General Information of the Disease (ID: DIS00038)
Name
Breast cancer
ICD
ICD-11: 2C60
Full List of Target(s) of This Ferroptosis-centered Disease
Voltage-dependent anion-selective channel protein 2 (VDAC2)
In total 1 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [1]
Target for Ferroptosis Driver
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Robustaflavone Investigative
Responsed Regulator E3 ubiquitin-protein ligase NEDD4 (NEDD4) Suppressor
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
Response regulation Robustaflavone A strikingly induced MCF-7 nonapoptotic cell death through ferroptosis by enhancing the expression of VDAC2 channels and reducing the expression of Nedd4 E3 ubiquitin ligase, leading to lipid peroxidation and ROS production. The results suggested that RF-A has potential as a novel breast cancer treatment through its regulation of the mitochondrial VDAC2 and Nedd4 pathways.
Phospholipid hydroperoxide glutathione peroxidase (GPX4)
In total 10 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [2]
Target for Ferroptosis Suppressor
Responsed Disease Breast adenocarcinoma [ICD-11: 2C60]
Responsed Drug JQ1 Investigative
Responsed Regulator Bromodomain-containing protein 4 (BRD4) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Apoptosis hsa04210
Autophagy hsa04140
Cell Process Cell ferroptosis
Cell apoptosis
Cell autophagy
Cell proliferation
Cell migration
Cell invasion
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Hs-578T cells Invasive breast carcinoma Homo sapiens CVCL_0332
NCI-H1299 cells Lung large cell carcinoma Homo sapiens CVCL_0060
A-549 cells Lung adenocarcinoma Homo sapiens CVCL_0023
MCF-10A cells Normal Homo sapiens CVCL_0598
In Vivo Model
Female athymic BALB/c nude mice (4-6-week old) were obtained from Beijing Vital River Laboratory Animal Technology Co., Ltd. (Beijing, China). Approximately 1 x 107 cells (A549) in 200 uL of serum-free medium and Matrigel solution were injected directly into the right axilla of each mouse. Tumor growth was measured with calipers every 3 days.

    Click to Show/Hide
Response regulation Ferroptosis was induced under (+)-JQ1 treatment and BRD4 knockdown, indicating that (+)-JQ1 induces ferroptosis via BRD4 inhibition in breast adenocarcinoma. In addition, expression of the ferroptosis-associated genes GPX4, SLC7A11, and SLC3A2 was downregulated under (+)-JQ1 treatment. Moreover, JQ1 treatment and BRD4 knockdown led to decreased FTH1 expression.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [3]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Ketamine Investigative
Responsed Regulator Histone acetyltransferase KAT5 (KAT5) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Apoptosis hsa04210
Cell Process Cell ferroptosis
Cell apoptosis
Cell proliferation
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
T-47D cells Invasive breast carcinoma Homo sapiens CVCL_0553
Response regulation The treatment of Ketamine induced the levels of MDA, lipid ROS, and Fe2+, while KAT5 or GPX4 overexpression could reverse this effect in breast cancer cells. Ketamine suppresses proliferation and induces ferroptosis and apoptosis of breast cancer cells by targeting KAT5/GPX4 axis. Ketamine may serve as a potential therapeutic strategy for breast cancer.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [4]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Metformin Investigative
Responsed Regulator hsa-miR-324-3p (miRNA) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Glutathione metabolism hsa00480
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
In Vivo Model
Six-week-old athymic nude mice were obtained from Nanjing Biomedical Research Institute of Nanjing University (Nanjing, China). Mice were divided into five groups: sham group, metformin group, metformin + NC group, metformin + miR-324-3p overexpression group, and metformin + miR-324-3p knockdown group (n = 6 in each group). Mice were injected with 3 x 106 MDA-MB-231 cells subcutaneously into the right flank. For the miR-324-3p overexpression or knockdown in the mice, two groups of mice were treated with miR-324-3p overexpression or knockdown lentivirus (GenePharma), respectively, by intratumoral injection of 50 ul of lentivirus (4 x 107 IU/ml) after the tumor cell injection. One day after tumor cell inoculation, the sham-treated group was treated with PBS and metformin-treated groups were treated with 200 mg/kg metformin every 2 days through intraperitoneal injection.

    Click to Show/Hide
Response regulation Metformin promotes ferroptosis of breast cancer by targeting the miR-324-3p/GPX4 axis. The effect of miR-324-3p was mediated by directly targeting glutathione peroxidase 4 (GPX4). Metformin could act as a potential anti-cancer agent through the induction of ferroptosis.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [5]
Target for Ferroptosis Suppressor
Responsed Disease Triple-negative breast cancer [ICD-11: 2C60]
Responsed Drug Simvastatin Investigative
Responsed Regulator 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
In Vivo Model
MDA-MB-231 cells were injected to subcutaneous of mice to build a tumor model. When the tumor volume reaches about 60 mm3, all mice were randomly divided into five groups (n = 5) for various treatments. Then, mice were treated with PBS, Fe3O4@PCBMA, SIM, Fe3O4-SIM and Fe3O4@PCBMA-SIM through injected intravenously. The injected doses of SIM were 4 mg/kg body weight in each mouse on days 0, 3, 6, and 9.

    Click to Show/Hide
Response regulation The study presented the ferroptosis nanomedicine by loading simvastatin (SIM), a ferroptosis drugs, into zwitterionic polymer coated of magnetic nanoparticles (Fe3O4@PCBMA), thereby improving the therapeutic effect of triple negative breast cancer. SIM could inhibit the expression of HMGCR to downregulate the mevalonate (MVA) pathway and glutathione peroxidase 4 (GPX4), thereby inducing cancer cell ferroptosis.
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target [10]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Etoposide Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
MCF-10A cells Normal Homo sapiens CVCL_0598
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Response regulation The combined treatment of etoposide and erastin synergistically induced oxidative stress and lipid peroxidation, while suppressing glutathione peroxidase activity in breast cancer cells. More importantly, the combination treatment synergistically increased iron accumulation, which was associated with altered expression of IREB2/FPN1. Additionally, ferroptosis-regulating proteins ACSF2 and GPX4 were altered more potently by the combination treatment, compared to untreated cells and erastin treatment alone (p<0.05).
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target [19]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Lycium barbarumpolysaccharide Investigative
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Response regulation Lycium barbarum polysaccharide (LBP) effectively inhibited proliferation of breast cancer cells and promoted ferroptosis by modulation of the xCT/GPX4 pathway. GPX4 inactivity and repression of SLC7A11 (the gene for xCT) result in ROS accumulation, thereby modulating ferroptosis.
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target [20]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Tubastatin A Investigative
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
In Vivo Model
Female 5-week-old athymic nude mice were obtained from Sun Yat-sen University. All mice were kept under specific-pathogen free conditions in the animal facility of Sun Yat-sen University Cancer Centre. Cancer cells were suspended and counted in 1 x DMEM, and 2 x 106 MDA-MB-231 cells were injected into mice subcutaneously. When the tumours reached 50-100 mm3, the mice were randomly assigned to different treatment groups. Tumours were irradiated with a JL Shepherd Mark I-68A irradiator at a dose of 10 Gy. Tub was dissolved in solvent containing 1% DMSO, 30% polyethylene glycol, 1% Tween 80 and 68% H2O and then subcutaneously administered to mice at a dose of 2.5 mg/kg once a day. Lipro-1 diluted in PBS was intraperitoneally injected daily at a dose of 10 mg/kg. Tub or Lipro-1 was administered three times before irradiation followed by continued daily administration until the endpoint, as indicated in the corresponding figures.

