Next Article in Journal
Proton Treatment Suppresses Exosome Production in Head and Neck Squamous Cell Carcinoma
Previous Article in Journal
Reply to Jue et al. Overcoming Understaging and Undergrading in Upper Tract Urothelial Carcinoma. Comment on “Ghoreifi et al. Modern Kidney-Sparing Management of Upper Tract Urothelial Carcinoma. Cancers 2023, 15, 4495”
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Correction

Correction: Jannoo et al. Targeting and Sensitization of Breast Cancer Cells to Killing with a Novel Interleukin-13 Receptor α2-Specific Hybrid Cytolytic Peptide. Cancers 2023, 15, 2772

by
Riaz Jannoo
1,
William Walker
2 and
Venkateswarlu Kanamarlapudi
2,*
1
UCL ECMC GCLP Facility, UCL Cancer Institute, University College London, London WC1E 6DD, UK
2
Institute of Life Science, School of Medicine, Swansea University, Singleton Park, Swansea SA2 8PP, UK
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(5), 1006; https://doi.org/10.3390/cancers16051006
Submission received: 11 January 2024 / Accepted: 30 January 2024 / Published: 29 February 2024
In the original publication [1], there was a mistake in Figures 4 and 9 and reference [43] when published. The headers for the top two tables were inaccurate in Figure 4b. Incorrect images were used for MCF-10A (untreated), MCF-7 (Phor21) and MDA-MB-231 (Phor21) in Figure 9. The corrected Figure 4 and Figure 9 and Reference [43] appears below.
Reference [43]: Gkretsi, V.; Stylianou, A.; Louca, M.; Stylianopoulos T. Identification of Ras suppressor-1 (RSU-1) as a potential breast cancer metastasis biomarker using a three-dimensional in vitro approach. Oncotarget 2017, 8, 27364–27379. https://doi.org/10.18632/oncotarget.16062.
The authors apologize for any inconvenience caused and state that the scientific conclusions are unaffected. This correction was approved by the Academic Editor. The original publication has also been updated.

