• 제목/요약/키워드: TNBC

검색결과 48건 처리시간 0.029초

ABT-737 ameliorates docetaxel resistance in triple negative breast cancer cell line

  • Hwang, Eunjoo;Hwang, Seong-Hye;Kim, Jongjin;Park, Jin Hyun;Oh, Sohee;Kim, Young A;Hwang, Ki-Tae
    • Annals of Surgical Treatment and Research
    • /
    • 제95권5호
    • /
    • pp.240-248
    • /
    • 2018
  • Purpose: This study aimed to validate the synergistic effect of ABT-737 on docetaxel using MDA-MB-231, a triple negative breast cancer (TNBC) cell line overexpressing B-cell lymphoma-2 (Bcl-2). Methods: Western blot analysis was performed to assess expression levels of Bcl-2 family proteins and caspase-related molecules. Cell viability was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Cell cycle distribution was determined by flow cytometry analysis. Benzyloxycarbonyl-Val-Ala-Asp(OMe)-fluoromethylketone (z-VAD-fmk) was used for pretreatment to assess the role of caspases. Results: Cell viability of MDA-MB-231 after combination treatment with ABT-737 and docetaxel was significantly lower than that after docetaxel or ABT-737 monotherapy based on MTT assay (both P < 0.001), with a combination index of 0.41. The proportion of sub-G1 population after combination treatment was significantly higher than that after docetaxel or ABT-737 monotherapy (P = 0.001, P = 0.003, respectively). Pretreatment with z-VAD-fmk completely restored cell viability of MDA-MB-231 from apoptotic cell death induced by combination therapy (P = 0.001). Although pro-caspase-8 or Bid did not show significant change in expression level, pro-casepase-9 showed significantly decreased expression after combination treatment. Cleaved caspase-3 showed increased expression while poly (ADP-ribose) polymerase cleavage was induced after combination treatment. However, hypoxia-inducible factor 1-alpha and aldehyde dehydrogenase 1 totally lost their expression after combination treatment. Conclusion: Combination of ABT-737 with docetaxel elicits synergistic therapeutic effect on MDA-MB-231, a TNBC cell line overexpressing Bcl-2, mainly by activating the intrinsic pathway of apoptosis. Therefore, adjunct of ABT-737 to docetaxel might be a new therapeutic option to overcome docetaxel resistance of TNBCs overexpressing Bcl-2.

The expression of Rab5 and its effect on invasion, migration and exosome secretion in triple negative breast cancer

  • Lei Qiao;Chao Dong;Jiaojiao Zhang;Gang Sun
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제27권2호
    • /
    • pp.157-165
    • /
    • 2023
  • Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer and current therapeutic strategies are limited in their effectiveness. The expressions of Rab5 and the M2 tumor-associated macrophage marker CD163 in tissues were detected by Western blot. The migration and invasion of cells were determined using a Transwell assay. The expressions of the exosome markers were evaluated by Western blot. The polarization of human macrophages (THP-1) was determined by incubation of THP-1 cells with conditioned medium or exosomes collected from MDA-MB-231 cells with indicated transfections or by a coculture system of THP-1 and MDA-MB-231 cells. The M1 and M2 macrophage markers were evaluated by qRT-PCR. The expression of Rab5 in TNBC was significantly higher than that in normal breast tissue. Rab5 expressions in triple-negative and luminal A breast cancer were higher than those in other molecular subtypes. Higher CD163 expression was observed in triple-negative breast cancer and in triple-negative and luminal B subtypes. Rab5 knockdown suppressed but Rab5 overexpression promoted the migration and invasion capacity of MDA-MB-231 cells. The levels of CD63 and CD9 in the medium of Rab5 knockdown cells were lower than those in control cells, whereas higher levels of CD63 and CD9 were observed in Rab5 overexpression cells. Rab5 knockdown decreased the excretion but did not alter the diameter of the exosomes. Knockdown of Rab5 facilitated the anti-tumor polarization of macrophages, which was partially reversed by Rab5 overexpression. Therefore, Rab5 is expected to be a potential therapeutic target for triple-negative breast cancer.