    Click to Show/Hide
Response regulation Tubastatin A (Tub) as a novel GPX4 inhibitor that induced ferroptosis through large-scale drug screening. We showed that IR-mediated GPX4 expression restrained ferroptosis to drive radioresistance in breast cancer.
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target [15]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator Plasmanylethanolamine desaturase (PEDS1) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Autophagy hsa04140
Cell Process Cell ferroptosis
Cell autophagy
Cell proliferation
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
In Vivo Model
Twenty BALB/c nude female mice (4- to 6-week-old) were purchased from Charles River Laboratories (Beijing, China) to establish tumorigenesis (5 mice in each). We injected MCF-7 or MDA-MB-231 cells (4 x 106) transfected with the stable knockdown and over-expressing TMEM189 into the flanks of mice to construct tumorigenesis models, respectively. Meanwhile, the sh-Con and empty vector were served as the control groups for each model.

    Click to Show/Hide
Response regulation TMEM189 (PEDS1) could inhibit autophagy to mediate ferroptosis in breast cancer cells. Moreover, TMEM189 ablation strongly up-regulated LC3BII and transferrin receptor 1 (TfR1) expression levels in breast cancer cells, whereas down-regulated p62 and GPX4.
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target [21]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator RUNX1-IT1 (IncRNA) Suppressor
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model T-47D cells Invasive breast carcinoma Homo sapiens CVCL_0553
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
In Vivo Model
MDA-MB-231 cells with stable RUNX1-IT1 knockdown were injected into NOD/SCID mice, which were randomly divided into three groups, five in each group. The protocol of establishment of breast cancer orthotopic transplantation model was described in our previous study.

    Click to Show/Hide
Response regulation RUNX1-IT1 promotes breast cancer carcinogenesis through blocking ferroptosis via elevating GPX4, targeting of the previously unappreciated regulatory axis of RUNX1-IT1/IGF2BP1/GPX4 may be a promising treatment for patient with breast cancer.
Experiment 10 Reporting the Ferroptosis-centered Disease Response by This Target [21]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator Insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) Suppressor
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model T-47D cells Invasive breast carcinoma Homo sapiens CVCL_0553
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
In Vivo Model
MDA-MB-231 cells with stable RUNX1-IT1 knockdown were injected into NOD/SCID mice, which were randomly divided into three groups, five in each group. The protocol of establishment of breast cancer orthotopic transplantation model was described in our previous study.

    Click to Show/Hide
Response regulation RUNX1-IT1 promotes breast cancer carcinogenesis through blocking ferroptosis via elevating GPX4, targeting of the previously unappreciated regulatory axis of RUNX1-IT1/IGF2BP1/GPX4 may be a promising treatment for patient with breast cancer.
Ferritin heavy chain (FTH1)
In total 1 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [2]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Breast adenocarcinoma [ICD-11: 2C60]
Responsed Drug JQ1 Investigative
Responsed Regulator Bromodomain-containing protein 4 (BRD4) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Apoptosis hsa04210
Autophagy hsa04140
Cell Process Cell ferroptosis
Cell apoptosis
Cell autophagy
Cell proliferation
Cell migration
Cell invasion
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Hs-578T cells Invasive breast carcinoma Homo sapiens CVCL_0332
NCI-H1299 cells Lung large cell carcinoma Homo sapiens CVCL_0060
A-549 cells Lung adenocarcinoma Homo sapiens CVCL_0023
MCF-10A cells Normal Homo sapiens CVCL_0598
In Vivo Model
Female athymic BALB/c nude mice (4-6-week old) were obtained from Beijing Vital River Laboratory Animal Technology Co., Ltd. (Beijing, China). Approximately 1 x 107 cells (A549) in 200 uL of serum-free medium and Matrigel solution were injected directly into the right axilla of each mouse. Tumor growth was measured with calipers every 3 days.

    Click to Show/Hide
Response regulation Ferroptosis was induced under (+)-JQ1 treatment and BRD4 knockdown, indicating that (+)-JQ1 induces ferroptosis via BRD4 inhibition in breast adenocarcinoma. In addition, expression of the ferroptosis-associated genes GPX4, SLC7A11, and SLC3A2 was downregulated under (+)-JQ1 treatment. Moreover, JQ1 treatment and BRD4 knockdown led to decreased FTH1 expression.
Cystine/glutamate transporter (SLC7A11)
In total 12 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [2]
Target for Ferroptosis Suppressor
Responsed Disease Breast adenocarcinoma [ICD-11: 2C60]
Responsed Drug JQ1 Investigative
Responsed Regulator Bromodomain-containing protein 4 (BRD4) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Apoptosis hsa04210
Autophagy hsa04140
Cell Process Cell ferroptosis
Cell apoptosis
Cell autophagy
Cell proliferation
Cell migration
Cell invasion
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Hs-578T cells Invasive breast carcinoma Homo sapiens CVCL_0332
NCI-H1299 cells Lung large cell carcinoma Homo sapiens CVCL_0060
A-549 cells Lung adenocarcinoma Homo sapiens CVCL_0023
MCF-10A cells Normal Homo sapiens CVCL_0598
In Vivo Model
Female athymic BALB/c nude mice (4-6-week old) were obtained from Beijing Vital River Laboratory Animal Technology Co., Ltd. (Beijing, China). Approximately 1 x 107 cells (A549) in 200 uL of serum-free medium and Matrigel solution were injected directly into the right axilla of each mouse. Tumor growth was measured with calipers every 3 days.

    Click to Show/Hide
Response regulation Ferroptosis was induced under (+)-JQ1 treatment and BRD4 knockdown, indicating that (+)-JQ1 induces ferroptosis via BRD4 inhibition in breast adenocarcinoma. In addition, expression of the ferroptosis-associated genes GPX4, SLC7A11, and SLC3A2 was downregulated under (+)-JQ1 treatment. Moreover, JQ1 treatment and BRD4 knockdown led to decreased FTH1 expression.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [6]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Lidocaine Investigative
Responsed Regulator hsa-miR-382-5p (miRNA) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Apoptosis hsa04210
Cell Process Cell ferroptosis
Cell apoptosis
Cell proliferation
Cell migration
Cell invasion
In Vitro Model SK-OV-3 cells Ovarian serous cystadenocarcinoma Homo sapiens CVCL_0532
T-47D cells Invasive breast carcinoma Homo sapiens CVCL_0553
In Vivo Model
SPF-level male nude mice aged 56weeks and weighted around 20 g were purchased from Vitalriver (China). All mice were maintained in a 12-hour circadian rhythm, and had free access to water and food. Cancer cells were subcutaneously injected into the right flank of mice. Lidocaine was administrated to mice at a dose of 1.5 mg per kg injected through the vail tails. For control group, the mice were treated with saline. Tumor volume and mice body weight were monitored every 5 days.

