Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1682 2023-12-12 18:32:38 |
2 format change Meta information modification 1682 2023-12-13 03:31:31 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Molodtsova, D.; Guryev, D.V.; Osipov, A.N. Conditioned Media from Radioresistant and Chemoresistant Cancer Cells. Encyclopedia. Available online: https://encyclopedia.pub/entry/52628 (accessed on 01 July 2024).
Molodtsova D, Guryev DV, Osipov AN. Conditioned Media from Radioresistant and Chemoresistant Cancer Cells. Encyclopedia. Available at: https://encyclopedia.pub/entry/52628. Accessed July 01, 2024.
Molodtsova, Daria, Denis V. Guryev, Andreyan N. Osipov. "Conditioned Media from Radioresistant and Chemoresistant Cancer Cells" Encyclopedia, https://encyclopedia.pub/entry/52628 (accessed July 01, 2024).
Molodtsova, D., Guryev, D.V., & Osipov, A.N. (2023, December 12). Conditioned Media from Radioresistant and Chemoresistant Cancer Cells. In Encyclopedia. https://encyclopedia.pub/entry/52628
Molodtsova, Daria, et al. "Conditioned Media from Radioresistant and Chemoresistant Cancer Cells." Encyclopedia. Web. 12 December, 2023.
Conditioned Media from Radioresistant and Chemoresistant Cancer Cells
Edit

Chemoradiation therapy (CRT) is a commonly indicated treatment in the case of cancer. In combination with surgery or alone, it offers a relief of condition or even a cure for some patients. Resistance to chemo- or radiotherapy is the main obstacle to consistent treatment outcomes in oncology patients.

radioresistance chemoresistance cancer cell conditioned media secretory factors oncosomes exosomes miRNA

1. Introduction

Chemoradiation therapy (CRT) is a commonly indicated treatment in the case of cancer. In combination with surgery or alone, it offers a relief of condition or even a cure for some patients. In cases when cancer cannot be abolished with CRT, resistance is often the case. Resistance can be a pre-existing tumor state due to a genetic profile or presence of cancer stem cells (CSCs), cancer-associated fibroblasts (CAFs), or mesenchymal stem cells (MSCs), or it can be acquired. Previous treatment with radiation or chemotherapy (CT) can lead to the selection of surviving cells with a resistant phenotype and/or the transformation of surviving cancer cells into resistant cells. This resistant phenotype allows cancer cells to escape death after future treatment and maintain a viable tumor cell population. Various mechanisms have been described that are associated with resistance, and many are shared between radioresistant and chemoresistant cells. Increased repair capacity, abrogation of cell cycle arrest, apoptosis evasion, tumor heterogeneity, activation of CSCs, mutation, and tumor microenvironment (TME) are common mechanisms leading to both radio- and chemoresistance. When anti-cancer therapy is administered, tumor cells exert a complex response that includes a state-specific secretory profile that orchestrates communication between cells to help them adapt. A variety of molecules, including cytokines, interleukins, mRNA, and ncRNA, such as miRNA and lncRNA, as well as others, are released into the extracellular space and can cause changes in the cells that uptake them and also reflect the cell-specific state.

2. Conditioned Media

To reveal the influence of TME on the development of resistance of cancer cells, experiments with conditioned media (CM) were conducted. CM contains various paracrine factors and oncosomes that convey molecular signals and aid in cellular communication. Among the resistance-associated secretory molecules, circular RNAs (circRNA) and cytokines have been identified. Cytokines bind to cellular receptors and initiate a signal cascade, while circRNA can enter by endocytosis. circITGB6 was found to be associated with cisplatin resistance in M2 macrophages, while circATP8B4 facilitates higher viability after ionizing radiation (IR) treatment [1][2]. In addition to circRNA, interleukin-11 (IL-11), secreted by cancer-associated fibroblasts, previously treated with cisplatin, also induced a significantly higher viability of A549 cells following cisplatin exposure by activating the IL-11R/STAT3 anti-apoptotic signaling pathway [3].
Other studies explored radioresistance induction and showed that CM from previously irradiated cells also showed protective effects in the A549 cells upon additional X-ray exposure, leading to lower apoptotic rates [4]. CM from three types of previously irradiated lung cancer cells facilitated decreased cell death upon irradiation of sensitive cells via increased plasminogen activator inhibitor-1 (PAI-1) that upregulated AKT and ERK1/2 pathways and inhibited caspase-3 activity [5].
In vivo TME contains senescent cells that are formed after anti-cancer therapy that express the senescence-associated secretory phenotype (SASP). Senescent cells are more resistant to exposure due to their dormant state, polyploidy, and apoptosis evasion via senescent cell anti-apoptotic pathways, and they secrete various cytokines such as TGFβ1, TGFβ3, IL1β, IL-6, IL-8, CXCL1, CXCL2, and CXCL5 in addition to miRNA containing oncosomes that can promote tumor progression and confer subsequent therapy resistance [6][7]. Therapeutic approaches help to ablate senescent cells and were shown to improve treatment outcomes.

3. Diagnostic Biomarkers of Resistance

The detection of secretory factors holds potential as a diagnostic biomarker for tumor cell resistance status to predict a therapy response. miRNAs are a promising tool in biomarker development, as they are highly stable molecules in circulation [8]. Minimally invasive diagnostic approaches have been made using plasma levels of some miRNAs. miR-208a holds potential as a serum biomarker of NSCLC radioresistance, while other detected differentially expressed miRNAs await further investigation [9]. miR-29a-3p and miR-150-5p from blood were also found to be reflective of NSCLC radioresistance [10]. Eleven serum miRNAs were predictive of NSCLC patients’ resistance to RT [11]. Various circulatory exosome-shuttled miRNAs were also predictive of chemotherapy resistance status, as presented in a review [12]. Candidate secretory miRNAs involved with radio- and chemoresistance of NSCLC as an example are presented in Table 1, and they can be a starting point in the development of a minimally invasive diagnostic panel. Other diagnostic approaches are based on liquid and tumor biopsies, which can include miRNA from isolated oncosomes or other secretory factors, as well as intercellular miRNA analysis. Candidate intercellular miRNAs predictive of chemo- or radioresistance status have been proposed in numerous studies and are summarized in reviews [13][14].
Table 1. Secretory miRNA (exosomal and circulatory) involved in the radio- and chemoresistance of NSCLC. 