Mouse models of breast cancer in preclinical research

  • Park, Mi Kyung;Lee, Chang Hoon;Lee, Ho
    • Laboraroty Animal Research
    • /
    • 제34권4호
    • /
    • pp.160-165
    • /
    • 2018
  • Breast cancer remains the second leading cause of cancer death among woman, worldwide, despite advances in identifying novel targeted therapies and the development of treating strategies. Classification of clinical subtypes (ER+, PR+, HER2+, and TNBC (Triple-negative)) increases the complexity of breast cancers, which thus necessitates further investigation. Mouse models used in breast cancer research provide an essential approach to examine the mechanisms and genetic pathway in cancer progression and metastasis and to develop and evaluate clinical therapeutics. In this review, we summarize tumor transplantation models and genetically engineered mouse models (GEMMs) of breast cancer and their applications in the field of human breast cancer research and anti-cancer drug development. These models may help to improve the knowledge of underlying mechanisms and genetic pathways, as well as creating approaches for modeling clinical tumor subtypes, and developing innovative cancer therapy.

Anti-migration and anti-invasion effects of LY-290181 on breast cancer cell lines through the inhibition of Twist1

  • Jiyoung Park;Sewoong Lee;Haelim Yoon;Eunjeong Kang;Sayeon Cho
    • BMB Reports
    • /
    • 제56권7호
    • /
    • pp.410-415
    • /
    • 2023
  • Breast cancer has become the most common cancer among women worldwide. Among breast cancers, metastatic breast cancer is associated with the highest mortality rate. Twist1, one of the epithelial-mesenchymal transition-regulating transcription factors, is known to promote the intravasation of breast cancer cells into metastatic sites. Therefore, targeting Twist1 to develop anti-cancer drugs might be a valuable strategy. In this study, LY-290181 dose-dependently inhibited migration, invasion, and multicellular tumor spheroid invasion in breast cancer cell lines. These anti-cancer effects of LY-290181 were mediated through the down-regulation of Twist1 protein levels. LY-290181 inhibited extracellular signal-regulated kinase and c-Jun N-terminal kinase signaling pathways. Therefore, our findings suggest that LY-290181 may serve as a basis for future research and development of an anti-cancer agent targeting metastatic cancers.

Cisplatin Plus Gemcitabine for Treatment of Breast Cancer Patients with Brain Metastases: a Preferential Option for Triple Negative Patients?

  • Erten, Cigdem;Demir, Lutfiye;Somali, Isil;Alacacioglu, Ahmet;Kucukzeybek, Yuksel;Akyol, Murat;Can, Alper;Dirican, Ahmet;Bayoglu, Vedat;Tarhan, Mustafa Oktay
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제14권6호
    • /
    • pp.3711-3717
    • /
    • 2013
  • Background: To assess the efficacy and tolerability of Cisplatin plus Gemcitabine combination in patients with brain metastases (BM) from breast cancer (BC). Materials and Methods: Eighteen BC patients with BM who were treated with Cisplatin plus Gemcitabine regimen between 2003-2011 were evaluated. Results: A median of 6 cycles of this regimen were received, in fifteen patients (83.3%) as first-line chemotherapy, in 2 as second-line and in 1 as third-line after diagnosis of BM. Dose reduction was performed in 11 (61.1%) patients; major reasons were neutropenia and leukopenia. Grade III neutropenia and Grade II trombocytopenia rates were 33.3% and 16.7% respectively. Overall response rate (ORR; complete+partial response rate) was 33.4% (n=6) for the entire study population; triple negative patients achieved an 66.6% ORR while hormone receptor (HR) positive patients had 25% and HER2 positive patients 12.5%. Median progression-free survival was 5.6 months (2.4-8.8 months, 95%CI) and longer in patients with triple negative breast cancer (TNBC) (median 7.4 months, 95%CI, 2.4-12.3 months) than the patients with other subtypes (median 5 months for HER2 positive and 3.6 months for HR positive patients). Median PFS of the patients with TNBC who received this regimen as first-line was 9.2 months (5.2-13.2 months, 95%CI). Conclusions: Cisplatin plus Gemcitabine may be a treatment option for patients with BM from breast cancer. Longer PFS and higher response rates are results that support the usage of this regimen especially for the triple negative subtype. However, further prospective and randomized trials are clearly required to provide more exact information.