    Click to Show/Hide
Response regulation The ovarian and breast cancer cell proliferation was suppressed while cell apoptosis was induced by lidocaine in vitro. Lidocaine attenuated invasion and migration of ovarian and breast cancer cells as well. Regarding the mechanism, lidocaine downregulated solute carrier family 7 member 11 (SLC7A11) expression by enhancing microRNA-382-5p (miR-382-5p) in the cells.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [7]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Metformin Investigative
Responsed Regulator Ubiquitin-fold modifier 1 (UFM1) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
T-47D cells Invasive breast carcinoma Homo sapiens CVCL_0553
HCC1937 cells Breast ductal carcinoma Homo sapiens CVCL_0290
Bcap37 cells Breast carcinoma Homo sapiens CVCL_0164
HBL-100 cells Normal Homo sapiens CVCL_4362
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
In Vivo Model
T47D xenografts were established in 5-week-old nude mice (Shanghai SLAC Laboratory Animal Corporation) by inoculating 1 x 107 cells mixed with Matrigel (BD Biosciences) at 1:1 ratio (volume) into the abdominal mammary fat pad. When the tumor reached 50-100 mm3, the mice were assigned randomly into different treatment groups (DMSO, Metformin, SAS, and Metformin + SAS groups). Metformin (200 mg/kg/day) was provided in drinking water. Sulfasalazine was dissolved in dimethyl sulfoxide (DMSO), diluted in PBS, and then intraperitoneally injected into mice at a dose of 250 mg/kg once a day.

    Click to Show/Hide
Response regulation Metformin reduces the protein stability of SLC7A11, which is a critical ferroptosis regulator, by inhibiting its UFMylation process. Furthermore, metformin combined with sulfasalazine, the system xc-inhibitor, can work in a synergistic manner to induce ferroptosis and inhibit the proliferation of breast cancer cells.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [8]
Target for Ferroptosis Suppressor
Responsed Disease Triple-negative breast cancer [ICD-11: 2C60]
Responsed Drug Polyphyllin III Investigative
Responsed Regulator Krueppel-like factor 4 (KLF4) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Hs-578T cells Invasive breast carcinoma Homo sapiens CVCL_0332
MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
T-47D cells Invasive breast carcinoma Homo sapiens CVCL_0553
HBL-100 cells Normal Homo sapiens CVCL_4362
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
MDA-MB-453 cells Breast adenocarcinoma Homo sapiens CVCL_0418
In Vivo Model
MDA-MB-231 xenografts were established in 5 week-old BALB/C nude mice (Shanghai SLAC Laboratory Animal Corporation) by inoculating 1 x 106 cells mixed with Matrigel (BD Biosciences) at a 1:1 ratio into the abdominal mammary fat pad. When the tumor reached 50-100 mm3, the mice were assigned randomly into different treatment groups (DMSO, PPIII, SAS, and PPIII + SAS groups), and each group consisted of 5 mice. PPIII (5 mg/kg/day) and SAS (200 mg/kg/day) were dissolved in dimethyl sulfoxide (DMSO), diluted in PBS, and then intraperitoneally injected into mice at a dose of 10 ml/kg/d once a day.

    Click to Show/Hide
Response regulation Polyphyllin III, which is a major saponin extracted fromParis polyphyllarhizomes, exerted its proliferation-inhibitory effect on MDA-MB-231 triple-negative breast cancer cells mainly through ACSL4-mediated lipid peroxidation elevation and ferroptosis induction. Polyphyllin III treatment also induced KLF4-mediated protective upregulation of xCT(SLC7A11), which is the negative regulator of ferroptosis.
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target [31]
Target for Ferroptosis Suppressor
Responsed Disease Breast adenocarcinoma [ICD-11: 2C60]
Responsed Drug Sulfasalazine Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
Response regulation The combined effects of vorinostat with salazosulfapyridine (SASP) depend on the accumulation of ROS caused by a decrease in intracellular GSH levels, possibly due to SASP-mediated inhibition of xCT. xCT (coded by the SLC7A11 gene), a light chain subunit of the glutamate-cystine antiporter system Xc(-) in Breast adenocarcinoma.
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target [7]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Sulfasalazine Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
T-47D cells Invasive breast carcinoma Homo sapiens CVCL_0553
HCC1937 cells Breast ductal carcinoma Homo sapiens CVCL_0290
Bcap37 cells Breast carcinoma Homo sapiens CVCL_0164
HBL-100 cells Normal Homo sapiens CVCL_4362
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
In Vivo Model
T47D xenografts were established in 5-week-old nude mice (Shanghai SLAC Laboratory Animal Corporation) by inoculating 1 x 107 cells mixed with Matrigel (BD Biosciences) at 1:1 ratio (volume) into the abdominal mammary fat pad. When the tumor reached 50-100 mm3, the mice were assigned randomly into different treatment groups (DMSO, Metformin, SAS, and Metformin + SAS groups). Metformin (200 mg/kg/day) was provided in drinking water. Sulfasalazine was dissolved in dimethyl sulfoxide (DMSO), diluted in PBS, and then intraperitoneally injected into mice at a dose of 250 mg/kg once a day.

    Click to Show/Hide
Response regulation Metformin reduces the protein stability of SLC7A11, which is a critical ferroptosis regulator, by inhibiting its UFMylation process. Furthermore, metformin combined with sulfasalazine, the system xc-inhibitor, can work in a synergistic manner to induce ferroptosis and inhibit the proliferation of breast cancer cells.
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target [32]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator Circ-BGN (circRNA) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ubiquitin mediated proteolysis hsa04120
Cell Process Cell ferroptosis
In Vitro Model BT-474 cells Invasive breast carcinoma Homo sapiens CVCL_0179
SK-BR-3 cells Breast adenocarcinoma Homo sapiens CVCL_0033
In Vivo Model
A total of 24 NOD/SCID mice were purchased from Model Animal Research Center and grown under specific-pathogen-free condition. Mice were randomly divided into three groups (n = 8 per group), BT474-Tr cells were inoculated orthotopically onto the abdominal mammary fat pad. After 1 week, mice were treated with erastin (15 mg/kg intraperitoneal, twice every other day). Erastin was dissolved in 5% DMSO + corn oil (C8267, Sigma). To better dissolve erastin, we warmed the tube at 37 water bath and shook it gently. At the end of the sixth week, all mice were sacrificed, tumor tissues were collected and weighed.

