Araştırma Makalesi


DOI :10.26650/EurJBiol.2024.1531120   IUP :10.26650/EurJBiol.2024.1531120    Tam Metin (PDF)

Notch and Hedgehog Signalling Axis Drive Senescence in HER2-Positive Breast Cancer Resistant to Trastuzumab

Asiye Büşra Boz Erİdris Er

Objective: Cellular senescence halts the proliferation of damaged or preneoplastic cells, playing a vital role in cancer control. In HER2-positive breast cancer, resistance to trastuzumab, a HER2-targeted monoclonal antibody, remains a significant obstacle. Although the trastuzumab and cilengitide combination reduces stemness and epithelial-mesenchymal transition, its effect on senescence remains unclear. Additionally, inhibiting the Notch and Hedgehog pathways can induce senescence by impairing proliferation, stemness, and cell cycle progression, making them promising therapeutic targets. This study aimed to evaluate the effect of trastuzumab/cilengitide on cellular senescence in HER2-positive trastuzumab-resistant breast cancer cells and to elucidate the roles of Notch and Hedgehog signalling in this process.

Materials and Methods:: HER2-positive breast cancer cell lines HCC1954 and SKBR3, along with their trastuzumab-resistant variants, were treated with trastuzumab, cilengitide, or both. Senescence markers were assessed by real-time PCR. Notch and Hedgehog pathway activity was evaluated, with additional experiments using specific inhibitors Fli06 (Notch) and GANT61 (Hedgehog).

Results: The trastuzumab-cilengitide combination significantly upregulated senescence markers relative to monotherapy. This response was associated with a marked decrease in Notch and Hedgehog pathway activity. Further combined inhibition of these pathways enhanced senescence marker expression, underscoring their involvement in drug-induced senescence.

Conclusion: The trastuzumab-cilengitide combination induces senescence in trastuzumab-resistant HER2-positive breast cancer cells, potentially through Notch and Hedgehog inhibition. These findings support targeting senescence pathways as a novel strategy to overcome trastuzumab resistance and improve therapeutic outcomes. Further research is warranted to assess the clinical potential of such combination therapies.