Type of Resistance

miRNA

Tyrosine kinase inhibitors

mir-BART14, mir-1469, mir-16-1, mir-196, mir-4791, mir-4796, mir-548aq, mir-72, mir-H19, mir-138-2, mir-153, mir-585, mir-4803, mir-744, mir-769 [15]

mir-184, mir-3913 [16]

mir-658, mir-564 [17]

mir-1468, mir-23 [18]

mir-136 [19]

mir-214 [20]

mir-210 [21]

mir-615 [22]

Cisplatin

mir-20a [23]

mir-193a [24]

mir-524 [25]

mir-4443 [26]

mir-1246 [27]

mir-425 [28]

mir-103a [29]

mir-1273a [30]

mir-100 [31]

IR

mir-196a [32]

mir-208a [9]

mir-29a, mir-150 [10]

The Human miRNA Disease Database [33] contains miRNA–disease associations, including cancer and others, from 19,280 scientific articles as of today. Cancer-associated miRNA biomarkers have been validated, and a “miRview-mets2” panel was created for the clinical identification of metastatic cancer origins, in addition to other clinical miRNA-based tests [34]. Creating a resistance status database would be helpful to current diagnostics in the clinic to select a more efficient treatment regimen.

3.1. Therapeutic Approaches

Since the resistance-mediating effects of oncosomes were characterized, the inhibition of their secretion, biogenesis, or uptake was attempted in combination with anticancer therapy that led to tumor sensitization and higher antineoplastic efficiency in multiple in vitro studies, as summarized in a review [12]. The administration of exosome inhibitors heparin and simvastatin can help alleviate the detrimental effects of the oncosome injection derived from resistant cells in mice [35][36]. Alkylation of TME also reduces oncosome release, and intraperitoneal injections of proton pump inhibitors in combination with chemotherapy in mice led to decreased plasma exosome levels; however, no differences in tumor weight were noted due to selected time intervals [37].
Upon the discovery of secretory regulatory RNA factors conveying chemo- and radioresistance, approaches were made to up- or downregulate them in vivo. Injections of exosomes containing mir-214 antagomir sensitized lung tumors in mice, pre-treated with oncosomes from gefitinib-resistant PC9 cells [20]. As an example, antisense oligonucleotide targeting allowed for the knockout of circITGB6 in vivo with intraperitoneal injections, and in combination with cisplatin treatment, it led to significantly lower ovarian circITGB6-transfected tumor size and increased survival in mice compared to cisplatin alone [1]. Knockdown of IL-11 in mice with lung cancer also led to better effects of cisplatin treatment [3]. Inhibition of a paracrine factor PAI-1 in mice with lung cancer by oral administration of tiplaxtitnin successfully sensitized tumors to radiation therapy and led to a significantly decreased tumor volume [5].
The animal studies discussed above provide a valuable model for the in vivo investigation of therapeutic opportunities and successful outcomes proceeding further to clinical trials. Preliminary investigations were carried out on cell cultures, while 3D cultures were also used to simulate tumor formation in vitro. Recent neo-organoid developments are a very promising treatment based on cell integration into a 3D scaffold with the following implantation into the body. Neo-organoid implantation with Matrigel-imbedded MSCs overexpressing IL-12 led to significantly better results, as compared to non-genetically modified MSCs with 67% of mice with breast cancer xenografts being completely tumor free 55 days after treatment [38].
Such approaches pave the way to abrogate the subset of resistance-acquiring cancer tumor cells via the acquisition of secreted factors. However, the question remains open as to how kill the resistant and CSCs that are also present as a subset of a heterogenous tumor in this model. Today, the major directions in CSC-targeted therapy research include immunotherapy, inhibition of key signaling pathways, inhibition of DNA repair, and awakening quiescent CSCs [39][40]. Studies of differentially expressed intracellular miRNAs between resistant and sensitive cancer cells point out the miRNA control of cancer cells’ response to IR or CT, and its direct manipulation can be used to sensitize the tumor prior to therapy. Table 2 presents a summary of the candidate deregulated miRNA of resistant NSCLC cells as an example of possible therapeutic targets.
Table 2. Most commonly differentially expressed miRNA of radio- and chemoresistant NSCLC cells. IR—ionizing radiation; EGFR-TKI—epidermal growth factor receptor tyrosine kinase inhibitors; 5FU—5-fluorouracil.
miRNA therapeutic approaches for the treatment of cancer are gaining attention, with some of them in clinical trials already [96][97][98]. Several biopharmaceutical companies are developing and implementing miRNA-based therapeutics and are trying to overcome the challenges associated with tumor tissue specificity, off-target activity due to miRNA pleiotropic nature, and toxicity with novel drug delivery systems and combinations with other medications [99][100]. Targeting resistance-conferring miRNA for tumor sensitization in combination with RT or CT also has a potential to overcome resistance and provide more satisfactory therapeutic results (Figure 1). One of the ways to implement it was attempted with intra-tumoral injections of exosomes derived from MSCs transfected with selected miRNAs to sensitize cancer cells to CT, as summarized in a review [101].
Figure 1. Development of tumor chemo- and radioresistance. Abbreviations: IR—ionizing radaiation; CT—chemotherapy; RT—radiotherapy; CRT—chemoradiotherapy.
Abolishment of senescent cells improves anti-cancer therapy results and can involve senolytic agents, such as chimeric antigen receptor T cells against uPAR marker and proteolysis-targeting chimera technology, in addition to other natural and targeted senolytic compounds that cause senescent cell death [102][103][104]. Senomorphic agents block SASP effects without causing senescent cell death [105]. Senotherapeutics are used as an adjuvant therapy to ablate senescent cells formed after CT or RT and lead to a better response in some patients; however, the treatment outcomes are not consistent [106].