    Click to Show/Hide
Response regulation Knockdown of circ-BGN inhibited breast cancer cell viability and notably restored its sensitivity to trastuzumab. Further, we found that circ-BGN could directly bind to OTUB1 and SLC7A11, enhancing OTUB1-mediated SLC7A11 deubiquitination and thereby inhibiting ferroptosis.
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target [33]
Target for Ferroptosis Suppressor
Responsed Disease Breast adenocarcinoma [ICD-11: 2C60]
Responsed Regulator Mucin-1 (MUC1) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MDA-MB-468 cells Breast adenocarcinoma Homo sapiens CVCL_0419
MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
HEK-293T cells Normal Homo sapiens CVCL_0063
Response regulation MUC1-C binds directly with CD44v and in turn promotes stability of xCT (SLC7A11) in the cell membrane in breast adenocarcinoma. The interaction between 2MUC1-C and xCT is further supported by the demonstration that targeting xCT with silencing or the inhibitor sulfasalazine suppresses MUC1 gene transcription by increasing histone and DNA methylation on the MUC1 promoter.
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target [26]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator hsa-miR-5096 (miRNA) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MDA-MB-468 cells Breast adenocarcinoma Homo sapiens CVCL_0419
MDA-MB-453 cells Breast adenocarcinoma Homo sapiens CVCL_0418
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
SK-BR-3 cells Breast adenocarcinoma Homo sapiens CVCL_0033
T-47D cells Invasive breast carcinoma Homo sapiens CVCL_0553
MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
ZR-75-1 cells Invasive breast carcinoma Homo sapiens CVCL_0588
MCF-10A cells Normal Homo sapiens CVCL_0598
In Vivo Model
Mating was setup 2 days prior to injection day and zebrafish embryos were collected and incubated in E3 embryo medium (5 mM NaCl, 0.17 mM KCl, 10 mM HEPES, 0.33 mM MgSO4·7H2O, 0.33 mM CaCl2·6H2O, and 0.00001% methylene blue) containing 0.2 mM N-phenyl-thiourea (PTU) (catalog no: P7629, Sigma-Aldrich). Two days post-fertilization, the chorion was removed manually using fine forceps, and the embryos were anesthetized using E3 medium containing 200 mg/L Ethyl 3-aminobenzoate methanesulfonate (Tricaine) (catalog no: A5040, Sigma-Aldrich). Anesthetized embryos were mounted in 0.7% low melting agarose containing 200 ug/ml of Tricaine and were microinjected with 500 cells in the yolk sac using Nanoject III (catalog no: 3-000-207; Drummond Scientific Company, PA, USA). At 1 day post-injection (dpi), embryos with similar graft size were selected and imaged using both bright field and RFP channels and incubated in E3-PTU medium containing 5 ug/ml doxycycline at 34 until further imaging. At 4 dpi, embryos were anesthetized and imaged again using both bright field and RFP channels using Olympus IX-73 microscope. Cells that migrated outside the yolk sac (injection site) were represented by a notable fluorescent dot and considered a metastatic event; these were counted manually for all embryos in each experimental group.

    Click to Show/Hide
Response regulation The present study demonstrated that miR-5096 targets and downregulates SLC7A11, thereby providing a mechanistic basis for ferroptosis in human breast cancer cells. In addition, miR-5096 induced cell death via ferroptosis, characterized by mitochondrial shrinkage with partial loss of cristae with simultaneous changes in ACSL4, ROS, lipid ROS, OH-, reactive iron, GSH, and MMP levels.
Experiment 10 Reporting the Ferroptosis-centered Disease Response by This Target [32]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator Ubiquitin thioesterase OTUB1 (OTUB1) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ubiquitin mediated proteolysis hsa04120
Cell Process Cell ferroptosis
In Vitro Model BT-474 cells Invasive breast carcinoma Homo sapiens CVCL_0179
SK-BR-3 cells Breast adenocarcinoma Homo sapiens CVCL_0033
In Vivo Model
A total of 24 NOD/SCID mice were purchased from Model Animal Research Center and grown under specific-pathogen-free condition. Mice were randomly divided into three groups (n = 8 per group), BT474-Tr cells were inoculated orthotopically onto the abdominal mammary fat pad. After 1 week, mice were treated with erastin (15 mg/kg intraperitoneal, twice every other day). Erastin was dissolved in 5% DMSO + corn oil (C8267, Sigma). To better dissolve erastin, we warmed the tube at 37 water bath and shook it gently. At the end of the sixth week, all mice were sacrificed, tumor tissues were collected and weighed.

    Click to Show/Hide
Response regulation Knockdown of circ-BGN inhibited breast cancer cell viability and notably restored its sensitivity to trastuzumab. Further, we found that circ-BGN could directly bind to OTUB1 and SLC7A11, enhancing OTUB1-mediated SLC7A11 deubiquitination and thereby inhibiting ferroptosis.
Experiment 11 Reporting the Ferroptosis-centered Disease Response by This Target [34]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator E3 ubiquitin-protein ligase NEDD4-like (NEDD4L) Driver
Pathway Response Fatty acid metabolism hsa01212
Ubiquitin mediated proteolysis hsa04120
Cell Process Cell ferroptosis
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
ZR-75-1 cells Invasive breast carcinoma Homo sapiens CVCL_0588
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Response regulation SLC7A11 expression was regulated by both ESR1 and NEDD4L, in opposite ways. For the first time, the study elucidated that ESR1 and NEDD4L functioned together after radiation treatment and finally induced ferroptosis in breast cancer cells, which provides novel insight into the guidance of clinical treatment of breast cancer.
Experiment 12 Reporting the Ferroptosis-centered Disease Response by This Target [34]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator Estrogen receptor (ESR1) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ubiquitin mediated proteolysis hsa04120
Cell Process Cell ferroptosis
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
ZR-75-1 cells Invasive breast carcinoma Homo sapiens CVCL_0588
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Response regulation SLC7A11 expression was regulated by both ESR1 and NEDD4L, in opposite ways. For the first time, the study elucidated that ESR1 and NEDD4L functioned together after radiation treatment and finally induced ferroptosis in breast cancer cells, which provides novel insight into the guidance of clinical treatment of breast cancer.
4F2 cell-surface antigen heavy chain (SLC3A2)
In total 1 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [2]
Target for Ferroptosis Suppressor
Responsed Disease Breast adenocarcinoma [ICD-11: 2C60]
Responsed Drug JQ1 Investigative
Responsed Regulator Bromodomain-containing protein 4 (BRD4) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Apoptosis hsa04210
Autophagy hsa04140
Cell Process Cell ferroptosis
Cell apoptosis
Cell autophagy
Cell proliferation
Cell migration
Cell invasion
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Hs-578T cells Invasive breast carcinoma Homo sapiens CVCL_0332
NCI-H1299 cells Lung large cell carcinoma Homo sapiens CVCL_0060
A-549 cells Lung adenocarcinoma Homo sapiens CVCL_0023
MCF-10A cells Normal Homo sapiens CVCL_0598
In Vivo Model
Female athymic BALB/c nude mice (4-6-week old) were obtained from Beijing Vital River Laboratory Animal Technology Co., Ltd. (Beijing, China). Approximately 1 x 107 cells (A549) in 200 uL of serum-free medium and Matrigel solution were injected directly into the right axilla of each mouse. Tumor growth was measured with calipers every 3 days.