Reference

  1. Jannoo, R.; Walker, W.; Kanamarlapudi, V. Targeting and Sensitization of Breast Cancer Cells to Killing with a Novel Interleukin-13 Receptor α2-Specific Hybrid Cytolytic Peptide. Cancers 2023, 15, 2772. [Google Scholar] [CrossRef] [PubMed]
Figure 4. Treatment of representative breast cancer cell lines with Pep-1-Phor21. (a) Dose-dependent effect of Pep-1-Phor21 (O), Pep-1 (■), or Phor21 (∆) on the viability of non-tumorigenic (MCF-10A), non-TNBC (MCF-7), and TNBC cells (MDA-MB 231, LM2). Cells were treated with different concentrations of Pep-1-Phor21 (effective concentration range = 0–120 µM, a 5-fold serial dilution) and their viability was assessed after 3 h, 6 h, or 24 h of the treatment by Alamar Blue assay. (b) The IC50 of peptides for various cell lines for different incubation times. (c) The cytotoxic effect of individual peptides on cell lines was also assessed by CellTox assay. MCF-10A and MCF-7 cells were treated for 3 h with 120 µM Pep-1-Phor21 (maximum concentration used in the dose-response analysis, Alamar Blue, Figure 4a). MDA-MB 231 and LM2 cells were treated with Pep-1-Phor21 at 24 µM (maximal effective concentration against LM2, as determined in dose-response analysis, Alamar Blue, Figure 4a). Pep-1-Phor21 had a significant cytotoxic effect only against IL-13Rα2-expressing TNBC cells (MDA-MB-231, LM2; relative cytotoxicity = 85.2% ± 5.4 and 96.9% ± 0.38, respectively, versus non-treated cells). Data = mean value ± SEM of three independent experiments (* p ≤ 0.05; ** p ≤ 0.01; *** p ≤ 0.001; **** p ≤ 0.0001).
Figure 4. Treatment of representative breast cancer cell lines with Pep-1-Phor21. (a) Dose-dependent effect of Pep-1-Phor21 (O), Pep-1 (■), or Phor21 (∆) on the viability of non-tumorigenic (MCF-10A), non-TNBC (MCF-7), and TNBC cells (MDA-MB 231, LM2). Cells were treated with different concentrations of Pep-1-Phor21 (effective concentration range = 0–120 µM, a 5-fold serial dilution) and their viability was assessed after 3 h, 6 h, or 24 h of the treatment by Alamar Blue assay. (b) The IC50 of peptides for various cell lines for different incubation times. (c) The cytotoxic effect of individual peptides on cell lines was also assessed by CellTox assay. MCF-10A and MCF-7 cells were treated for 3 h with 120 µM Pep-1-Phor21 (maximum concentration used in the dose-response analysis, Alamar Blue, Figure 4a). MDA-MB 231 and LM2 cells were treated with Pep-1-Phor21 at 24 µM (maximal effective concentration against LM2, as determined in dose-response analysis, Alamar Blue, Figure 4a). Pep-1-Phor21 had a significant cytotoxic effect only against IL-13Rα2-expressing TNBC cells (MDA-MB-231, LM2; relative cytotoxicity = 85.2% ± 5.4 and 96.9% ± 0.38, respectively, versus non-treated cells). Data = mean value ± SEM of three independent experiments (* p ≤ 0.05; ** p ≤ 0.01; *** p ≤ 0.001; **** p ≤ 0.0001).
Cancers 16 01006 g004
Figure 9. Confocal cell imaging of breast cancer spheroids treated with Pep-1-Phor21. Indicated cell lines established as spheroids (48 h) were treated with Pep-1-Phor21 or Phor21 (3 h, 30 µM) and assessed for the presence of live and dead cells using confocal fluorescent microscopy. IL-13Rα2-positive TNBC spheroids (MDA-MB-231, LM2) exhibited diffuse dead cell staining (red fluorescence, EthD-1) with the concomitant disruption of spheroid integrity after Pep-1-Phor21 treatment. Some foci of live cells (green fluorescence, Vybrant DiO) were detectable within the core of the disrupted spheroid after 3 h. In contrast, IL-13Rα2-negative MCF-10A breast epithelial and non-TNBC MCF-7 cells exhibited only viable cell staining with no observable loss of spheroid structure post-treatment.
Figure 9. Confocal cell imaging of breast cancer spheroids treated with Pep-1-Phor21. Indicated cell lines established as spheroids (48 h) were treated with Pep-1-Phor21 or Phor21 (3 h, 30 µM) and assessed for the presence of live and dead cells using confocal fluorescent microscopy. IL-13Rα2-positive TNBC spheroids (MDA-MB-231, LM2) exhibited diffuse dead cell staining (red fluorescence, EthD-1) with the concomitant disruption of spheroid integrity after Pep-1-Phor21 treatment. Some foci of live cells (green fluorescence, Vybrant DiO) were detectable within the core of the disrupted spheroid after 3 h. In contrast, IL-13Rα2-negative MCF-10A breast epithelial and non-TNBC MCF-7 cells exhibited only viable cell staining with no observable loss of spheroid structure post-treatment.
Cancers 16 01006 g009
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jannoo, R.; Walker, W.; Kanamarlapudi, V. Correction: Jannoo et al. Targeting and Sensitization of Breast Cancer Cells to Killing with a Novel Interleukin-13 Receptor α2-Specific Hybrid Cytolytic Peptide. Cancers 2023, 15, 2772. Cancers 2024, 16, 1006. https://doi.org/10.3390/cancers16051006

AMA Style

Jannoo R, Walker W, Kanamarlapudi V. Correction: Jannoo et al. Targeting and Sensitization of Breast Cancer Cells to Killing with a Novel Interleukin-13 Receptor α2-Specific Hybrid Cytolytic Peptide. Cancers 2023, 15, 2772. Cancers. 2024; 16(5):1006. https://doi.org/10.3390/cancers16051006

Chicago/Turabian Style

Jannoo, Riaz, William Walker, and Venkateswarlu Kanamarlapudi. 2024. "Correction: Jannoo et al. Targeting and Sensitization of Breast Cancer Cells to Killing with a Novel Interleukin-13 Receptor α2-Specific Hybrid Cytolytic Peptide. Cancers 2023, 15, 2772" Cancers 16, no. 5: 1006. https://doi.org/10.3390/cancers16051006

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop