Open Access

High p62 and ALDH1A3 Reduce the Effectiveness of Endocrine Therapy in Luminal B Breast Cancer

YUKI MAEMURA 1*
AYAKA OZAKI 1*
RYOSUKE CHIWAKI 1
MISAKI ENOMOTO 1
YUNA TADA 1
YUKA NAGASHIMA 1
HAYATO ISHII 1
KANA NOHATA 1
TAKAHIRO KASAI 1
RANMAN OKIYAMA 1
YOHSUKE HARADA 1
SHOMA TAMORI 1,2
SHIGEO OHNO 3
KAZUNORI SASAKI 3
  &  
KAZUNORI AKIMOTO 1,2
*These Authors contributed equally to this work

1Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Tokyo, Japan

2Research Division of Medical Data Science, Research Institute for Science and Technology, Tokyo University of Science, Chiba, Japan

3Laboratory of Cancer Biology, Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo, Japan

Cancer Diagnosis & Prognosis Nov-Dec; 5(6): 677-697 DOI: 10.21873/cdp.10484
Received 23 August 2025 | Revised 10 September 2025 | Accepted 11 September 2025
Corresponding author
Kazunori Akimoto, Ph.D., Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan. Tel: +81 358761567, e-mail: akimoto@rs.tus.ac.jp
Download PDF pdf image icon

Abstract

Background/Aim
High expression of p62 and ALDH1A3 indicates a poor clinical outcome in luminal B breast cancer, and p62 is involved in the progression of ALDH1-positive luminal B breast cancer stem cells. However, the association between endocrine therapy and high p62 and ALDH1A3 expression, in luminal B breast cancer remains unclear.
Materials and Methods
Two datasets with gene expression and clinical data for patients with primary breast cancer (METABRIC, n=2,509; The Cancer Genome Atlas, n=1,084) were downloaded and statistically analyzed. To evaluate the association between the p62 and ALDH1A3 expression levels and endocrine therapy, including tamoxifen and aromatase inhibitor, in patients with luminal B breast cancer, disease-specific survival was examined using Kaplan-Meier and multivariate Cox regression analyses.
Results
Patients with p62high ALDH1A3high luminal B breast cancer treated with endocrine therapy exhibited a poor prognosis. Moreover, patients with p62high ALDH1A3high luminal B breast cancer treated with tamoxifen showed a trend towards a poor prognosis, but those treated with aromatase inhibitors showed a significantly poor prognosis. These results suggest that endocrine therapy, especially aromatase inhibitors, exhibits a reduced effectiveness against p62high ALDH1A3high luminal B tumors.
Conclusion
p62 and ALDH1A3 could be used together as a prognostic biomarker for predicting the efficacy of endocrine therapy for luminal B breast cancer.
Keywords: p62, ALDH1A3, endocrine therapy, breast cancer, luminal B

Introduction

Breast cancer has the highest incidence rate and is the leading cause of cancer-related death among women worldwide (1). Among the breast cancer subtypes, luminal A and luminal B express estrogen receptor (ER) and/or progesterone receptor, and certain luminal B tumors express HER2 and highly express the proliferation marker Ki-67 (MKI67) (2,3). Additionally, luminal B breast cancer has a poorer prognosis among the breast cancer subtypes (2-10). The main treatments for luminal B breast cancer entail surgery, radiotherapy and drug therapy, which include endocrine therapy, HER2-targeted therapy, and chemotherapy (2,3,11). However, since the luminal B subtype still exhibits a poorer prognosis, further stratification and the development of prognostic markers and therapeutic targets are required for improving the quality of life of patients. It is also important to identify effective prognostic biomarkers to assess the effectiveness of treatments for luminal B breast cancer.

Cancer stem cells (CSCs) have stem-like properties such as self-renewal, multi-differentiation and tumorigenicity (12,13). Since CSCs are resistant to conventional drug therapy and radiotherapy, the development of targeted therapies against CSCs is necessary to improve the clinical outcomes of patients (12-15). Aldehyde dehydrogenase 1 (ALDH1) is a detoxifying enzyme that converts aldehydes into carboxylic acids and is known as a CSC marker (16). Among the ALDH1 genes, ALDH1A3 is known to be a CSC marker in several cancer types (17,18). ALDH1A3 expression is correlated with tumor grade, metastasis and the clinical outcome of breast cancer (19-21), and it is a significant contributory factor to ALDH1 activity in this disease (20,22). ALDH1A3 expression is also associated with chemoresistance and radioresistance in several cancer types (17,22,23). However, the association between ALDH1A3 expression and the effectiveness of endocrine therapy in luminal B breast cancer remains to be elucidated.

p62 is a multifunctional scaffold protein that is involved in various biological and cellular activities, including the NF-κB signaling pathway, antioxidant response and autophagy (24-34). p62 is highly expressed in a number of cancer types (35-45), and high p62 expression is corelated with a poor clinical outcome in several cancer types (38,41,43,46-49). The p62 gene and protein are also highly expressed in breast cancer (50-52), and high p62 expression is corelated with a poor clinical outcome (51-55). Furthermore, high expression of p62 and ALDH1A3 indicates a poor clinical outcome in luminal B breast cancer, and p62 is involved in the cancerous progression of ALDH1-positive luminal B breast CSCs (55). Moreover, co-expression of p62 and ALDH1A3 in luminal B breast cancer reduces the effectiveness of radiotherapy (56). However, the association between endocrine therapy and high p62 and ALDH1A3 expression, in luminal B breast cancer remains unclear.

The results of the present study revealed that p62high ALDH1A3high luminal B tumors are associated with a reduced effectiveness of endocrine therapy, especially aromatase inhibitor therapy.

Materials and Methods

Datasets. The Molecular Taxonomy of Breast Cancer International Consortium (METABRIC; n=2,509) (57,58) and The Cancer Genome Atlas (TCGA; n=1,084) (59) datasets were downloaded from cBioportal (60,61) on August 1, 2024. The details of the data are shown in Figure 1 and have also been reported previously (62).

