH1)-expressing cells are enriched in triple-negative breast cancer. Int. J. Biol. Markers. 2013;28:e357–364. doi: 10.5301/jbm.5000048.
    1. Creighton CJ, et al. Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features. Proc. Natl. Acad. Sci. USA. 2009;106:13820–13825. doi: 10.1073/pnas.0905718106.
    1. Tanei T, et al. Association of breast cancer stem cells identified by aldehyde dehydrogenase 1 expression with resistance to sequential Paclitaxel and epirubicin-based chemotherapy for breast cancers. Clin. Cancer Res. 2009;15:4234–4241. doi: 10.1158/1078-0432.CCR-08-1479.
    1. Colacino JA, et al. Heterogeneity of human breast stem and progenitor cells as revealed by transcriptional profiling. Stem Cell Rep. 2018;10:1596–1609. doi: 10.1016/j.stemcr.2018.03.001.
    1. Yao H, et al. Triple-negative breast cancer: is there a treatment on the horizon? Oncotarget. 2017;8:1913–1924. doi: 10.18632/oncotarget.12284.
    1. Prieto K, et al. Polyphenol-rich extract induces apoptosis with immunogenic markers in melanoma cells through the ER stress-associated kinase PERK. Cell Death Discov. 2019;5:134. doi: 10.1038/s41420-019-0214-2.
    1. Tesniere A, et al. Immunogenic death of colon cancer cells treated with oxaliplatin. Oncogene. 2010;29:482–491. doi: 10.1038/onc.2009.356.
    1. Schmittgen TD, Zakrajsek BA. Effect of experimental treatment on housekeeping gene expression: validation by real-time, quantitative RT-PCR. J. Biochem. Biophys. Methods. 2000;46:69–81. doi: 10.1016/S0165-022X(00)00129-9.
    1. Uruena C, et al. Petiveria alliacea extracts uses multiple mechanisms to inhibit growth of human and mouse tumoral cells. BMC Complement. Altern. Med. 2008;8:60. doi: 10.1186/1472-6882-8-60.
    1. Ianevski A, He L, Aittokallio T, Tang J. SynergyFinder: a web application for analyzing drug combination dose-response matrix data. Bioinformatics. 2017;33:2413–2415. doi: 10.1093/bioinformatics/btx162.
    1. Yadav B, Wennerberg K, Aittokallio T, Tang J. Searching for drug synergy in complex dose-response landscapes using an interaction potency model. Comput. Struct. Biotechnol. J. 2015;13:504–513. doi: 10.1016/j.csbj.2015.09.001.
    1. Martins I, et al. Molecular mechanisms of ATP secretion during immunogenic cell death. Cell Death Differ. 2014;21:79–91. doi: 10.1038/cdd.2013.75.
    1. Lasso P, et al. Prophylactic vs. therapeutic treatment with P2Et polyphenol-rich extract has opposite effects on tumor growth. Front. Oncol. 2018;8:356. doi: 10.3389/fonc.2018.00356.
    1. Lehmann BD, et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J. Clin. Invest. 2011;121:2750–2767. doi: 10.1172/JCI45014.
    1. Park SY, et al. Heterogeneity for stem cell-related markers according to tumor subtype and histologic stage in breast cancer. Clin. Cancer Res. 2010;16:876–887. doi: 10.1158/1078-0432.CCR-09-1532.
    1. O'Conor CJ, Chen T, Gonzalez I, Cao D, Peng Y. Cancer stem cells in triple-negative breast cancer: a potential target and prognostic marker. Biomark. Med. 2018;12:813–820. doi: 10.2217/bmm-2017-0398.
    1. Pommier SJ, et al. Surgical resection of breast cancers: molecular analysis of cancer stem cells in residual disease. Surgery. 2019;165:1008–1013. doi: 10.1016/j.surg.2019.01.001.
    1. Song X, et al. Hypoxia-induced resistance to cisplatin and doxorubicin in non-small cell lung cancer is inhibited by silencing of HIF-1alpha gene. Cancer Chemother. Pharmacol. 2006;58:776–784. doi: 10.1007/s00280-006-0224-7.
    1. Abdel-Daim MM, El-Tawil OS, Bungau SG, Atanasov AG. Applications of antioxidants in metabolic disorders and degenerative diseases: mechanistic approach. Oxid. Med. Cell Longev. 2019;2019:4179676. doi: 10.1155/2019/4179676.
    1. Pluchino KM, Hall MD, Goldsborough AS, Callaghan R, Gottesman MM. Collateral sensitivity as a strategy against cancer multidrug resistance. Drug Resist. Update. 2012;15:98–105. doi: 10.1016/j.drup.2012.03.002.
    1. Zhou Q, Bennett LL, Zhou S. Multifaceted ability of naturally occurring polyphenols against metastatic cancer. Clin. Exp. Pharmacol. Physiol. 2016;43:394–409. doi: 10.1111/1440-1681.12546.
    1. Liu Y, et al. Lack of correlation of stem cell markers in breast cancer stem cells. Br. J. Cancer. 2014;110:2063–2071. doi: 10.1038/bjc.2014.105.
    1. Pan M, et al. Inhibition of breast cancer growth via miR-7 suppressing ALDH1A3 activity concomitant with decreasing breast cancer stem cell subpopulation. J. Cell Physiol. 2020;235:1405–1416. doi: 10.1002/jcp.29059.
    1. Clark DW, Palle K. Aldehyde dehydrogenases in cancer stem cells: potential as therapeutic targets. Ann. Transl. Med. 2016;4:518. doi: 10.21037/atm.2016.11.82.
    1. Visus C, et al. Targeting ALDH(bright) human carcinoma-initiating cells with ALDH1A1-specific CD8(+) T cells. Clin. Cancer Res. 2011;17:6174–6184. doi: 10.1158/1078-0432.CCR-11-1111.

Source: PubMed

3
Subskrybuj