Anahtar Kelimeler: HER2-positiveSenescenceNotchHedgehog

PDF Görünüm

Referanslar

  • Bousset L, Gil J. Targeting senescence as an anticancer therapy. Mol Oncol. 2022;16(21):3855-3880. google scholar
  • McHugh D, Gil J. Senescence and aging: Causes, consequences, and therapeutic avenues. J Cell Biol. 2018;217(1):65-77. google scholar
  • Boz Er AB. Integrin beta3 reprogramming stemness in HER2-positive breast cancer cell lines. Biology (Basel). 2024;13(6). doi:10.3390/biology13060429 google scholar
  • Boz Er AB, Er I. Targeting ITG3 to overcome trastuzumab re-sistance through epithelial-mesenchymal transition regulation in HER2-positive breast cancer. Int J Mol Sci. 2024;25(16):8640. doi: 10.3390/ijms25168640 google scholar
  • Pan X, Yi M, Liu C, et al. Cilengitide, an alphavbeta3-integrin inhibitor, enhances the efficacy of anti-programmed cell death-1 therapy in a murine melanoma model. Bioengineered. 2022;13(2):4557-4572. google scholar
  • Pang X, He X, Qiu Z, et al. Targeting integrin pathways: Mech-anisms and advances in therapy. Signal Transduct Target Ther. 2023;8(1):1. doi:10.1038/s41392-022-01259-6 google scholar
  • Kitsugi K, Noritake H, Matsumoto M, et al. Inhibition of inte-grin binding to ligand arg-gly-asp motif induces AKT-mediated cellular senescence in hepatic stellate cells. Mol Cell Biochem. 2023;479(10):2697-2710. google scholar
  • Fujita M, Sasada M, Eguchi M, et al. Induction of cellular senes-cence in fibroblasts through beta1-integrin activation by tenascin-C-derived peptide and its protumor effect. Am J Cancer Res. 2021;11(9):4364-4379. google scholar
  • Rapisarda V, Borghesan M, Miguela V, et al. Integrin beta 3 regulates cellular senescence by activating the TGF-beta pathway. Cell Rep. 2017;18(10):2480-2493. google scholar
  • Franovic A, Elliott KC, Seguin L, Camargo MF, Weis SM, Cheresh DA. Glioblastomas require integrin alphavbeta3/PAK4 signaling to escape senescence. Cancer Res. 2015;75(21):4466-4473. google scholar
  • Wang B, Han J, Elisseeff JH, Demaria M. The senescence-associated secretory phenotype and its physiological and pathological implications. Nat Rev Mol Cell Biol. 2024. doi:10.1038/s41580-024-00727-x google scholar
  • Borah A, Kumar DS. Chapter 8 - Targeting the Hedgehog and Notch Signaling Pathways in Cancer Stem Cells. In: Dammacco F, Silvestris F, eds. Oncogenomics. Academic Press; 2019:103-120. google scholar
  • Yoshida Y, Hayashi Y, Suda M, et al. Notch signal-ing regulates the lifespan of vascular endothelial cells via a p16-dependent pathway. PLoS One. 2014;9(6):e100359. doi:10.1371/journal.pone.0100359 google scholar
  • Lainez-Gonzalez D, Serrano-Lopez J, Alonso-Dominguez JM. Understanding the Notch signaling pathway in acute myeloid leukemia stem cells: From Hematopoiesis to Neoplasia. Cancers (Basel). 2022;14(6). doi:10.3390/cancers14061459 google scholar
  • Zou Y, Wu S, Hu Q, et al. Sonic hedgehog restrains the ubiquitin-dependent degradation of SP1 to inhibit neuronal/glial senes-cence associated phenotypes in chemotherapy-induced periph-eral neuropathy via the TRIM25-CXCL13 axis. J Adv Res. 2024. doi:10.1016/j.jare.2024.03.006 google scholar
  • Cho A, Park SR, Kim SR, et al. An endogenous anti-aging factor, sonic hedgehog, suppresses endometrial stem cell aging through SERPINB2. Mol Ther. 2019;27(7):1286-1298. google scholar
  • Kumari R, Jat P. Mechanisms of cellular senescence: Cell cycle arrest and senescence associated secretory phenotype. Front Cell Dev Biol. 2021;9:645593. doi:10.3389/fcell.2021.645593 google scholar
  • Swain SM, Shastry M, Hamilton E. Targeting HER2-positive breast cancer: Advances and future directions. Nat Rev Drug Discov. 2023;22(2):101-126. google scholar
  • Takebe N, Miele L, Harris PJ, et al. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nat Rev Clin Oncol. 2015;12(8):445-464. doi:10.1038/nrclinonc.2015.61 google scholar
  • Zagozdzon R, Gallagher WM, Crown J. Truncated HER2: Im-plications for HER2-targeted therapeutics. Drug Discov Today. 2011;16(17-18):810-816. doi:10.1016/j.drudis.2011.06.003 google scholar
  • Prasad D, Baldelli E, Blais EM, et al. Functional activation of the AKT-mTOR signalling axis in a real-world metastatic breast cancer cohort. Br J Cancer. 2024;131:1543-1554 google scholar
  • Khan SU, Fatima K, Aisha S, Malik F. Unveiling the mechanisms and challenges of cancer drug resistance. Cell Commun Signal. 2024;22(1):109. doi:10.1186/s12964-023-01302-1 google scholar
  • Maass KF, Kulkarni C, Betts AM, Wittrup KD. Determina-tion of cellular processing rates for a trastuzumab-maytansinoid antibody-drug conjugate (ADC) highlights key parameters for ADC design. AAPS J. 2016;18(3):635-646. google scholar
  • Ali K, Nabeel M, Mohsin F, et al. Recent developments in tar-geting breast cancer stem cells (BCSCs): A descriptive review of therapeutic strategies and emerging therapies. Med Oncol. 2024;41(5):112. doi:10.1007/s12032-024-02347-z google scholar
  • Duro-Sanchez S, Nadal-Serrano M, Lalinde-Gutierrez M, et al. Therapy-induced senescence enhances the efficacy of HER2-targeted antibody-drug conjugates in breast cancer. Cancer Res. 2022;82(24):4670-4679. google scholar
  • Billimoria R, Bhatt P. Senescence in cancer: Advances in detection and treatment modalities. Biochem Pharmacol. 2023;215:115739. doi:10.1016/j.bcp.2023.115739 google scholar
  • Di Micco R, Krizhanovsky V, Baker D, d’Adda di Fagagna F. Cellular senescence in ageing: from mechanisms to thera- peutic opportunities. Nat Rev Mol Cell Biol. 2021;22(2):75-95. doi:10.1038/s41580-020-00314-w google scholar
  • Nowell CS, Radtke F. Notch as a tumour suppressor. Nat Rev Cancer. 2017;17(3):145-159. google scholar
  • Andersson ER, Sandberg R, Lendahl U. Notch signaling: Simplicity in design, versatility in function. Development. 2011;138(17):3593-3612. google scholar