Statistical analysis. The details of the statistical analyses have been reported previously (62). The optimal cutoff determined by the Youden’s index following receiver operating characteristic analysis was used to divide each group into the p62 or/and ALDH1A3 high and low gene expression groups for disease-specific survival (DSS) analysis (Table I). Kaplan-Meier curves for DSS were plotted and then compared using the log-rank (Cochran-Mantel-Haenszel) test. Multiplicity was adjusted using the Holm test as a post hoc test. A multivariate Cox regression analysis for DSS was performed to evaluate the influence of gene expression and to estimate the adjusted hazard ratios (HRs) with the age at diagnosis as a confounding factor in Table II, Table III, Table IV and Table V. In Table II, chemotherapy and radiotherapy were also included as additional confounding factors. Two-sided p<0.05 was considered to indicate a statistically significant difference. All statistical analyses were conducted using R software (version 4.3.0) and BellCurve for Excel (version 4.05; Social Survey Research Information Co., Ltd., Tokyo, Japan).

Results

Endocrine therapy is insufficient for treating p62high luminal A breast cancer but sufficient for p62high luminal B breast cancer in the METABRIC dataset. High p62 expression is associated with poorer clinical outcomes in luminal A and luminal B breast cancer (55). However, the association between endocrine therapy and high p62 expression in the luminal A and luminal B breast cancer subtypes remains unclear. Therefore, to examine the clinical outcome of patients with p62high luminal A and luminal B breast cancer treated with endocrine therapy, Kaplan-Meier and multivariate analyses for DSS were performed. The overall workflow of the present study is shown in Figure 1. We first analyzed the gene expression data from patients with breast cancer in the METABRIC dataset. We performed Kaplan-Meier analysis to compare the DSS of patients with p62high or p62low luminal A and luminal B breast cancer who did or did not receive endocrine therapy. Patients with p62high luminal A breast cancer who did not receive endocrine therapy exhibited a good prognosis and those who received endocrine therapy displayed a poorer prognosis (p=0.0035) (Figure 2A). Although patients with p62high luminal B breast cancer who did not receive endocrine therapy exhibited a poorer prognosis (p<0.001), those who did receive endocrine therapy showed an improved prognosis (Figure 2B).

Next, multivariate Cox regression analysis of DSS was performed with age, chemotherapy and radiotherapy as confounding factors. As with the Kaplan-Meier analysis, patients with p62high luminal A breast cancer who received endocrine therapy exhibited poorer clinical outcomes [without endocrine therapy: hazard ratio (HR)=1.66, 95% confidence interval (CI)=0.85-3.25, p=0.142; with endocrine therapy: HR=1.57, 95%CI=1.06-2.33, p=0.024] (Table II). Patients with p62high luminal B breast cancer who did not receive endocrine therapy exhibited poorer clinical outcomes (HR=3.84, 95%CI=1.64-8.99, p=0.002) and those who received endocrine therapy exhibited improved clinical outcomes (HR=1.41, 95%CI=0.90-2.19, p=0.132) (Table II). These results suggest that endocrine therapy may be insufficient for treating p62high luminal A breast cancer.

Endocrine therapy is insufficient for treating p62highALDH1A3high luminal B breast cancer in the METABRIC dataset. We have previously shown that high p62 and ALDH1A3 expression is associated with poorer clinical outcomes in luminal B breast cancer (55). Moreover, the effectiveness of radiotherapy is reduced in luminal B breast cancer with p62 and ALDH1A3 co-expression (56). However, the association between endocrine therapy and high p62 and ALDH1A3 expression, in luminal B breast cancer remains unclear. Therefore, we next performed Kaplan-Meier analysis for patients with p62highALDH1A3high luminal A and luminal B breast cancer who did or did not receive endocrine therapy. First, the effect of high ALDH1A3 expression in luminal A and luminal B breast cancer was examined. Kaplan-Meier analyses showed that patients with ALDH1A3high luminal A and luminal B breast cancer treated without endocrine therapy did not have poor clinical outcomes compared with ALDH1A3low patients (Figure 2C and D). Additionally, multivariate Cox regression analysis showed that patients with ALDH1A3high luminal A and luminal B breast cancer treated without endocrine therapy did not have poorer clinical outcomes compared with ALDH1A3low patients (Table II). However, ALDH1A3high luminal B breast cancer treated with endocrine therapy was predictive of poorer clinical outcomes in Kaplan-Meier analysis (p=0.032) and poorer tendency in multivariate Cox regression analysis (HR=1.75, 95%CI=0.99-3.09, p=0.055) compared with ALDH1A3low (Figure 2C and D and Table II).

Next, Kaplan-Meier analyses showed that patients with p62high ALDH1A3high luminal A and luminal B breast cancer treated without endocrine therapy did not have poorer clinical outcomes than the other patient groups (p62high ALDH1A3low, p62low ALDH1A3high and p62low ALDH1A3low) with the luminal A and luminal B subtypes (Figure 2E and F). Notably, in patients treated with endocrine therapy, there was a significant difference between the survival rates of those with p62high ALDH1A3high luminal B breast cancer and the other patients with the luminal B subtype (p=0.030) (Figure 2F). The comparison among the four groups indicated that p62high ALDH1A3high did not have the poorest survival of the four groups of patients with the luminal A breast cancer subtype, whereas p62high ALDH1A3high had the poorest survival among the four groups of patients with the luminal B breast cancer subtype (p=0.067) (Figure 2G and H). Next, a multivariate Cox regression analysis for DSS was performed with age at diagnosis as a confounding factor (Table II). Multivariate Cox regression analysis also showed that p62high ALDH1A3high was not predictive of poorer clinical outcomes of patients with luminal A breast cancer treated without endocrine therapy compared with the other patient groups (p62high ALDH1A3low, p62low ALDH1A3high and p62low ALDH1A3low) with the luminal A subtype (Table II). However, p62high ALDH1A3high was predictive of poorer clinical outcomes of patients with luminal B breast cancer treated with endocrine therapy compared with other patient groups with the luminal B subtype (HR=1.52, 95%CI=1.05-2.20, p=0.028) (Table II). These results suggest that p62 may be involved in cancer progression and contribute to the poor prognosis of patients with ALDH1A3-positive CSC-enriched luminal B breast cancer. These results also suggest that endocrine therapy may be insufficient for treating p62high ALDH1A3high luminal B breast cancer.