References

  1. Li, H.; Luo, F.; Jiang, X.; Zhang, W.; Xiang, T.; Pan, Q.; Cai, L.; Zhao, J.; Weng, D.; Li, Y.; et al. CircITGB6 promotes ovarian cancer cisplatin resistance by resetting tumor-associated macrophage polarization toward the M2 phenotype. J. ImmunoTherapy Cancer 2022, 10, e004029.
  2. Zhao, M.; Xu, J.; Zhong, S.; Liu, Y.; Xiao, H.; Geng, L.; Liu, H. Expression profiles and potential functions of circular RNAs in extracellular vesicles isolated from radioresistant glioma cells. Oncol. Rep. 2019, 41, 1893–1900.
  3. Tao, L.; Huang, G.; Wang, R.; Pan, Y.; He, Z.; Chu, X.; Song, H.; Chen, L. Cancer-associated fibroblasts treated with cisplatin facilitates chemoresistance of lung adenocarcinoma through IL-11/IL-11R/STAT3 signaling pathway. Sci. Rep. 2016, 6, 38408.
  4. Yoshino, H.; Nawamaki, M.; Murakami, K.; Kashiwakura, I. Effects of irradiated cell conditioned medium on the response of human lung cancer cells to anticancer treatment in vitro. World Acad. Sci. J. 2019, 1, 92–97.
  5. Kang, J.; Kim, W.; Kwon, T.; Youn, H.; Kim, J.S.; Youn, B. Plasminogen activator inhibitor-1 enhances radioresistance and aggressiveness of non-small cell lung cancer cells. Oncotarget 2016, 7, 23961–23974.
  6. Chambers, C.R.; Ritchie, S.; Pereira, B.A.; Timpson, P. Overcoming the senescence-associated secretory phenotype (SASP): A complex mechanism of resistance in the treatment of cancer. Mol. Oncol. 2021, 15, 3242–3255.
  7. Hoare, M.; Ito, Y.; Kang, T.-W.; Weekes, M.P.; Matheson, N.J.; Patten, D.A.; Shetty, S.; Parry, A.J.; Menon, S.; Salama, R.; et al. NOTCH1 mediates a switch between two distinct secretomes during senescence. Nat. Cell Biol. 2016, 18, 979–992.
  8. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402.
  9. Tang, Y.; Cui, Y.; Li, Z.; Jiao, Z.; Zhang, Y.; He, Y.; Chen, G.; Zhou, Q.; Wang, W.; Zhou, X.; et al. Radiation-induced miR-208a increases the proliferation and radioresistance by targeting p21 in human lung cancer cells. J. Exp. Clin. Cancer Res. 2016, 35, 7.
  10. Dinh, T.-K.T.; Fendler, W.; Chałubińska-Fendler, J.; Acharya, S.S.; O’Leary, C.; Deraska, P.V.; D’Andrea, A.D.; Chowdhury, D.; Kozono, D. Circulating miR-29a and miR-150 correlate with delivered dose during thoracic radiation therapy for non-small cell lung cancer. Radiat. Oncol. 2016, 11, 61.
  11. Sun, Y.; Hawkins, P.G.; Bi, N.; Dess, R.T.; Tewari, M.; Hearn, J.W.D.; Hayman, J.A.; Kalemkerian, G.P.; Lawrence, T.S.; Ten Haken, R.K.; et al. Serum MicroRNA Signature Predicts Response to High-Dose Radiation Therapy in Locally Advanced Non-Small Cell Lung Cancer. Int. J. Radiat. Oncol. Biol. Phys. 2018, 100, 107–114.
  12. Mostafazadeh, M.; Samadi, N.; Kahroba, H.; Baradaran, B.; Haiaty, S.; Nouri, M. Potential roles and prognostic significance of exosomes in cancer drug resistance. Cell Biosci. 2021, 11, 1.
  13. Ahmad, P.; Sana, J.; Slavik, M.; Slampa, P.; Smilek, P.; Slaby, O. MicroRNAs Involvement in Radioresistance of Head and Neck Cancer. Dis. Markers 2017, 2017, 8245345.
  14. Long, L.; Zhang, X.; Bai, J.; Li, Y.; Wang, X.; Zhou, Y. Tissue-specific and exosomal miRNAs in lung cancer radiotherapy: From regulatory mechanisms to clinical implications. Cancer Manag. Res. 2019, 11, 4413–4424.
  15. Sorokin, M.; Kholodenko, R.; Grekhova, A.; Suntsova, M.; Pustovalova, M.; Vorobyeva, N.; Kholodenko, I.; Malakhova, G.; Garazha, A.; Nedoluzhko, A.; et al. Acquired resistance to tyrosine kinase inhibitors may be linked with the decreased sensitivity to X-ray irradiation. Oncotarget 2017, 9, 5111–5124.
  16. Li, X.; Chen, C.; Wang, Z.; Liu, J.; Sun, W.; Shen, K.; Lv, Y.; Zhu, S.; Zhan, P.; Lv, T.; et al. Elevated exosome-derived miRNAs predict osimertinib resistance in non-small cell lung cancer. Cancer Cell Int. 2021, 21, 428.
  17. Azuma, Y.; Yokobori, T.; Mogi, A.; Yajima, T.; Kosaka, T.; Iijima, M.; Shimizu, K.; Shirabe, K.; Kuwano, H. Cancer exosomal microRNAs from gefitinib-resistant lung cancer cells cause therapeutic resistance in gefitinib-sensitive cells. Surg. Today 2020, 50, 1099–1106.