    Click to Show/Hide
Response regulation Ferroptosis was induced under (+)-JQ1 treatment and BRD4 knockdown, indicating that (+)-JQ1 induces ferroptosis via BRD4 inhibition in breast adenocarcinoma. In addition, expression of the ferroptosis-associated genes GPX4, SLC7A11, and SLC3A2 was downregulated under (+)-JQ1 treatment. Moreover, JQ1 treatment and BRD4 knockdown led to decreased FTH1 expression.
Unspecific Target
In total 34 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [9]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Erastin Investigative
Responsed Regulator Discoidin domain-containing receptor 2 (DDR2) Driver
Pathway Response Ferroptosis hsa04216
Hippo signaling pathway hsa04390
Cell adhesion molecules hsa04514
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model SUM52PE cells Breast carcinoma Homo sapiens CVCL_3425
ZR-75-1 cells Invasive breast carcinoma Homo sapiens CVCL_0588
BT-474 cells Invasive breast carcinoma Homo sapiens CVCL_0179
MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
BT-20 cells Invasive breast carcinoma of no special type Homo sapiens CVCL_0178
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Hs-578T cells Invasive breast carcinoma Homo sapiens CVCL_0332
MDA-MB-157 cells Breast carcinoma Homo sapiens CVCL_0618
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
Response regulation Discoidin Domain Receptor Tyrosine Kinase 2 (DDR2), the receptor for collagen I, is highly expressed in ferroptosis-sensitive recurrent tumor cells and human mesenchymal breast cancer cells. Erastin treatment induces DDR2 upregulation and phosphorylation, independent of collagen I. Furthermore, DDR2 knockdown in recurrent tumor cells reduces clonogenic proliferation.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [10]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Etoposide Investigative
Responsed Regulator Medium-chain acyl-CoA ligase ACSF2, mitochondrial (ACSF2) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
MCF-10A cells Normal Homo sapiens CVCL_0598
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Response regulation The combined treatment of etoposide and erastin synergistically induced oxidative stress and lipid peroxidation, while suppressing glutathione peroxidase activity in breast cancer cells. More importantly, the combination treatment synergistically increased iron accumulation, which was associated with altered expression of IREB2/FPN1. Additionally, ferroptosis-regulating proteins ACSF2 and GPX4 were altered more potently by the combination treatment, compared to untreated cells and erastin treatment alone (p<0.05).
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [11]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug I-BET151 Investigative
Responsed Regulator Bromodomain-containing protein 4 (BRD4) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
MDA-MB-468 cells Breast adenocarcinoma Homo sapiens CVCL_0419
SK-BR-3 cells Breast adenocarcinoma Homo sapiens CVCL_0033
In Vivo Model
Athymic nu/nu mice (5 to 6-week-old female) were acquired from the Silaike Experimental Animal Co. Ltd (Shanghai, China). All mice were randomly allocated to different groups, 1 x 106 MDA-MB-231 cells were subcutaneously inoculated into the right flanks. Ten days later, mice were intraperitoneally injected with PBS including DMSO, JQ1 (50 mg/kg), Bufalin (1 mg/kg) and SR1848 (50 mg/kg), every 2-4 days for 8 times, and the tumor volumes and mice weigh were monitored and recorded every 2-3 days.

    Click to Show/Hide
Response regulation BET (BRD2, BRD3, BRD4, BRDT) inhibitors (BETi) exert an excellent anti-cancer activity in breast cancer. BETi JQ1 and I-BET151 exhibited anti-cancer effects in breast cancer by inducing ferroptosis. NCOA3 as a coactivator synergized with NR5A2 to prevent BETi-induced ferroptosis. Mechanistically, NR5A2 synergized with NCOA3 to increase expression of NRF2, a transcription factor that controls the expression of many antioxidant genes.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [11]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug I-BET151 Investigative
Responsed Regulator Bromodomain-containing protein 2 (BRD2) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
MDA-MB-468 cells Breast adenocarcinoma Homo sapiens CVCL_0419
SK-BR-3 cells Breast adenocarcinoma Homo sapiens CVCL_0033
In Vivo Model
Athymic nu/nu mice (5 to 6-week-old female) were acquired from the Silaike Experimental Animal Co. Ltd (Shanghai, China). All mice were randomly allocated to different groups, 1 x 106 MDA-MB-231 cells were subcutaneously inoculated into the right flanks. Ten days later, mice were intraperitoneally injected with PBS including DMSO, JQ1 (50 mg/kg), Bufalin (1 mg/kg) and SR1848 (50 mg/kg), every 2-4 days for 8 times, and the tumor volumes and mice weigh were monitored and recorded every 2-3 days.

    Click to Show/Hide
Response regulation BET (BRD2, BRD3, BRD4, BRDT) inhibitors (BETi) exert an excellent anti-cancer activity in breast cancer. BETi JQ1 and I-BET151 exhibited anti-cancer effects in breast cancer by inducing ferroptosis. NCOA3 as a coactivator synergized with NR5A2 to prevent BETi-induced ferroptosis. Mechanistically, NR5A2 synergized with NCOA3 to increase expression of NRF2, a transcription factor that controls the expression of many antioxidant genes.
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target [11]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug I-BET151 Investigative
Responsed Regulator Bromodomain-containing protein 3 (BRD3) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
MDA-MB-468 cells Breast adenocarcinoma Homo sapiens CVCL_0419
SK-BR-3 cells Breast adenocarcinoma Homo sapiens CVCL_0033
In Vivo Model
Athymic nu/nu mice (5 to 6-week-old female) were acquired from the Silaike Experimental Animal Co. Ltd (Shanghai, China). All mice were randomly allocated to different groups, 1 x 106 MDA-MB-231 cells were subcutaneously inoculated into the right flanks. Ten days later, mice were intraperitoneally injected with PBS including DMSO, JQ1 (50 mg/kg), Bufalin (1 mg/kg) and SR1848 (50 mg/kg), every 2-4 days for 8 times, and the tumor volumes and mice weigh were monitored and recorded every 2-3 days.