Endocrine therapy is insufficient for treating p62highALDH1A3high luminal B breast cancer in TCGA dataset. To confirm the above results from the METABRIC dataset analysis, we next examined TCGA dataset as another cohort. Of the p62high groups of patients with luminal A and luminal B breast cancer who did or did not receive endocrine therapy, those with the luminal B subtype treated with endocrine therapy showed a poor prognosis (p=0.0021) (Figure 3A and B). The ALDH1A3high groups of patients with luminal A and luminal B breast cancer who did or did not receive endocrine therapy did not show a poor prognosis (Figure 3C and D). Next, Kaplan-Meier analyses showed that patients with p62high ALDH1A3high luminal A and luminal B breast cancer treated without endocrine therapy did not have poorer clinical outcomes than the other patient groups (p62high ALDH1A3low, p62low ALDH1A3high and p62low ALDH1A3low) with the luminal A and luminal B subtypes (Figure 3E and F). For patients treated with endocrine therapy, there was a significant difference between the survival rates of those with p62high ALDH1A3high luminal B breast cancer and the other patient groups with the luminal B subtype (luminal B: p<0.001) (Figure 3F). The comparison among the four groups indicated that the p62high ALDH1A3high group did not have the poorest survival of the four groups of patients with the luminal A subtype (Figure 3G), whereas the p62high ALDH1A3high group had the poorest survival of the four groups of patients with the luminal B subtype (p=0.0013) (Figure 3H). Subsequently, multivariate Cox regression analysis for DSS was performed with age as a confounding factor, and similar results were obtained as those in the Kaplan-Meier analysis (Table III). These results also suggest that endocrine therapy may be an insufficient treatment option for patients with p62high ALDH1A3high luminal B breast cancer.

Tamoxifen is sufficient for treating p62high luminal A and luminal B breast cancer in TCGA dataset. Next, the endocrine therapy group was divided into two groups, those treated with tamoxifen and those treated with aromatase inhibitors, to examine in more detail the relationship between high p62 and ALDH1A3 expression, and endocrine therapy. Of the p62high groups of patients with luminal A and luminal B breast cancer who did or did not receive tamoxifen treatment as endocrine therapy, those with the luminal B subtype treated without tamoxifen showed a poor prognosis (p=0.0045) (Figure 4A, B and Table IV). The ALDH1A3high groups of patients with luminal A and luminal B breast cancer who did or did not receive tamoxifen treatment as endocrine therapy did not show a significant difference (Figure 4C, D and Table IV).

Tamoxifen is insufficient for treating p62high ALDH1A3high luminal A breast cancer in TCGA dataset. In patients treated with tamoxifen, there was a significant difference between the survival rates of those with p62high ALDH1A3high luminal A breast cancer and the other patient groups with the luminal A subtype, and patients with p62high ALDH1A3high luminal B breast cancer treated with tamoxifen tended to have a poor outcome (luminal A with tamoxifen: p=0.013; luminal B with tamoxifen therapy: p=0.054) (Figure 4E and F). The comparison among the four groups showed that the p62high ALDH1A3high group had the poorest survival of the four groups of patients with the luminal A and luminal B subtypes, although there was not a significant difference (Figure 4G and H).

Aromatase inhibitor therapy is insufficient for treating p62high luminal B breast cancer in TCGA dataset. Of the p62high groups of patients with luminal A and luminal B breast cancer who did or did not receive aromatase inhibitor treatment as endocrine therapy, those with the luminal B subtype showed a poor prognosis (p<0.001) (Figure 5A and B). The ALDH1A3high groups of patients with luminal A and luminal B breast cancer who did or did not receive aromatase inhibitor treatment as endocrine therapy did not show a poor prognosis (Figure 5C and D).

Aromatase inhibitor therapy is insufficient for treating p62high ALDH1A3high luminal B breast cancer in TCGA dataset. For patients treated with aromatase inhibitors, there was no significant difference between the survival rates of those with p62high ALDH1A3high luminal A breast cancer and other patients with the luminal A subtype (Figure 5E). However, patients with p62high ALDH1A3high luminal B breast cancer treated with aromatase inhibitors had a poor clinical outcome (p<0.001) (Figure 5F). The comparison among the four expression groups showed that p62high ALDH1A3high luminal A breast cancer did not show a poor prognosis (Figure 5G), but p62high ALDH1A3high indicated a poorer prognosis among the four groups of patients with the luminal B subtype (p<0.001) (Figure 5H). These results also suggest that aromatase inhibitor as endocrine therapy may be an insufficient treatment option for patients with p62high ALDH1A3high luminal B breast cancer.

Discussion

The present study demonstrated that the p62high ALDH1A3high luminal B subtype tumors treated with endocrine therapy, especially those treated with aromatase inhibitors, exhibited poor clinical outcomes. High p62 expression in cancer tissue at the gene and protein level contributes to cancer progression and metastasis (52,63-68) and is associated with a poor clinical outcome in patients with breast cancer, including the luminal B subtype (55). Therefore, p62 is expected to predict prognosis after medical treatment for several cancer types. A series of experiments at the cellular level have shown that p62 is also involved in cell proliferation, survival, antioxidant response, autophagy and tumor formation (24,26,31,55,69). In addition, p62 is involved in chemoresistance and radioresistance in cancer cells by enhancing the level of NF-κB signaling, mTOR signaling and autophagy flux (70,71). In breast cancer cell lines, p62 binds to vimentin and induces invasion and metastasis (52). In endometrial cancer cell lines, 17β-estradiol induces p62 phosphorylation and ERα expression via the PI3K/Akt/mTOR signaling pathway, and p62 phosphorylation releases ERα from the p62-KEAP1-ERα complex (72). Endocrine therapy activates the integrated stress response through NF-κB signaling in a subpopulation of ER-positive breast cancer cells (73). However, the mechanism linking high p62 expression to cancer progression and to the reduced effectiveness of endocrine therapy in luminal B breast cancer is unclear. Because aromatase inhibitors block estrogen production in postmenopausal adipose tissue, the estrogen-independent and p62-dependent signaling pathways that remain active in luminal B breast cancer cells may be responsible for the reduced therapeutic efficacy of aromatase inhibitors. This is supported by the results of the present study in which tamoxifen, which acts directly on the ER, showed a tendency towards a reduced effectiveness in p62high ALDH1A3high luminal B breast cancer (Figure 4). Thus, it will be important to clarify the detailed molecular mechanism in the future.