  18. Janpipatkul, K.; Trachu, N.; Watcharenwong, P.; Panvongsa, W.; Worakitchanon, W.; Metheetrairut, C.; Oranratnachai, S.; Reungwetwattana, T.; Chairoungdua, A. Exosomal microRNAs as potential biomarkers for osimertinib resistance of non-small cell lung cancer patients. Cancer Biomark. 2021, 31, 281–294.
  19. Gu, G.; Hu, C.; Hui, K.; Zhang, H.; Chen, T.; Zhang, X.; Jiang, X. Exosomal miR-136-5p Derived from Anlotinib-Resistant NSCLC Cells Confers Anlotinib Resistance in Non-Small Cell Lung Cancer Through Targeting PPP2R2A. Int. J. Nanomed. 2021, 16, 6329–6343.
  20. Zhang, Y.; Li, M.; Hu, C. Exosomal transfer of miR-214 mediates gefitinib resistance in non-small cell lung cancer. Biochem. Biophys. Res. Commun. 2018, 507, 457–464.
  21. Hisakane, K.; Seike, M.; Sugano, T.; Yoshikawa, A.; Matsuda, K.; Takano, N.; Takahashi, S.; Noro, R.; Gemma, A. Exosome-derived miR-210 involved in resistance to osimertinib and epithelial–mesenchymal transition in EGFR mutant non-small cell lung cancer cells. Thorac. Cancer 2021, 12, 1690–1698.
  22. Pan, R.; Zhou, H. Exosomal Transfer of lncRNA H19 Promotes Erlotinib Resistance in Non-Small Cell Lung Cancer via miR-615-3p/ATG7 Axis. Cancer Manag. Res. 2020, 12, 4283–4297.
  23. Shi, L.; Zhu, W.; Huang, Y.; Zhuo, L.; Wang, S.; Chen, S.; Zhang, B.; Ke, B. Cancer-associated fibroblast-derived exosomal microRNA-20a suppresses the PTEN/PI3K-AKT pathway to promote the progression and chemoresistance of non-small cell lung cancer. Clin. Transl. Med. 2022, 12, e989.
  24. Wu, H.; Mu, X.; Liu, L.; Wu, H.; Hu, X.; Chen, L.; Liu, J.; Mu, Y.; Yuan, F.; Liu, W.; et al. Bone marrow mesenchymal stem cells-derived exosomal microRNA-193a reduces cisplatin resistance of non-small cell lung cancer cells via targeting LRRC1. Cell Death Dis. 2020, 11, 801.
  25. Xie, H.; Yao, J.; Wang, Y.; Ni, B. Exosome-transmitted circVMP1 facilitates the progression and cisplatin resistance of non-small cell lung cancer by targeting miR-524-5p-METTL3/SOX2 axis. Drug Deliv. 2022, 29, 1257–1271.
  26. Song, Z.; Jia, G.; Ma, P.; Cang, S. Exosomal miR-4443 promotes cisplatin resistance in non-small cell lung carcinoma by regulating FSP1 m6A modification-mediated ferroptosis. Life Sci. 2021, 276, 119399.
  27. Liu, Z.; Zhao, W.; Yang, R. MiR-1246 is responsible for lung cancer cells-derived exosomes-mediated promoting effects on lung cancer stemness via targeting TRIM17. Environ. Toxicol. 2022, 37, 2651–2659.
  28. Kilic, S.; Lezaja, A.; Gatti, M.; Bianco, E.; Michelena, J.; Imhof, R.; Altmeyer, M. Phase separation of 53 BP 1 determines liquid-like behavior of DNA repair compartments. EMBO J. 2019, 38, e101379.
  29. Wang, H.; Huang, H.; Wang, L.; Liu, Y.; Wang, M.; Zhao, S.; Lu, G.; Kang, X. Cancer-associated fibroblasts secreted miR-103a-3p suppresses apoptosis and promotes cisplatin resistance in non-small cell lung cancer. Aging 2021, 13, 14456–14468.
  30. Zhao, X.; Li, M.; Dai, X.; Yang, Y.; Peng, Y.; Xu, C.; Dai, N.; Wang, D. Downregulation of exosomal miR-1273a increases cisplatin resistance of non-small cell lung cancer by upregulating the expression of syndecan binding protein. Oncol. Rep. 2020, 44, 2165–2173.
  31. Qin, X.; Yu, S.; Zhou, L.; Shi, M.; Hu, Y.; Xu, X.; Shen, B.; Liu, S.; Yan, D.; Feng, J. Cisplatin-resistant lung cancer cell–derived exosomes increase cisplatin resistance of recipient cells in exosomal miR-100–5p-dependent manner. Int. J. Nanomed. 2017, 12, 3721–3733.
  32. Yao, F.; Shi, W.; Fang, F.; Lv, M.Y.; Xu, M.; Wu, S.Y.; Huang, C.L. Exosomal miR-196a-5p enhances radioresistance in lung cancer cells by downregulating NFKBIA. Kaohsiung J. Med. Sci. 2023, 39, 554–564.
  33. Cui, C.; Zhong, B.; Fan, R.; Cui, Q. HMDD v4.0: A database for experimentally supported human microRNA-disease associations. Nucleic Acids Res. 2023, gkad717.
  34. Hydbring, P.; Badalian-Very, G. Clinical applications of microRNAs. F1000Research 2013, 2, 136.
  35. Mrowczynski, O.D.; Madhankumar, A.B.; Sundstrom, J.M.; Zhao, Y.; Kawasawa, Y.I.; Slagle-Webb, B.; Mau, C.; Payne, R.A.; Rizk, E.B.; Zacharia, B.E.; et al. Exosomes impact survival to radiation exposure in cell line models of nervous system cancer. Oncotarget 2018, 9, 36083–36101.