    Click to Show/Hide
Response regulation BET (BRD2, BRD3, BRD4, BRDT) inhibitors (BETi) exert an excellent anti-cancer activity in breast cancer. BETi JQ1 and I-BET151 exhibited anti-cancer effects in breast cancer by inducing ferroptosis. NCOA3 as a coactivator synergized with NR5A2 to prevent BETi-induced ferroptosis. Mechanistically, NR5A2 synergized with NCOA3 to increase expression of NRF2, a transcription factor that controls the expression of many antioxidant genes.
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target [11]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug I-BET151 Investigative
Responsed Regulator Bromodomain testis-specific protein (BRDT) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
MDA-MB-468 cells Breast adenocarcinoma Homo sapiens CVCL_0419
SK-BR-3 cells Breast adenocarcinoma Homo sapiens CVCL_0033
In Vivo Model
Athymic nu/nu mice (5 to 6-week-old female) were acquired from the Silaike Experimental Animal Co. Ltd (Shanghai, China). All mice were randomly allocated to different groups, 1 x 106 MDA-MB-231 cells were subcutaneously inoculated into the right flanks. Ten days later, mice were intraperitoneally injected with PBS including DMSO, JQ1 (50 mg/kg), Bufalin (1 mg/kg) and SR1848 (50 mg/kg), every 2-4 days for 8 times, and the tumor volumes and mice weigh were monitored and recorded every 2-3 days.

    Click to Show/Hide
Response regulation BET (BRD2, BRD3, BRD4, BRDT) inhibitors (BETi) exert an excellent anti-cancer activity in breast cancer. BETi JQ1 and I-BET151 exhibited anti-cancer effects in breast cancer by inducing ferroptosis. NCOA3 as a coactivator synergized with NR5A2 to prevent BETi-induced ferroptosis. Mechanistically, NR5A2 synergized with NCOA3 to increase expression of NRF2, a transcription factor that controls the expression of many antioxidant genes.
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target [12]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Isoliquiritin Investigative
Responsed Regulator Transcription factor p65 (RELA) Suppressor
Pathway Response Ferroptosis hsa04216
Apoptosis hsa04210
Gluconeogenesis hsa00010
NF-kappa B signaling pathway hsa04064
Cell Process Cell ferroptosis
Cell apoptosis
Cell proliferation
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
In Vivo Model
BALB/c nude mice (4 weeks old) were obtained from Charles river (Beijing, China). Mice were fed in a temperature-controlled SPF animal room with a 12-h cycle of light-dark. All animal experiments were ratified by the Animal Research Ethics Committee of Shenzhen Bao'an Traditional Chinese Medicine Hospital Group. 10 mice were subcutaneously inoculated with a total of 2106 of MDA-MB-231 cells and then divided into two groups at random, including the Model group and Iso group. Mice in the Iso group were intraperitoneally injected with 25?mg/kg/d Iso, while mice in the Model group were supplied with the same amount of phosphate buffer saline (PBS, Beyotime). Tumor volume was monitored every 4 d. Mice were sacrificed with the intraperitoneal introduction of sodium pentobarbital (100 mg/kg) after tumor cells treatment for 31 d, and the tumor samples were removed and weighed. Tumor volume was determined according to the formula: volume = 1/2 x length x width2.

    Click to Show/Hide
Response regulation Isoliquiritin notably counteracted the LPS-induced relative protein levels of p-p50/p50, p-p65/p65 (RELA), and IB, and the levels of ferroptosis, oxidative stress, glycolysis, and inflammation. Iso inhibited the NF-kB signaling to regulate ferroptosis and improved Dox-resistance in breast cancer.
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target [13]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Metformin Investigative
Responsed Regulator H19 (IncRNA) Suppressor
Pathway Response Glutamate metabolism hsa00250
Autophagy hsa04140
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell autophagy
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
Response regulation Metformin and lncRNA-H19 can regulate both autophagy and ferroptosis. Autophagy inducers and H19 can reverse the production of lipid reactive oxygen species and the inhibition of autophagy induced by metformin. Metformin may induce ferroptosis by inhibiting autophagy via H19, and this discovery may facilitate the development of novel therapies for the treatment of breast cancer.
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target [35]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Bazedoxifene Approved
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HT-1080 cells Fibrosarcoma Homo sapiens CVCL_0317
U2OS cells Osteosarcoma Homo sapiens CVCL_0042
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
HEK-293T cells Normal Homo sapiens CVCL_0063
NCI-H1299 cells Lung large cell carcinoma Homo sapiens CVCL_0060
A-549 cells Lung adenocarcinoma Homo sapiens CVCL_0023
T98G cells Glioblastoma Homo sapiens CVCL_0556
Caki-1 cells Clear cell renal cell carcinoma Homo sapiens CVCL_0234
A-375 cells Amelanotic melanoma Homo sapiens CVCL_0132
4T1 cells Mammary carcinoma Mus musculus CVCL_0125
EO771 cells Breast carcinoma Mus musculus CVCL_GR23
In Vivo Model
N2 Bristol (wild-type) nematodes were maintained on nematode growth media (NGM) plates seeded with bacteria (E. coliOP50) at 20. Experiments with dihomo--linolenic acid (DGLA) were performed using NGM plates formulated with 0.1% Tergitol NP40 (Sigma Chemicals) and 0.125 mM DGLA sodium salt (NuChek Prep, Inc.) or Tergitol alone (vehicle). Dry plates were seeded with OP50 and then three days later ferrostatin-1 or bazedoxifene were dissolved into the plates at a final concentration of 150 uM and allowed to dry for 30 min, before ~50 synchronized L1 larvae were transferred to each plate. Sterility was scored 72-96 h later, as determined by light microscopic examination of uterine embryos.

    Click to Show/Hide
Response regulation The FDA-approved drug bazedoxifene acts as a potent radical trapping antioxidant inhibitor of ferroptosis both in vitro and in vivo. Bazedoxifene potently suppressed ferroptosis in MDA-MB-231, E0771, and 4T1 breast cancer cell lines which do not express the estrogen receptor.
Experiment 10 Reporting the Ferroptosis-centered Disease Response by This Target [36]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Salinomycin Investigative
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model HMLER cells Normal Homo sapiens CVCL_DG85
U2OS cells Osteosarcoma Homo sapiens CVCL_0042
MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
In Vivo Model
MCF-7 cell cultures were collected, enzymatically dissociated, washed with PBS, and re-suspended in a PBS/Matrigel mixture (1:1 v/v). The mixture (0.1 mL) was then implanted in the mammary fat pad of 5-week-old female AthymicNude-Fox1nu mice bilaterally (Harlan, France). Mice received estradiol supplementation (0.4 mg/kg) the same day and 7 days from cell injection, and were observed and palpated for tumor appearance. Mice were treated with AM5 (1 mg/kg body weight/day) by means of intraperitoneal injections every 5 working days of the week.