The luminal B breast cancer subtype expresses HER2 and is treated with HER2-targeted therapy (2,3). Therefore, we investigated the prognosis (DSS) of patients with p62high luminal B breast cancer who received HER2-targeted therapy, but we were unable to perform a prognostic analysis as no patients who received HER2 targeted therapy died in the dataset. Additionally, since the luminal B type has a high proliferation rate, chemotherapy is usually also performed. It is important to examine the effects of HER2-targeted therapy and chemotherapy in patients with high p62 expression in luminal B subtype in the future.

p62 is involved in the maintenance of cancer stem-like properties by stabilizing MYC mRNA and NRF2 activation (51,74) in several cancer types. p62 is also involved in the stem-like properties of ALDH1-positive luminal B breast CSCs (55). Furthermore, p62high ALDH1A3high luminal B breast cancer exhibits reduced response to radiotherapy, and X-ray irradiation suppresses the tumor formation of p62-deficient, ALDH1-positive luminal B breast CSCs (56). These findings raise the possibility that the reduced effectiveness of endocrine therapy in p62high ALDH1A3high luminal B breast cancer results from the stem-like properties of ALDH1A3-positive luminal B breast CSCs. Therefore, it is necessary to reveal the mechanism by which p62 is involved in these properties. p62 binds to atypical protein kinase C such as PKCζ and PKCλ/ι via a PB1-PB1 domain interaction (25). PKCζhigh and PKCζhigh ALDH1A3high luminal B breast cancers exhibit reduced response to endocrine therapy and aromatase inhibitor treatment (62,75). Thus, p62 and PKCζ may be cooperatively involved in the reduced effectiveness of endocrine therapy in ALDH1A3-positive luminal B breast cancer.

Conclusion

The present study revealed that endocrine therapy, especially aromatase inhibitor therapy, exhibits reduced effectiveness in p62high ALDH1A3high luminal B subtype tumors for. Therefore, p62high and ALDH1A3high may be used as a prognostic biomarker for predicting the efficacy of endocrine therapy for luminal B breast cancer.

Conflicts of Interest

The Authors declare that they have no competing interests in relation to this study.

Authors’ Contributions

Conceptualization: YM, AO, KA; Formal analysis: YM, AO, RC, KN; Founding Acquisition: AO, RC, ST, SO, KS, KA; Investigation: YM, AO, RC, ME, KN, KS, KA; Methodology: YM, AO, KN, KA; Project Administration: KA; Supervision: KA; Validation: RC, ME, YT, HI, KN, TK, RO, YH, ST, KS; Visualization: YM, AO, ME; Writing - Original Draft Preparation: YM, AO, YN, HI, KA; Writing - Review & Editing: YM, AO, RC, ME, YT, YN, HI, KN, TK, RO, YH, ST, SO, KS, KA.

Funding

The present study was supported by the Tokyo University of Science Grant for President’s Research Promotion, JST Moonshot R&D (grant no. JPMJMS2022-21), Grant-in-Aid for Scientific Research (C) (grant no. 20K08936, 24K12517), Grant-in-Aid for Research Activity Start-up (grant no. 21K20732), Grant-in-Aid for Early-Career Scientists (grant no. 23K14352), JST SPRING (grant no. JPMJSP2151), Grant-in-Aid for Special Research in Subsidies for ordinary expenses of private schools from The Promotion and Mutual Aid Corporation for Private Schools of Japan, Grant from Institute for Environmental & Gender-specific Medicine, Juntendo University.

Artificial Intelligence (AI) Disclosure

No artificial intelligence (AI) tools, including large language models or machine learning software, were used in the preparation, analysis, or presentation of this manuscript.