  36. Tang, C.-H.; Sento, S.; Sasabe, E.; Yamamoto, T. Application of a Persistent Heparin Treatment Inhibits the Malignant Potential of Oral Squamous Carcinoma Cells Induced by Tumor Cell-Derived Exosomes. PLoS ONE 2016, 11, e0148454.
  37. Lebedeva, I.V.; Federici, C.; Petrucci, F.; Caimi, S.; Cesolini, A.; Logozzi, M.; Borghi, M.; D’Ilio, S.; Lugini, L.; Violante, N.; et al. Exosome Release and Low pH Belong to a Framework of Resistance of Human Melanoma Cells to Cisplatin. PLoS ONE 2014, 9, e88193.
  38. Eliopoulos, N.; Francois, M.r.; Boivin, M.-N.l.; Martineau, D.; Galipeau, J. Neo-Organoid of Marrow Mesenchymal Stromal Cells Secreting Interleukin-12 for Breast Cancer Therapy. Cancer Res. 2008, 68, 4810–4818.
  39. Huang, T.; Song, X.; Xu, D.; Tiek, D.; Goenka, A.; Wu, B.; Sastry, N.; Hu, B.; Cheng, S.-Y. Stem cell programs in cancer initiation, progression, and therapy resistance. Theranostics 2020, 10, 8721–8743.
  40. Schulz, A.; Meyer, F.; Dubrovska, A.; Borgmann, K. Cancer Stem Cells and Radioresistance: DNA Repair and Beyond. Cancers 2019, 11, 862.
  41. Zhang, C.-C.; Li, Y.; Feng, X.-Z.; Li, D.-B. Circular RNA circ_0001287 inhibits the proliferation, metastasis, and radiosensitivity of non-small cell lung cancer cells by sponging microRNA miR-21 and up-regulating phosphatase and tensin homolog expression. Bioengineered 2021, 12, 414–425.
  42. Zhang, J.; Zhang, C.; Hu, L.; He, Y.; Shi, Z.; Tang, S.; Chen, Y. Abnormal Expression of miR-21 and miR-95 in Cancer Stem-Like Cells is Associated with Radioresistance of Lung Cancer. Cancer Investig. 2015, 33, 165–171.
  43. Sun, Y.; Liu, W.; Zhao, Q.; Zhang, R.; Wang, J.; Pan, P.; Shang, H.; Liu, C.; Wang, C. Down-Regulating the Expression of miRNA-21 Inhibits the Glucose Metabolism of A549/DDP Cells and Promotes Cell Death Through the PI3K/AKT/mTOR/HIF-1α Pathway. Front. Oncol. 2021, 11, 653596.
  44. Gao, W.; Lu, X.; Liu, L.; Xu, J.; Feng, D.; Shu, Y. MiRNA-21. Cancer Biol. Ther. 2014, 13, 330–340.
  45. Dong, Z.; Ren, L.I.; Lin, L.I.; Li, J.; Huang, Y.; Li, J. Effect of microRNA-21 on multidrug resistance reversal in A549/DDP human lung cancer cells. Mol. Med. Rep. 2015, 11, 682–690.
  46. Li, B.; Ren, S.; Li, X.; Wang, Y.; Garfield, D.; Zhou, S.; Chen, X.; Su, C.; Chen, M.; Kuang, P.; et al. MiR-21 overexpression is associated with acquired resistance of EGFR-TKI in non-small cell lung cancer. Lung Cancer 2014, 83, 146–153.
  47. Wang, S.; Su, X.; Bai, H.; Zhao, J.; Duan, J.; An, T.; Zhuo, M.; Wang, Z.; Wu, M.; Li, Z.; et al. Identification of plasma microRNA profiles for primary resistance to EGFR-TKIs in advanced non-small cell lung cancer (NSCLC) patients with EGFR activating mutation. J. Hematol. Oncol. 2015, 8, 127.
  48. Huang, W.-C.; Yadav, V.K.; Cheng, W.-H.; Wang, C.-H.; Hsieh, M.-S.; Huang, T.-Y.; Lin, S.-F.; Yeh, C.-T.; Kuo, K.-T. The MEK/ERK/miR-21 Signaling Is Critical in Osimertinib Resistance in EGFR-Mutant Non-Small Cell Lung Cancer Cells. Cancers 2021, 13, 6005.
  49. Lee, J.W.; Shen, H.; Zhu, F.; Liu, J.; Xu, T.; Pei, D.; Wang, R.; Qian, Y.; Li, Q.; Wang, L.; et al. Alteration in Mir-21/PTEN Expression Modulates Gefitinib Resistance in Non-Small Cell Lung Cancer. PLoS ONE 2014, 9, e103305.
  50. Ding, S.; Zheng, Y.; Xu, Y.; Zhao, X.; Zhong, C. MiR-21/PTEN signaling modulates the chemo-sensitivity to 5-fluorouracil in human lung adenocarcinoma A549 cells. Int. J. Clin. Exp. Pathol. 2019, 12, 2339–2352.
  51. Su, C.; Cheng, X.; Li, Y.; Han, Y.; Song, X.; Yu, D.; Cao, X.; Liu, Z. MiR-21 improves invasion and migration of drug-resistant lung adenocarcinoma cancer cell and transformation of EMT through targetingHBP1. Cancer Med. 2018, 7, 2485–2503.
  52. Zhang, Y.-Q.; Chen, R.-L.; Shang, L.-Q.; Yang, S.-M. Nicotine-induced miR-21-3p promotes chemoresistance in lung cancer by negatively regulating FOXO3a. Oncol. Lett. 2022, 24, 260.
  53. Liu, Z.-L.; Wang, H.; Liu, J.; Wang, Z.-X. MicroRNA-21 (miR-21) expression promotes growth, metastasis, and chemo- or radioresistance in non-small cell lung cancer cells by targeting PTEN. Mol. Cell. Biochem. 2012, 372, 35–45.