Atıflar

Biçimlendirilmiş bir atıfı kopyalayıp yapıştırın veya seçtiğiniz biçimde dışa aktarmak için seçeneklerden birini kullanın


DIŞA AKTAR



APA

Boz Er, A.B., & Er, İ. (2024). Notch and Hedgehog Signalling Axis Drive Senescence in HER2-Positive Breast Cancer Resistant to Trastuzumab. European Journal of Biology, 83(2), 213-221. https://doi.org/10.26650/EurJBiol.2024.1531120


AMA

Boz Er A B, Er İ. Notch and Hedgehog Signalling Axis Drive Senescence in HER2-Positive Breast Cancer Resistant to Trastuzumab. European Journal of Biology. 2024;83(2):213-221. https://doi.org/10.26650/EurJBiol.2024.1531120


ABNT

Boz Er, A.B.; Er, İ. Notch and Hedgehog Signalling Axis Drive Senescence in HER2-Positive Breast Cancer Resistant to Trastuzumab. European Journal of Biology, [Publisher Location], v. 83, n. 2, p. 213-221, 2024.


Chicago: Author-Date Style

Boz Er, Asiye Büşra, and İdris Er. 2024. “Notch and Hedgehog Signalling Axis Drive Senescence in HER2-Positive Breast Cancer Resistant to Trastuzumab.” European Journal of Biology 83, no. 2: 213-221. https://doi.org/10.26650/EurJBiol.2024.1531120


Chicago: Humanities Style

Boz Er, Asiye Büşra, and İdris Er. Notch and Hedgehog Signalling Axis Drive Senescence in HER2-Positive Breast Cancer Resistant to Trastuzumab.” European Journal of Biology 83, no. 2 (Dec. 2024): 213-221. https://doi.org/10.26650/EurJBiol.2024.1531120


Harvard: Australian Style

Boz Er, AB & Er, İ 2024, 'Notch and Hedgehog Signalling Axis Drive Senescence in HER2-Positive Breast Cancer Resistant to Trastuzumab', European Journal of Biology, vol. 83, no. 2, pp. 213-221, viewed 22 Dec. 2024, https://doi.org/10.26650/EurJBiol.2024.1531120


Harvard: Author-Date Style

Boz Er, A.B. and Er, İ. (2024) ‘Notch and Hedgehog Signalling Axis Drive Senescence in HER2-Positive Breast Cancer Resistant to Trastuzumab’, European Journal of Biology, 83(2), pp. 213-221. https://doi.org/10.26650/EurJBiol.2024.1531120 (22 Dec. 2024).


MLA

Boz Er, Asiye Büşra, and İdris Er. Notch and Hedgehog Signalling Axis Drive Senescence in HER2-Positive Breast Cancer Resistant to Trastuzumab.” European Journal of Biology, vol. 83, no. 2, 2024, pp. 213-221. [Database Container], https://doi.org/10.26650/EurJBiol.2024.1531120


Vancouver

Boz Er AB, Er İ. Notch and Hedgehog Signalling Axis Drive Senescence in HER2-Positive Breast Cancer Resistant to Trastuzumab. European Journal of Biology [Internet]. 22 Dec. 2024 [cited 22 Dec. 2024];83(2):213-221. Available from: https://doi.org/10.26650/EurJBiol.2024.1531120 doi: 10.26650/EurJBiol.2024.1531120


ISNAD

Boz Er, AsiyeBüşra - Er, İdris. Notch and Hedgehog Signalling Axis Drive Senescence in HER2-Positive Breast Cancer Resistant to Trastuzumab”. European Journal of Biology 83/2 (Dec. 2024): 213-221. https://doi.org/10.26650/EurJBiol.2024.1531120



ZAMAN ÇİZELGESİ


Gönderim09.08.2024
Kabul11.11.2024
Çevrimiçi Yayınlanma19.12.2024

LİSANS


Attribution-NonCommercial (CC BY-NC)

This license lets others remix, tweak, and build upon your work non-commercially, and although their new works must also acknowledge you and be non-commercial, they don’t have to license their derivative works on the same terms.


PAYLAŞ




İstanbul Üniversitesi Yayınları, uluslararası yayıncılık standartları ve etiğine uygun olarak, yüksek kalitede bilimsel dergi ve kitapların yayınlanmasıyla giderek artan bilimsel bilginin yayılmasına katkıda bulunmayı amaçlamaktadır. İstanbul Üniversitesi Yayınları açık erişimli, ticari olmayan, bilimsel yayıncılığı takip etmektedir.