References

1 Bray F Laversanne M Sung H Ferlay J Siegel RL Soerjomataram I & Jemal A Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74(3) 229 - 263 2024. DOI: 10.3322/caac.21834
2 Prat A Pineda E Adamo B Galván P Fernández A Gaba L Díez M Viladot M Arance A & Muñoz M Clinical implications of the intrinsic molecular subtypes of breast cancer. Breast 24 Suppl. 24 S26 - S35 2015. DOI: 10.1016/j.breast.2015.07.008
3 Ades F Zardavas D Bozovic-Spasojevic I Pugliano L Fumagalli D de Azambuja E Viale G Sotiriou C & Piccart M Luminal B breast cancer: molecular characterization, clinical management, and future perspectives. J Clin Oncol. 32(25) 2794 - 2803 2014. DOI: 10.1200/JCO.2013.54.1870
4 Langlands FE Horgan K Dodwell DD & Smith L Breast cancer subtypes: response to radiotherapy and potential radiosensitisation. Br J Radiol. 86(1023) 20120601 2013. DOI: 10.1259/bjr.20120601
5 Laurberg T Tramm T Nielsen T Alsner J Nord S Myhre S Sørlie T Leung S Fan C Perou C Gelmon K Overgaard J Voduc D Prat A & Cheang MCU Intrinsic subtypes and benefit from postmastectomy radiotherapy in node-positive premenopausal breast cancer patients who received adjuvant chemotherapy - results from two independent randomized trials. Acta Oncol. 57(1) 38 - 43 2018. DOI: 10.1080/0284186X.2017.1401735
6 Chia SK Bramwell VH Tu D Shepherd LE Jiang S Vickery T Mardis E Leung S Ung K Pritchard KI Parker JS Bernard PS Perou CM Ellis MJ & Nielsen TO A 50-gene intrinsic subtype classifier for prognosis and prediction of benefit from adjuvant tamoxifen. Clin Cancer Res. 18(16) 4465 - 4472 2012. DOI: 10.1158/1078-0432.CCR-12-0286
7 Dowsett M Sestak I Lopez-Knowles E Sidhu K Dunbier AK Cowens JW Ferree S Storhoff J Schaper C & Cuzick J Comparison of PAM50 risk of recurrence score with oncotype DX and IHC4 for predicting risk of distant recurrence after endocrine therapy. J Clin Oncol. 31(22) 2783 - 2790 2013. DOI: 10.1200/JCO.2012.46.1558
8 Pu M Messer K Davies SR Vickery TL Pittman E Parker BA Ellis MJ Flatt SW Marinac CR Nelson SH Mardis ER Pierce JP & Natarajan L Research-based PAM50 signature and long-term breast cancer survival. Breast Cancer Res Treat. 179(1) 197 - 206 2020. DOI: 10.1007/s10549-019-05446-y
9 Prat A Parker JS Karginova O Fan C Livasy C Herschkowitz JI He X & Perou CM Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res. 12(5) R68 2010. DOI: 10.1186/bcr2635
10 Gnant M Filipits M Greil R Stoeger H Rudas M Bago-Horvath Z Mlineritsch B Kwasny W Knauer M Singer C Jakesz R Dubsky P Fitzal F Bartsch R Steger G Balic M Ressler S Cowens JW Storhoff J Ferree S Schaper C Liu S Fesl C Nielsen TO & Austrian Breast Colorectal Cancer Study Group Predicting distant recurrence in receptor-positive breast cancer patients with limited clinicopathological risk: using the PAM50 Risk of Recurrence score in 1478 postmenopausal patients of the ABCSG-8 trial treated with adjuvant endocrine therapy alone. Ann Oncol. 25(2) 339 - 345 2014. DOI: 10.1093/annonc/mdt494
11 Burguin A Diorio C & Durocher F Breast cancer treatments: updates and new challenges. J Pers Med. 11(8) 808 2021. DOI: 10.3390/jpm11080808
12 Reya T Morrison SJ Clarke MF & Weissman IL Stem cells, cancer, and cancer stem cells. Nature. 414(6859) 105 - 111 2001. DOI: 10.1038/35102167
13 Visvader JE & Lindeman GJ Cancer stem cells: current status and evolving complexities. Cell Stem Cell. 10(6) 717 - 728 2012. DOI: 10.1016/j.stem.2012.05.007
14 Diehn M Cho RW Lobo NA Kalisky T Dorie MJ Kulp AN Qian D Lam JS Ailles LE Wong M Joshua B Kaplan MJ Wapnir I Dirbas FM Somlo G Garberoglio C Paz B Shen J Lau SK Quake SR Brown JM Weissman IL & Clarke MF Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature. 458(7239) 780 - 783 2009. DOI: 10.1038/nature07733
15 Nguyen NP Almeida FS Chi A Nguyen LM Cohen D Karlsson U & Vinh-Hung V Molecular biology of breast cancer stem cells: Potential clinical applications. Cancer Treat Rev. 36(6) 485 - 491 2010. DOI: 10.1016/j.ctrv.2010.02.016
16 Ginestier C Hur MH Charafe-Jauffret E Monville F Dutcher J Brown M Jacquemier J Viens P Kleer CG Liu S Schott A Hayes D Birnbaum D Wicha MS & Dontu G ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell. 1(5) 555 - 567 2007. DOI: 10.1016/j.stem.2007.08.014
17 McLean ME MacLean MR Cahill HF Arun RP Walker OL Wasson MD Fernando W Venkatesh J & Marcato P The expanding role of cancer stem cell marker ALDH1A3 in cancer and beyond. Cancers (Basel). 15(2) 492 2023. DOI: 10.3390/cancers15020492
18 Marcato P Dean CA Giacomantonio CA & Lee PW Aldehyde dehydrogenase: Its role as a cancer stem cell marker comes down to the specific isoform. Cell Cycle. 10(9) 1378 - 1384 2011. DOI: 10.4161/cc.10.9.15486
19 Opdenaker LM Arnold KM Pohlig RT Padmanabhan JS Flynn DC & Sims-Mourtada J Immunohistochemical analysis of aldehyde dehydrogenase isoforms and their association with estrogen-receptor status and disease progression in breast cancer. Breast Cancer (Dove Med Press). 6 205 - 209 2014. DOI: 10.2147/BCTT.S73674