  54. Li, H.; Zhao, S.; Chen, X.; Feng, G.; Chen, Z.; Fan, S. MiR-145 modulates the radiosensitivity of non-small cell lung cancer cells by suppression of TMOD3. Carcinogenesis 2022, 43, 288–296.
  55. Zhang, H.; Luo, Y.; Xu, W.; Li, K.; Liao, C. Silencing long intergenic non-coding RNA 00707 enhances cisplatin sensitivity in cisplatin-resistant non-small-cell lung cancer cells by sponging miR-145. Oncol. Lett. 2019, 18, 6261–6268.
  56. Bar, J.; Gorn-Hondermann, I.; Moretto, P.; Perkins, T.J.; Niknejad, N.; Stewart, D.J.; Goss, G.D.; Dimitroulakos, J. miR Profiling Identifies Cyclin-Dependent Kinase 6 Downregulation as a Potential Mechanism of Acquired Cisplatin Resistance in Non–Small-Cell Lung Carcinoma. Clin. Lung Cancer 2015, 16, e121–e129.
  57. Chang, Y.-F.; Lim, K.-H.; Chiang, Y.-W.; Sie, Z.-L.; Chang, J.; Ho, A.-S.; Cheng, C.-C. STAT3 induces G9a to exacerbate HER3 expression for the survival of epidermal growth factor receptor-tyrosine kinase inhibitors in lung cancers. BMC Cancer 2019, 19, 959.
  58. Yu, C.; Li, B.; Wang, J.; Zhang, Z.; Li, S.; Lei, S.; Wang, Q. miR-145-5p Modulates Gefitinib Resistance by Targeting NRAS and MEST in Non-Small Cell Lung Cancer. Ann. Clin. Lab. Sci. 2021, 51, 625–637.
  59. Wang, Y.; Lian, Y.M.; Ge, C.Y. MiR-145 changes sensitivity of non-small cell lung cancer to gefitinib through targeting ADAM19. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 5831–5839.
  60. Pan, Y.; Chen, J.; Tao, L.; Zhang, K.; Wang, R.; Chu, X.; Chen, L. Long noncoding RNA ROR regulates chemoresistance in docetaxel-resistant lung adenocarcinoma cells via epithelial mesenchymal transition pathway. Oncotarget 2017, 8, 33144–33158.
  61. Chang, W.-W.; Wang, B.-Y.; Chen, S.-H.; Chien, P.-J.; Sheu, G.-T.; Lin, C.-H. miR-145-5p Targets Sp1 in Non-Small Cell Lung Cancer Cells and Links to BMI1 Induced Pemetrexed Resistance and Epithelial–Mesenchymal Transition. Int. J. Mol. Sci. 2022, 23, 15352.
  62. Guan, X.; Guan, Y. miR-145-5p attenuates paclitaxel resistance and suppresses the progression in drug-resistant breast cancer cell lines. Neoplasma 2020, 67, 972–981.
  63. Zheng, F.; Xu, R. CircPVT1 contributes to chemotherapy resistance of lung adenocarcinoma through miR-145-5p/ABCC1 axis. Biomed. Pharmacother. 2020, 124, 109828.
  64. Fukuda, K.; Takeuchi, S.; Arai, S.; Katayama, R.; Nanjo, S.; Tanimoto, A.; Nishiyama, A.; Nakagawa, T.; Taniguchi, H.; Suzuki, T.; et al. Epithelial-to-Mesenchymal Transition Is a Mechanism of ALK Inhibitor Resistance in Lung Cancer Independent of ALK Mutation Status. Cancer Res. 2019, 79, 1658–1670.
  65. Gao, H.-X.; Yan, L.; Li, C.; Zhao, L.-M.; Liu, W. miR-200c regulates crizotinib-resistant ALK-positive lung cancer cells by reversing epithelial-mesenchymal transition via targeting ZEB1. Mol. Med. Rep. 2016, 14, 4135–4143.
  66. Fukuda, K.; Takeuchi, S.; Arai, S.; Kita, K.; Tanimoto, A.; Nishiyama, A.; Yano, S. Glycogen synthase kinase-3 inhibition overcomes epithelial-mesenchymal transition-associated resistance to osimertinib in EGFR-mutant lung cancer. Cancer Sci. 2020, 111, 2374–2384.
  67. Zhou, G.; Zhang, F.; Guo, Y.; Huang, J.; Xie, Y.; Yue, S.; Chen, M.; Jiang, H.; Li, M. miR-200c enhances sensitivity of drug-resistant non-small cell lung cancer to gefitinib by suppression of PI3K/Akt signaling pathway and inhibites cell migration via targeting ZEB1. Biomed. Pharmacother. 2017, 85, 113–119.
  68. Zhao, Y.-f.; Han, M.-l.; Xiong, Y.-j.; Wang, L.; Fei, Y.; Shen, X.; Zhu, Y.; Liang, Z.-q. A miRNA-200c/cathepsin L feedback loop determines paclitaxel resistance in human lung cancer A549 cells in vitro through regulating epithelial–mesenchymal transition. Acta Pharmacol. Sin. 2017, 39, 1034–1047.
  69. Zhu, W.; Xu, H.; Zhu, D.; Zhi, H.; Wang, T.; Wang, J.; Jiang, B.; Shu, Y.; Liu, P. miR-200bc/429 cluster modulates multidrug resistance of human cancer cell lines by targeting BCL2 and XIAP. Cancer Chemother. Pharmacol. 2011, 69, 723–731.
  70. Zhao, J.; Fu, W.; Liao, H.; Dai, L.; Jiang, Z.; Pan, Y.; Huang, H.; Mo, Y.; Li, S.; Yang, G.; et al. The regulatory and predictive functions of miR-17 and miR-92 families on cisplatin resistance of non-small cell lung cancer. BMC Cancer 2015, 15, 731.