    Click to Show/Hide
Response regulation A synthetic derivative of salinomycin, which we named ironomycin (AM5), exhibits a more potent and selective activity against breast cancer stem cells (CSCs) in vitro and in vivo, by accumulating and sequestering iron in lysosomes.
Experiment 11 Reporting the Ferroptosis-centered Disease Response by This Target [37]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Siramesine Terminated
Pathway Response Ferroptosis hsa04216
Autophagy hsa04140
Cell Process Cell ferroptosis
Cell autophagy
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
SK-BR-3 cells Breast adenocarcinoma Homo sapiens CVCL_0033
Response regulation The combination of siramesine, a lysosome disruptor, and lapatinib, a dual tyrosine kinase inhibitor, has been shown to synergistically induce cell death in breast cancer cells mediated by ferroptosis. Siramesine and lapatinib initially induced ferroptosis but changes to an autophagy induced cell death after 24 hours.
Experiment 12 Reporting the Ferroptosis-centered Disease Response by This Target [38]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Ferroptocide Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model ES-2 cells Ovarian clear cell adenocarcinoma Homo sapiens CVCL_3509
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
4T1 cells Mammary carcinoma Mus musculus CVCL_0125
A-549 cells Lung adenocarcinoma Homo sapiens CVCL_0023
MCF-10A cells Normal Homo sapiens CVCL_0598
HFF-1 cells Normal Homo sapiens CVCL_3285
Response regulation As ferroptocide induces a regulated, non-apoptotic mode of cell death, this compound (and possibly other pro-ferroptotic agents) have the potential to synergize with the immune system for the treatment of breast cancer.
Experiment 13 Reporting the Ferroptosis-centered Disease Response by This Target [39]
Responsed Disease HER2 positivebreast cancer [ICD-11: 2C60]
Responsed Drug Herceptin Investigative
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model CHO-S/H9C2 cells Normal Cricetulus griseus CVCL_A0TS
Response regulation Herceptin induced injury, oxidative stress, mitochondrial dysfunction and ferroptosis in H9c2 cells, which could be attenuated by Fer-1. These findings provided insights into the pathogenic mechanism that underlie Herceptin-induced cardiomyopathy, which potentially provides a novel therapeutic target for the prevention of cardiotoxicity in HER2 breast cancer treatment.
Experiment 14 Reporting the Ferroptosis-centered Disease Response by This Target [37]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Lapatinib Investigative
Pathway Response Ferroptosis hsa04216
Autophagy hsa04140
Cell Process Cell ferroptosis
Cell autophagy
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
SK-BR-3 cells Breast adenocarcinoma Homo sapiens CVCL_0033
Response regulation The combination of siramesine, a lysosome disruptor, and lapatinib, a dual tyrosine kinase inhibitor, has been shown to synergistically induce cell death in breast cancer cells mediated by ferroptosis. Siramesine and lapatinib initially induced ferroptosis but changes to an autophagy induced cell death after 24 hours.
Experiment 15 Reporting the Ferroptosis-centered Disease Response by This Target [40]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Neratinib Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
Cell metastasis
In Vitro Model 67NR cells Breast carcinoma Mus musculus CVCL_9723
4T1.2 cells Breast carcinoma Mus musculus CVCL_GR32
4T1 cells Mammary carcinoma Mus musculus CVCL_0125
MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
BT-474 cells Invasive breast carcinoma Homo sapiens CVCL_0179
SK-BR-3 cells Breast adenocarcinoma Homo sapiens CVCL_0033
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
In Vivo Model
Female BALB/C mice (5/box) were maintained in a specific pathogen-free environment. For initial characterisation of metastatic spread, TBCP-1 cells (5 x 105) were injected into the left cardiac ventricle of 6-8-week-old female BALB/C mice. The mice were monitored daily and sacrificed after 3 weeks or earlier if signs of metastases became apparent (weight loss > 10%, ruffled fur, lethargy, rapid breathing).

    Click to Show/Hide
Response regulation Neratinib promotes a non-apoptotic form of cell death termed ferroptosis. Importantly, metastasis assays demonstrate that neratinib potently inhibits breast cancer growth and metastasis, including to the brain, and prolongs survival, particularly when used as a neoadjuvant therapy.
Experiment 16 Reporting the Ferroptosis-centered Disease Response by This Target [41]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Piperlongumine Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MIA PaCa-2 cells Pancreatic ductal adenocarcinoma Homo sapiens CVCL_0428
PANC-1 cells Pancreatic ductal adenocarcinoma Homo sapiens CVCL_0480
CFPAC-1 cells Pancreatic ductal adenocarcinoma Homo sapiens CVCL_1119
BxPC-3 cells Pancreatic ductal adenocarcinoma Homo sapiens CVCL_0186
Response regulation Piperlongumine (PL) is a natural product with cytotoxic properties restricted to cancer cells by significantly increasing intracellular reactive oxygen species (ROS) levels. The study demonstrated that PL induced cancer cell death through, at least in part, the induction of ferroptosis. And the triple combined treatment with PL, CNA and SSZ is highly effective against pancreatic cancer.
Experiment 17 Reporting the Ferroptosis-centered Disease Response by This Target [42]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Soyauxinium chloride Investigative
Pathway Response Apoptosis hsa04210
Ferroptosis hsa04216
Necroptosis hsa04217
Cell Process Cell ferroptosis
Cell apoptosis
Cell necroptosis
In Vitro Model U-87MG cells Glioblastoma Homo sapiens CVCL_0022
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Response regulation The prominent cytotoxic potential of soyauxinium chloride (SCHL) on a panel of 18 human and animal cancer cell lines, including MDR phenotypes. This investigated indoloquinazoline alkaloid induced apoptosis in CCRF-CEM cellsviacaspases activation, MMP alteration and increase of ROS production, and caused ferroptosis and necroptosis in breast cancer.
Experiment 18 Reporting the Ferroptosis-centered Disease Response by This Target [31]
Responsed Disease Breast adenocarcinoma [ICD-11: 2C60]
Responsed Drug Vorinostat Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
Response regulation The combined effects of vorinostat with salazosulfapyridine (SASP) depend on the accumulation of ROS caused by a decrease in intracellular GSH levels in breast adenocarcinoma, possibly due to SASP-mediated inhibition of xCT. xCT (coded by the SLC7A11 gene), a light chain subunit of the glutamate-cystine antiporter system Xc(-).
Experiment 19 Reporting the Ferroptosis-centered Disease Response by This Target [43]
Responsed Disease Triple-negative breast cancer [ICD-11: 2C60]
Responsed Regulator Bromodomain-containing protein 4 (BRD4) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MCF-10A cells Normal Homo sapiens CVCL_0598
HEK-293T cells Normal Homo sapiens CVCL_0063
HCC38 cells Breast ductal carcinoma Homo sapiens CVCL_1267
MDA-MB-468 cells Breast adenocarcinoma Homo sapiens CVCL_0419
HCC1143 cells Breast ductal carcinoma Homo sapiens CVCL_1245
HCC1937 cells Breast ductal carcinoma Homo sapiens CVCL_0290
HCC70 cells Breast ductal carcinoma Homo sapiens CVCL_1270
HCC38 cells Breast ductal carcinoma Homo sapiens CVCL_1267
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
SUM159 cells Breast pleomorphic carcinoma Homo sapiens CVCL_5423
Hs-578T cells Invasive breast carcinoma Homo sapiens CVCL_0332
In Vivo Model
For mouse xenograft models, MDA-MB-231 cells (2 x 106 per mouse) expressing green fluorescent protein (GFP) luciferase were implanted bilaterally into the fat pads of the fourth inguinal mammary gland of 6-week-old female athymic nude-Foxn1nu mice. Twenty-three days later, mice were randomized into four groups (n = 10 mice per group) and treated with the following: (i) vehicle; (ii) OTX015, daily through oral gavage (25 mg/kg); OTX015 (100 mg/ml stock in DMSO) was diluted in vehicle solution containing 2% DMSO, 30% polyethylene glycol (PEG)300, and 5% Tween 80; (iii) SB225022, which was intraperitoneally administered 5 days a week, at 5 mg/kg prepared in PBS; or (iv) OTX015 + SB225022 combination.