20 Marcato P Dean CA Pan D Araslanova R Gillis M Joshi M Helyer L Pan L Leidal A Gujar S Giacomantonio CA & Lee PWK Aldehyde dehydrogenase activity of breast cancer stem cells is primarily due to isoform ALDH1A3 and its expression is predictive of metastasis. Stem Cells. 29(1) 32 - 45 2011. DOI: 10.1002/stem.563
21 Marcato P Dean CA Liu RZ Coyle KM Bydoun M Wallace M Clements D Turner C Mathenge EG Gujar SA Giacomantonio CA Mackey JR Godbout R & Lee PW Aldehyde dehydrogenase 1A3 influences breast cancer progression via differential retinoic acid signaling. Mol Oncol. 9(1) 17 - 31 2015. DOI: 10.1016/j.molonc.2014.07.010
22 Croker AK Rodriguez-Torres M Xia Y Pardhan S Leong HS Lewis JD & Allan AL Differential functional roles of ALDH1A1 and ALDH1A3 in mediating metastatic behavior and therapy resistance of human breast cancer cells. Int J Mol Sci. 18(10) 2039 2017. DOI: 10.3390/ijms18102039
23 Mao P Joshi K Li J Kim SH Li P Santana-Santos L Luthra S Chandran UR Benos PV Smith L Wang M Hu B Cheng SY Sobol RW & Nakano I Mesenchymal glioma stem cells are maintained by activated glycolytic metabolism involving aldehyde dehydrogenase 1A3. Proc Natl Acad Sci U S A. 110(21) 8644 - 8649 2013. DOI: 10.1073/pnas.1221478110
24 Moscat J & Diaz-Meco MT p62 at the crossroads of autophagy, apoptosis, and cancer. Cell. 137(6) 1001 - 1004 2009. DOI: 10.1016/j.cell.2009.05.023
25 Moscat J Diaz-Meco MT & Wooten MW Of the atypical PKCs, Par-4 and p62: recent understandings of the biology and pathology of a PB1-dominated complex. Cell Death Differ. 16(11) 1426 - 1437 2009. DOI: 10.1038/cdd.2009.119
26 Fan L Yin S Zhang E & Hu H Role of p62 in the regulation of cell death induction. Apoptosis. 23(3-4) 187 - 193 2018. DOI: 10.1007/s10495-018-1445-z
27 Puls A Schmidt S Grawe F & Stabel S Interaction of protein kinase C zeta with ZIP, a novel protein kinase C-binding protein. Proc Natl Acad Sci U S A. 94(12) 6191 - 6196 1997. DOI: 10.1073/pnas.94.12.6191
28 Sanchez P De Carcer G Sandoval IV Moscat J & Diaz-Meco MT Localization of atypical protein kinase C isoforms into lysosome-targeted endosomes through interaction with p62. Mol Cell Biol. 18(5) 3069 - 3080 1998. DOI: 10.1128/MCB.18.5.3069
29 Sanz L Sanchez P Lallena MJ Diaz-Meco MT & Moscat J The interaction of p62 with RIP links the atypical PKCs to NF-kappaB activation. EMBO J. 18(11) 3044 - 3053 1999. DOI: 10.1093/emboj/18.11.3044
30 Sanz L Diaz-Meco MT Nakano H & Moscat J The atypical PKC-interacting protein p62 channels NF-kappaB activation by the IL-1-TRAF6 pathway. EMBO J. 19(7) 1576 - 1586 2000. DOI: 10.1093/emboj/19.7.1576
31 Jain A Lamark T Sjøttem E Larsen KB Awuh JA Øvervatn A McMahon M Hayes JD & Johansen T p62/SQSTM1 is a target gene for transcription factor NRF2 and creates a positive feedback loop by inducing antioxidant response element-driven gene transcription. J Biol Chem. 285(29) 22576 - 22591 2010. DOI: 10.1074/jbc.M110.118976
32 Bjørkøy G Lamark T Brech A Outzen H Perander M Overvatn A Stenmark H & Johansen T p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J Cell Biol. 171(4) 603 - 614 2005. DOI: 10.1083/jcb.200507002
33 Pankiv S Clausen TH Lamark T Brech A Bruun JA Outzen H Øvervatn A Bjørkøy G & Johansen T p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem. 282(33) 24131 - 24145 2007. DOI: 10.1074/jbc.M702824200
34 Ichimura Y Kumanomidou T Sou YS Mizushima T Ezaki J Ueno T Kominami E Yamane T Tanaka K & Komatsu M Structural basis for sorting mechanism of p62 in selective autophagy. J Biol Chem. 283(33) 22847 - 22857 2008. DOI: 10.1074/jbc.M802182200
35 Kitamura H Torigoe T Asanuma H Hisasue SI Suzuki K Tsukamoto T Satoh M & Sato N Cytosolic overexpression of p62 sequestosome 1 in neoplastic prostate tissue. Histopathology. 48(2) 157 - 161 2006. DOI: 10.1111/j.1365-2559.2005.02313.x
36 Ellis RA Horswell S Ness T Lumsdon J Tooze SA Kirkham N Armstrong JL & Lovat PE Prognostic impact of p62 expression in cutaneous malignant melanoma. J Invest Dermatol. 134(5) 1476 - 1478 2014. DOI: 10.1038/jid.2013.497
37 Kuo WL Sharifi MN Lingen MW Ahmed O Liu J Nagilla M Macleod KF & Cohen EE p62/SQSTM1 accumulation in squamous cell carcinoma of head and neck predicts sensitivity to phosphatidylinositol 3-kinase pathway inhibitors. PLoS One. 9(3) e90171 2014. DOI: 10.1371/journal.pone.0090171
38 Liu JL Chen FF Lung J Lo CH Lee FH Lu YC & Hung CH Prognostic significance of p62/SQSTM1 subcellular localization and LC3B in oral squamous cell carcinoma. Br J Cancer. 111(5) 944 - 954 2014. DOI: 10.1038/bjc.2014.355
39 Ren F Shu G Liu G Liu D Zhou J Yuan L & Zhou J Knockdown of p62/sequestosome 1 attenuates autophagy and inhibits colorectal cancer cell growth. Mol Cell Biochem. 385(1-2) 95 - 102 2014. DOI: 10.1007/s11010-013-1818-0
40 Deng D Luo K Liu H Nie X Xue L Wang R Xu Y Cui J Shao N & Zhi F p62 acts as an oncogene and is targeted by miR-124-3p in glioma. Cancer Cell Int. 19 280 2019. DOI: 10.1186/s12935-019-1004-x