  71. Fröhlich, H.; Jiang, Z.; Yin, J.; Fu, W.; Mo, Y.; Pan, Y.; Dai, L.; Huang, H.; Li, S.; Zhao, J. miRNA 17 Family Regulates Cisplatin-Resistant and Metastasis by Targeting TGFbetaR2 in NSCLC. PLoS ONE 2014, 9, e94639.
  72. Chatterjee, A.; Chattopadhyay, D.; Chakrabarti, G. miR-16 targets Bcl-2 in paclitaxel-resistant lung cancer cells and overexpression of miR-16 along with miR-17 causes unprecedented sensitivity by simultaneously modulating autophagy and apoptosis. Cell. Signal. 2015, 27, 189–203.
  73. Mari, B.; Chatterjee, A.; Chattopadhyay, D.; Chakrabarti, G. miR-17-5p Downregulation Contributes to Paclitaxel Resistance of Lung Cancer Cells through Altering Beclin1 Expression. PLoS ONE 2014, 9, e95716.
  74. Yin, J.; Hu, W.; Pan, L.; Fu, W.; Dai, L.; Jiang, Z.; Zhang, F.; Zhao, J. let-7 and miR-17 promote self-renewal and drive gefitinib resistance in non-small cell lung cancer. Oncol. Rep. 2019, 42, 495–508.
  75. Gong, J.; He, L.; Ma, J.; Zhang, J.; Wang, L.; Wang, J. The relationship between miR-17-5p, miR-92a, and let-7b expression with non-small cell lung cancer targeted drug resistance. J. BUON 2017, 22, 454–461.
  76. Yu, G.; Zhong, N.; Chen, G.; Huang, B.; Wu, S. Downregulation of PEBP4, a target of miR-34a, sensitizes drug-resistant lung cancer cells. Tumor Biol. 2014, 35, 10341–10349.
  77. Luo, S.; Shen, M.; Chen, H.; Li, W.; Chen, C. Long non-coding RNA TP73-AS1 accelerates the progression and cisplatin resistance of non-small cell lung cancer by upregulating the expression of TRIM29 via competitively targeting microRNA-34a-5p. Mol. Med. Rep. 2020, 22, 3822–3832.
  78. Yang, X.; Sun, Q.; Song, Y.; Li, W.; Falzone, L. circHUWE1 Exerts an Oncogenic Role in Inducing DDP-Resistant NSCLC Progression Depending on the Regulation of miR-34a-5p/TNFAIP8. Int. J. Genom. 2021, 2021, 3997045.
  79. Zhou, J.-Y.; Chen, X.; Zhao, J.; Bao, Z.; Chen, X.; Zhang, P.; Liu, Z.-F.; Zhou, J.-Y. MicroRNA-34a overcomes HGF-mediated gefitinib resistance in EGFR mutant lung cancer cells partly by targeting MET. Cancer Lett. 2014, 351, 265–271.
  80. Xiong, R.; Sun, X.x.; Wu, H.r.; Xu, G.w.; Wang, G.x.; Sun, X.h.; Xu, M.q.; Xie, M.r. Mechanism research of miR-34a regulates Axl in non-small-cell lung cancer with gefitinib-acquired resistance. Thorac. Cancer 2019, 11, 156–165.
  81. Li, J.; Li, S.; Chen, Z.; Wang, J.; Chen, Y.; Xu, Z.; Jin, M.; Yu, W. miR-326 reverses chemoresistance in human lung adenocarcinoma cells by targeting specificity protein 1. Tumor Biol. 2016, 37, 13287–13294.
  82. Wei, J.; Meng, G.; Wu, J.; Wang, Y.; Zhang, Q.; Dong, T.; Bao, J.; Wang, C.; Zhang, J. MicroRNA-326 impairs chemotherapy resistance in non small cell lung cancer by suppressing histone deacetylase SIRT1-mediated HIF1α and elevating VEGFA. Bioengineered 2022, 13, 5685–5699.
  83. Wu, Y.; Cheng, K.; Liang, W.; Wang, X. lncRNA RPPH1 promotes non-small cell lung cancer progression through the miR-326/WNT2B axis. Oncol. Lett. 2020, 20, 105.
  84. Dong, C.; Yang, L.; Zhao, G. Circ-PGAM1 Enhances Matrine Resistance of Non-Small Cell Lung Cancer via the miR-326/CXCR5 Axis. Cancer Biother. Radiopharm. 2022.
  85. Zheng, Y.; Guo, Z.; Li, Y. Long non-coding RNA prostate cancer-associated transcript 6 inhibited gefitinib sensitivity of non-small cell lung cancer by serving as a competing endogenous RNA of miR-326 to up-regulate interferon-alpha receptor 2. Bioengineered 2022, 13, 3785–3796.
  86. Tang, W.; Yu, X.; Zeng, R.; Chen, L. LncRNA-ATB Promotes Cisplatin Resistance in Lung Adenocarcinoma Cells by Targeting the miR-200a/β-Catenin Pathway. Cancer Manag. Res. 2020, 12, 2001–2014.
  87. Liu, X.; Chen, L.; Wang, T. Overcoming cisplatin resistance of human lung cancer by sinomenine through targeting the miR-200a-3p-GLS axis. J. Chemother. 2022, 35, 357–366.
  88. Zhen, Q.; Liu, J.; Gao, L.; Liu, J.; Wang, R.; Chu, W.; Zhang, Y.; Tan, G.; Zhao, X.; Lv, B. MicroRNA-200a Targets EGFR and c-Met to Inhibit Migration, Invasion, and Gefitinib Resistance in Non-Small Cell Lung Cancer. Cytogenet. Genome Res. 2015, 146, 1–8.