Breast Tumor Cell Nuclei Segmentation in Histopathology Images using EfficientUnet++ and Multi-organ Transfer Learning

  • Dinh, Tuan Le;Kwon, Seong-Geun;Lee, Suk-Hwan;Kwon, Ki-Ryong
    • 한국멀티미디어학회논문지
    • /
    • 제24권8호
    • /
    • pp.1000-1011
    • /
    • 2021
  • In recent years, using Deep Learning methods to apply for medical and biomedical image analysis has seen many advancements. In clinical, using Deep Learning-based approaches for cancer image analysis is one of the key applications for cancer detection and treatment. However, the scarcity and shortage of labeling images make the task of cancer detection and analysis difficult to reach high accuracy. In 2015, the Unet model was introduced and gained much attention from researchers in the field. The success of Unet model is the ability to produce high accuracy with very few input images. Since the development of Unet, there are many variants and modifications of Unet related architecture. This paper proposes a new approach of using Unet++ with pretrained EfficientNet as backbone architecture for breast tumor cell nuclei segmentation and uses the multi-organ transfer learning approach to segment nuclei of breast tumor cells. We attempt to experiment and evaluate the performance of the network on the MonuSeg training dataset and Triple Negative Breast Cancer (TNBC) testing dataset, both are Hematoxylin and Eosin (H & E)-stained images. The results have shown that EfficientUnet++ architecture and the multi-organ transfer learning approach had outperformed other techniques and produced notable accuracy for breast tumor cell nuclei segmentation.

Radiation dosimetry of 89Zr labeled antibody estimated using the MIRD method and MCNP code

  • Saeideh Izadi Yazdi ;Mahdi Sadeghi ;Elham Saeedzadeh ;Mostafa Jalilifar
    • Nuclear Engineering and Technology
    • /
    • 제55권4호
    • /
    • pp.1265-1268
    • /
    • 2023
  • One important issue in using radiopharmaceuticals as therapeutic and imaging agents is predicting different organ absorbed dose following their injection. The present study aims at extrapolating dosimetry estimates to a female phantom from the animal data of 89Zr radionuclide accumulation using the Sparks-Idogan relationship. The absorbed dose of 89Zr radionuclide in different organs of the human body was calculated based on its distribution data in mice using both MIRD method and the MCNP simulation code. In this study, breasts, liver, heart wall, stomach, kidneys, lungs and spleen were considered as source and target organs. The highest and the lowest absorbed doses were respectively delivered to the liver (4.00E-02 and 3.43E-02 mGy/MBq) and the stomach (1.83E-03 and 1.66E-03 mGy/MBq). Moreover, there was a good agreement between the results obtained from both MIRD and MCNP methods. Therefore, according to the dosimetry results, [89Zr] DFO-CR011-PET/CT seems to be a suitable for diagnostic imaging of the breast anomalies for CDX-011 targeting gpNMB in patients with TNBC in the future.

KPNA3 promotes epithelial-mesenchymal transition by regulating TGF-β and AKT signaling pathways in MDA-MB-231, a triple-negative breast cancer cell line

  • Jaesung Choi;Jee-Hye Choi;Ho Woon Lee;Dongbeom Seo;Gavaachimed Lkhagvasuren;Jung-Woong Kim;Sang-Beom Seo;Kangseok Lee;Kwang-Ho Lee
    • BMB Reports
    • /
    • 제56권2호
    • /
    • pp.120-125
    • /
    • 2023
  • Karyopherin-α3 (KPNA3), a karyopherin-α isoform, is intimately associated with metastatic progression via epithelial-mesenchymal transition (EMT). However, the molecular mechanism underlying how KPNA3 acts as an EMT inducer remains to be elucidated. In this report, we identified that KPNA3 was significantly upregulated in cancer cells, particularly in triple-negative breast cancer, and its knockdown resulted in the suppression of cell proliferation and metastasis. The comprehensive transcriptome analysis from KPNA3 knockdown cells indicated that KPNA3 is involved in the regulation of numerous EMT-related genes, including the downregulation of GATA3 and E-cadherin and the up-regulation of HAS2. Moreover, it was found that KPNA3 EMT-mediated metastasis can be achieved by TGF-β or AKT signaling pathways; this suggests that the novel independent signaling pathways KPNA3-TGF-β-GATA3-HAS2/E-cadherin and KPNA3-AKT-HAS2/E-cadherin are involved in the EMT-mediated progress of TNBC MDA-MB-231 cells. These findings provide new insights into the divergent EMT inducibility of KPNA3 according to cell and cancer type.