41 Lei C Zhao B Liu L Zeng X Yu Z & Wang X Expression and clinical significance of p62 protein in colon cancer. Medicine (Baltimore). 99(3) e18791 2020. DOI: 10.1097/MD.0000000000018791
42 Li T Jiang D & Wu K p62 promotes bladder cancer cell growth by activating KEAP1/NRF2-dependent antioxidative response. Cancer Sci. 111(4) 1156 - 1164 2020. DOI: 10.1111/cas.14321
43 Yang W Niu L Zhao X Duan L Li Y Wang X Zhang Y Zhou W Liu J Zhao Q Han Y Fan D & Hong L Development and validation of a survival model based on autophagy-associated genes for predicting prognosis of hepatocellular carcinoma. Am J Transl Res. 12(10) 6705 - 6722 2020.
44 Kobayashi T Ishida M Miki H Matsumi Y Fukui T Hamada M Tsuta K & Sekimoto M p62 is a useful predictive marker for tumour regression after chemoradiation therapy in patients with advanced rectal cancer: an immunohistochemical study. Colorectal Dis. 23(5) 1083 - 1090 2021. DOI: 10.1111/codi.15486
45 Yu F Ma R Liu C Zhang L Feng K Wang M & Yin D SQSTM1/p62 promotes cell growth and triggers autophagy in papillary thyroid cancer by regulating the AKT/AMPK/mTOR signaling pathway. Front Oncol. 11 638701 2021. DOI: 10.3389/fonc.2021.638701
46 Inoue D Suzuki T Mitsuishi Y Miki Y Suzuki S Sugawara S Watanabe M Sakurada A Endo C Uruno A Sasano H Nakagawa T Satoh K Tanaka N Kubo H Motohashi H & Yamamoto M Accumulation of p62/SQSTM1 is associated with poor prognosis in patients with lung adenocarcinoma. Cancer Sci. 103(4) 760 - 766 2012. DOI: 10.1111/j.1349-7006.2012.02216.x
47 Iwadate R Inoue J Tsuda H Takano M Furuya K Hirasawa A Aoki D & Inazawa J High expression of SQSTM1/p62 protein is associated with poor prognosis in epithelial ovarian cancer. Acta Histochem Cytochem. 47(6) 295 - 301 2014. DOI: 10.1267/ahc.14048
48 Iwadate R Inoue J Tsuda H Takano M Furuya K Hirasawa A Aoki D & Inazawa J High expression of p62 protein is associated with poor prognosis and aggressive phenotypes in endometrial cancer. Am J Pathol. 185(9) 2523 - 2533 2015. DOI: 10.1016/j.ajpath.2015.05.008
49 Arai A Chano T Ikebuchi K Hama Y Ochi Y Tameno H & Shimada T p62/SQSTM1 levels predicts radiotherapy resistance in hypopharyngeal carcinomas. Am J Cancer Res. 7(4) 881 - 891 2017.
50 Thompson HGR Harris JW Wold BJ Lin F & Brody JP p62 overexpression in breast tumors and regulation by prostate-derived Ets factor in breast cancer cells. Oncogene. 22(15) 2322 - 2333 2003. DOI: 10.1038/sj.onc.1206325
51 Xu LZ Li SS Zhou W Kang ZJ Zhang QX Kamran M Xu J Liang DP Wang CL Hou ZJ Wan XB Wang HJ Lam EW Zhao ZW & Liu Q p62/SQSTM1 enhances breast cancer stem-like properties by stabilizing MYC mRNA. Oncogene. 36(3) 304 - 317 2017. DOI: 10.1038/onc.2016.202
52 Li SS Xu LZ Zhou W Yao S Wang CL Xia JL Wang HF Kamran M Xue XY Dong L Wang J Ding XD Bella L Bugeon L Xu J Zheng FM Dallman MJ Lam EWF & Liu Q p62/SQSTM1 interacts with vimentin to enhance breast cancer metastasis. Carcinogenesis. 38(11) 1092 - 1103 2017. DOI: 10.1093/carcin/bgx099
53 Luo RZ Yuan ZY Li M Xi SY Fu J & He J Accumulation of p62 is associated with poor prognosis in patients with triple-negative breast cancer. Onco Targets Ther. 6 883 - 888 2013. DOI: 10.2147/OTT.S46222
54 Yokota A Hiramoto M Hino H Tokuhisa M Miyazaki M Kazama H Takano N & Miyazawa K Sequestosome 1 (p62) accumulation in breast cancer cells suppresses progesterone receptor expression via argonaute 2. Biochem Biophys Res Commun. 531(2) 256 - 263 2020. DOI: 10.1016/j.bbrc.2020.07.058
55 Ozaki A Motomura H Tamori S Onaga C Nagashima Y Kotori M Matsuda C Matsuda A Mochizuki N Sato T Hara Y Sato K Miyagi Y Nagashima Y Hanawa T Harada Y Xiong Y Sasaki K Ohno S & Akimoto K High Expression of p62 and ALDH1A3 is associated with poor prognosis in luminal B breast cancer. Anticancer Res. 42(7) 3299 - 3312 2022. DOI: 10.21873/anticanres.15818
56 Ozaki A Matsuda A Maemura Y Tada Y Kasai T Nagashima Y Onaga C Hara Y Kitabatake K Tsukimoto M Tamori S Sasaki K Ohno S & Akimoto K Luminal B breast cancer coexpressing p62 and ALDH1A3 is less susceptible to radiotherapy. Anticancer Res. 44(1) 37 - 47 2024. DOI: 10.21873/anticanres.16786
57 Curtis C Shah SP Chin SF Turashvili G Rueda OM Dunning MJ Speed D Lynch AG Samarajiwa S Yuan Y Gräf S Ha G Haffari G Bashashati A Russell R McKinney S METABRIC Group Langerød A Green A Provenzano E Wishart G Pinder S Watson P Markowetz F Murphy L Ellis I Purushotham A Børresen-Dale AL Brenton JD Tavaré S Caldas C & Aparicio S The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 486(7403) 346 - 352 2012. DOI: 10.1038/nature10983
58 Pereira B Chin SF Rueda OM Vollan HK Provenzano E Bardwell HA Pugh M Jones L Russell R Sammut SJ Tsui DW Liu B Dawson SJ Abraham J Northen H Peden JF Mukherjee A Turashvili G Green AR McKinney S Oloumi A Shah S Rosenfeld N Murphy L Bentley DR Ellis IO Purushotham A Pinder SE Børresen-Dale AL Earl HM Pharoah PD Ross MT Aparicio S & Caldas C The somatic mutation profiles of 2,433 breast cancers refines their genomic and transcriptomic landscapes. Nat Commun. 7 11479 2016. DOI: 10.1038/ncomms11479