  89. Nishijima, N.; Seike, M.; Soeno, C.; Chiba, M.; Miyanaga, A.; Noro, R.; Sugano, T.; Matsumoto, M.; Kubota, K.; Gemma, A. miR-200/ZEB axis regulates sensitivity to nintedanib in non-small cell lung cancer cells. Int. J. Oncol. 2016, 48, 937–944.
  90. Rastogi, I.; Rajanna, S.; Webb, A.; Chhabra, G.; Foster, B.; Webb, B.; Puri, N. Mechanism of c-Met and EGFR tyrosine kinase inhibitor resistance through epithelial mesenchymal transition in non-small cell lung cancer. Biochem. Biophys. Res. Commun. 2016, 477, 937–944.
  91. Chen, J.; Liu, X.; Xu, Y.; Zhang, K.; Huang, J.; Pan, B.; Chen, D.; Cui, S.; Song, H.; Wang, R.; et al. TFAP2C-Activated MALAT1 Modulates the Chemoresistance of Docetaxel-Resistant Lung Adenocarcinoma Cells. Mol. Ther.-Nucleic Acids 2019, 14, 567–582.
  92. Jiang, M.; Qi, F.; Zhang, K.; Zhang, X.; Ma, J.; Xia, S.; Chen, L.; Yu, Z.; Chen, J.; Chen, D. MARCKSL1–2 reverses docetaxel-resistance of lung adenocarcinoma cells by recruiting SUZ12 to suppress HDAC1 and elevate miR-200b. Mol. Cancer 2022, 21, 150.
  93. Pan, B.; Feng, B.; Chen, Y.; Huang, G.; Wang, R.; Chen, L.; Song, H. MiR-200b regulates autophagy associated with chemoresistance in human lung adenocarcinoma. Oncotarget 2015, 6, 32805–32820.
  94. Chen, D.-Q.; Pan, B.-Z.; Huang, J.-Y.; Zhang, K.; Cui, S.-Y.; De, W.; Wang, R.; Chen, L.-B. HDAC 1/4-mediated silencing of microRNA-200b promotes chemoresistance in human lung adenocarcinoma cells. Oncotarget 2014, 5, 3333–3349.
  95. Feng, B.; Wang, R.; Song, H.-Z.; Chen, L.-B. MicroRNA-200b reverses chemoresistance of docetaxel-resistant human lung adenocarcinoma cells by targeting E2F3. Cancer 2012, 118, 3365–3376.
  96. Hong, D.S.; Kang, Y.-K.; Borad, M.; Sachdev, J.; Ejadi, S.; Lim, H.Y.; Brenner, A.J.; Park, K.; Lee, J.-L.; Kim, T.-Y.; et al. Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumours. Br. J. Cancer 2020, 122, 1630–1637.
  97. van Zandwijk, N.; Pavlakis, N.; Kao, S.C.; Linton, A.; Boyer, M.J.; Clarke, S.; Huynh, Y.; Chrzanowska, A.; Fulham, M.J.; Bailey, D.L.; et al. Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: A first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncol. 2017, 18, 1386–1396.
  98. Qi, Y.; Huang, Y.; Pang, L.; Gu, W.; Wang, N.; Hu, J.; Cui, X.; Zhang, J.; Zhao, J.; Liu, C.; et al. Prognostic value of the MicroRNA-29 family in multiple human cancers: A meta-analysis and systematic review. Clin. Exp. Pharmacol. Physiol. 2017, 44, 441–454.
  99. Chakraborty, C.; Sharma, A.R.; Sharma, G.; Lee, S.-S. Therapeutic advances of miRNAs: A preclinical and clinical update. J. Adv. Res. 2021, 28, 127–138.
  100. To, K.K.W.; Fong, W.; Tong, C.W.S.; Wu, M.; Yan, W.; Cho, W.C.S. Advances in the discovery of microRNA-based anticancer therapeutics: Latest tools and developments. Expert Opin. Drug Discov. 2019, 15, 63–83.
  101. Lin, Z.; Wu, Y.; Xu, Y.; Li, G.; Li, Z.; Liu, T. Mesenchymal stem cell-derived exosomes in cancer therapy resistance: Recent advances and therapeutic potential. Mol. Cancer 2022, 21, 179.
  102. Amor, C.; Feucht, J.; Leibold, J.; Ho, Y.-J.; Zhu, C.; Alonso-Curbelo, D.; Mansilla-Soto, J.; Boyer, J.A.; Li, X.; Giavridis, T.; et al. Senolytic CAR T cells reverse senescence-associated pathologies. Nature 2020, 583, 127–132.
  103. He, Y.; Zhang, X.; Chang, J.; Kim, H.-N.; Zhang, P.; Wang, Y.; Khan, S.; Liu, X.; Zhang, X.; Lv, D.; et al. Using proteolysis-targeting chimera technology to reduce navitoclax platelet toxicity and improve its senolytic activity. Nat. Commun. 2020, 11, 1996.
  104. Hu, L.; Li, H.; Zi, M.; Li, W.; Liu, J.; Yang, Y.; Zhou, D.; Kong, Q.-P.; Zhang, Y.; He, Y. Why Senescent Cells Are Resistant to Apoptosis: An Insight for Senolytic Development. Front. Cell Dev. Biol. 2022, 10, 822816.
  105. Zhang, L.; Pitcher, L.E.; Prahalad, V.; Niedernhofer, L.J.; Robbins, P.D. Targeting cellular senescence with senotherapeutics: Senolytics and senomorphics. FEBS J. 2022, 290, 1362–1383.
  106. Short, S.; Fielder, E.; Miwa, S.; von Zglinicki, T. Senolytics and senostatics as adjuvant tumour therapy. EBioMedicine 2019, 41, 683–692.
More
Information
Subjects: Cell Biology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 102
Revisions: 2 times (View History)
Update Date: 13 Dec 2023
1000/1000
Video Production Service