59 Liu J Lichtenberg T Hoadley KA Poisson LM Lazar AJ Cherniack AD Kovatich AJ Benz CC Levine DA Lee AV Omberg L Wolf DM Shriver CD Thorsson V Cancer Genome Atlas Research Network & Hu H An integrated TCGA pan-cancer clinical data resource to drive high-quality survival outcome analytics. Cell. 173(2) 400 - 416.e11 2018. DOI: 10.1016/j.cell.2018.02.052
60 Cerami E Gao J Dogrusoz U Gross BE Sumer SO Aksoy BA Jacobsen A Byrne CJ Heuer ML Larsson E Antipin Y Reva B Goldberg AP Sander C & Schultz N The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2(5) 401 - 404 2012. DOI: 10.1158/2159-8290.CD-12-0095
61 Gao J Aksoy BA Dogrusoz U Dresdner G Gross B Sumer SO Sun Y Jacobsen A Sinha R Larsson E Cerami E Sander C & Schultz N Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 6(269) pl1 2013. DOI: 10.1126/scisignal.2004088
62 Nagashima Y Sasaki K Chiwaki R Ishii H Nohata K Maemura Y Kasai T Ozaki A Tamori S Ohno S & Akimoto K High Expression of PKCζ And CTNNBIP1 is associated with poor prognosis in luminal B breast cancer. Cancer Genomics Proteomics. 22(4) 538 - 556 2025. DOI: 10.21873/cgp.20520
63 Chen J Gao Z Li X Shi Y Tang Z Liu W Zhang X Huang A Luo X Gao Q Ding G Song K Zhou J Fan J Fu X & Ding Z SQSTM1/p62 in intrahepatic cholangiocarcinoma promotes tumor progression via epithelial-mesenchymal transition and mitochondrial function maintenance. Cancer Med. 12(1) 459 - 471 2023. DOI: 10.1002/cam4.4908
64 Hennig P Fenini G Di Filippo M Karakaya T & Beer HD The pathways underlying the multiple roles of p62 in inflammation and cancer. Biomedicines. 9(7) 707 2021. DOI: 10.3390/biomedicines9070707
65 Qi JL He JR Liu CB Jin SM Yang X Bai HM & Ma YB SQSTM1/p62 regulate breast cancer progression and metastasis by inducing cell cycle arrest and regulating immune cell infiltration. Genes Dis. 9(5) 1332 - 1344 2021. DOI: 10.1016/j.gendis.2021.03.008
66 Li D He C Ye F Ye E He H Chen G & Zhang J p62 overexpression promotes bone metastasis of lung adenocarcinoma out of LC3-dependent autophagy. Front Oncol. 11 609548 2021. DOI: 10.3389/fonc.2021.609548
67 Umemura A He F Taniguchi K Nakagawa H Yamachika S Font-Burgada J Zhong Z Subramaniam S Raghunandan S Duran A Linares JF Reina-Campos M Umemura S Valasek MA Seki E Yamaguchi K Koike K Itoh Y Diaz-Meco MT Moscat J & Karin M p62, upregulated during preneoplasia, induces hepatocellular carcinogenesis by maintaining survival of stressed HCC-initiating cells. Cancer Cell. 29(6) 935 - 948 2016. DOI: 10.1016/j.ccell.2016.04.006
68 Saito T Ichimura Y Taguchi K Suzuki T Mizushima T Takagi K Hirose Y Nagahashi M Iso T Fukutomi T Ohishi M Endo K Uemura T Nishito Y Okuda S Obata M Kouno T Imamura R Tada Y Obata R Yasuda D Takahashi K Fujimura T Pi J Lee MS Ueno T Ohe T Mashino T Wakai T Kojima H Okabe T Nagano T Motohashi H Waguri S Soga T Yamamoto M Tanaka K & Komatsu M p62/Sqstm1 promotes malignancy of HCV-positive hepatocellular carcinoma through Nrf2-dependent metabolic reprogramming. Nat Commun. 7 12030 2016. DOI: 10.1038/ncomms12030
69 Reina-Campos M Shelton PM Diaz-Meco MT & Moscat J Metabolic reprogramming of the tumor microenvironment by p62 and its partners. Biochim Biophys Acta Rev Cancer. 1870(1) 88 - 95 2018. DOI: 10.1016/j.bbcan.2018.04.010
70 Yan XY Zhang Y Zhang JJ Zhang LC Liu YN Wu Y Xue YN Lu SY Su J & Sun LK p62/SQSTM1 as an oncotarget mediates cisplatin resistance through activating RIP1-NF-κB pathway in human ovarian cancer cells. Cancer Sci. 108(7) 1405 - 1413 2017. DOI: 10.1111/cas.13276
71 Wang Z Zhang J Li M Kong L & Yu J The expression of p-p62 and nuclear Nrf2 in esophageal squamous cell carcinoma and association with radioresistance. Thorac Cancer. 11(1) 130 - 139 2020. DOI: 10.1111/1759-7714.13252
72 Tsai CL Lin CY Chao A Lee YS Wu RC Tsai CN Yen CF & Chao AS GPR30 activation by 17β-estradiol promotes p62 phosphorylation and increases estrogen receptor α protein expression by inducing its release from a complex formed with KEAP1. J Pers Med. 11(9) 906 2021. DOI: 10.3390/jpm11090906
73 Semina SE Pal P Kansara NS Huggins RJ Alarid ET Greene GL & Frasor J Selective pressure of endocrine therapy activates the integrated stress response through NFκB signaling in a subpopulation of ER positive breast cancer cells. Breast Cancer Res. 24(1) 19 2022. DOI: 10.1186/s13058-022-01515-1
74 Ryoo IG Choi BH Ku SK & Kwak MK High CD44 expression mediates p62-associated NFE2L2/NRF2 activation in breast cancer stem cell-like cells: Implications for cancer stem cell resistance. Redox Biol. 17 246 - 258 2018. DOI: 10.1016/j.redox.2018.04.015
75 Nagashima Y Sasaki K Nohata K Chiwaki R Maemura Y Kasai T Ozaki A Tamori S Ohno S & Akimoto K High expression of PKCζ and ALDH1A3 is associated with poor prognosis in luminal B breast cancer. Anticancer Res. 45(10) 4357 - 4372 2025.