    Click to Show/Hide
Response regulation BRD4 transcript and protein levels are highly enriched in triple-negative breast cancer tumors and cell lines. Cotargeting of BET (BRD2, BRD3, BRD4, BRDT) and the proteasome applied at low doses could be a promising therapeutic approach for TNBC.
Experiment 20 Reporting the Ferroptosis-centered Disease Response by This Target [44]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator Prominin-2 (PROM2) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MCF-10A cells Normal Homo sapiens CVCL_0598
Hs-578T cells Invasive breast carcinoma Homo sapiens CVCL_0332
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Response regulation Prominin2 (PROM2) facilitates ferroptosis resistance in mammary epithelial and breast cancer cells. Mechanistically, prominin2 promotes the formation of ferritin-containing multivesicular bodies (MVBs) and exosomes that transport iron out of the cell, inhibiting ferroptosis.
Experiment 21 Reporting the Ferroptosis-centered Disease Response by This Target [45]
Responsed Disease Triple-negative breast cancer [ICD-11: 2C60]
Responsed Regulator Pre-mRNA-splicing regulator WTAP (WTAP) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
Cell migration
Cell invasion
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
ZR-75-30 cells Breast carcinoma Homo sapiens CVCL_1661
T-47D cells Invasive breast carcinoma Homo sapiens CVCL_0553
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
MCF-10A cells Normal Homo sapiens CVCL_0598
Response regulation WTAP knockdown promoted ferroptosis to suppress triple-negative breast cancer (TNBC) cell malignant behaviors, which were abrogated by NUPR1 overexpression. WTAP upregulated LCN2 by regulation of NUPR1 m6A modification, thereby suppressing ferroptosis to contribute to accelerate TNBC progression.
Experiment 22 Reporting the Ferroptosis-centered Disease Response by This Target [43]
Responsed Disease Triple-negative breast cancer [ICD-11: 2C60]
Responsed Regulator Bromodomain-containing protein 2 (BRD2) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MCF-10A cells Normal Homo sapiens CVCL_0598
HEK-293T cells Normal Homo sapiens CVCL_0063
HCC38 cells Breast ductal carcinoma Homo sapiens CVCL_1267
MDA-MB-468 cells Breast adenocarcinoma Homo sapiens CVCL_0419
HCC1143 cells Breast ductal carcinoma Homo sapiens CVCL_1245
HCC1937 cells Breast ductal carcinoma Homo sapiens CVCL_0290
HCC70 cells Breast ductal carcinoma Homo sapiens CVCL_1270
HCC38 cells Breast ductal carcinoma Homo sapiens CVCL_1267
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
SUM159 cells Breast pleomorphic carcinoma Homo sapiens CVCL_5423
Hs-578T cells Invasive breast carcinoma Homo sapiens CVCL_0332
In Vivo Model
For mouse xenograft models, MDA-MB-231 cells (2 x 106 per mouse) expressing green fluorescent protein (GFP) luciferase were implanted bilaterally into the fat pads of the fourth inguinal mammary gland of 6-week-old female athymic nude-Foxn1nu mice. Twenty-three days later, mice were randomized into four groups (n = 10 mice per group) and treated with the following: (i) vehicle; (ii) OTX015, daily through oral gavage (25 mg/kg); OTX015 (100 mg/ml stock in DMSO) was diluted in vehicle solution containing 2% DMSO, 30% polyethylene glycol (PEG)300, and 5% Tween 80; (iii) SB225022, which was intraperitoneally administered 5 days a week, at 5 mg/kg prepared in PBS; or (iv) OTX015 + SB225022 combination.

    Click to Show/Hide
Response regulation BRD4 transcript and protein levels are highly enriched in triple-negative breast cancer tumors and cell lines. Cotargeting of BET ( BRD2, BRD3, BRD4, BRDT) and the proteasome applied at low doses could be a promising therapeutic approach for TNBC.
Experiment 23 Reporting the Ferroptosis-centered Disease Response by This Target [43]
Responsed Disease Triple-negative breast cancer [ICD-11: 2C60]
Responsed Regulator Bromodomain-containing protein 3 (BRD3) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MCF-10A cells Normal Homo sapiens CVCL_0598
HEK-293T cells Normal Homo sapiens CVCL_0063
HCC38 cells Breast ductal carcinoma Homo sapiens CVCL_1267
MDA-MB-468 cells Breast adenocarcinoma Homo sapiens CVCL_0419
HCC1143 cells Breast ductal carcinoma Homo sapiens CVCL_1245
HCC1937 cells Breast ductal carcinoma Homo sapiens CVCL_0290
HCC70 cells Breast ductal carcinoma Homo sapiens CVCL_1270
HCC38 cells Breast ductal carcinoma Homo sapiens CVCL_1267
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
SUM159 cells Breast pleomorphic carcinoma Homo sapiens CVCL_5423
Hs-578T cells Invasive breast carcinoma Homo sapiens CVCL_0332
In Vivo Model
For mouse xenograft models, MDA-MB-231 cells (2 x 106 per mouse) expressing green fluorescent protein (GFP) luciferase were implanted bilaterally into the fat pads of the fourth inguinal mammary gland of 6-week-old female athymic nude-Foxn1nu mice. Twenty-three days later, mice were randomized into four groups (n = 10 mice per group) and treated with the following: (i) vehicle; (ii) OTX015, daily through oral gavage (25 mg/kg); OTX015 (100 mg/ml stock in DMSO) was diluted in vehicle solution containing 2% DMSO, 30% polyethylene glycol (PEG)300, and 5% Tween 80; (iii) SB225022, which was intraperitoneally administered 5 days a week, at 5 mg/kg prepared in PBS; or (iv) OTX015 + SB225022 combination.

    Click to Show/Hide
Response regulation BRD4 transcript and protein levels are highly enriched in triple-negative breast cancer tumors and cell lines. Cotargeting of BET (BRD2, BRD3, BRD4, BRDT) and the proteasome applied at low doses could be a promising therapeutic approach for TNBC.
Experiment 24 Reporting the Ferroptosis-centered Disease Response by This Target [43]
Responsed Disease Triple-negative breast cancer [ICD-11: 2C60]
Responsed Regulator Bromodomain testis-specific protein (BRDT) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MCF-10A cells Normal Homo sapiens CVCL_0598
HEK-293T cells Normal Homo sapiens CVCL_0063
HCC38 cells Breast ductal carcinoma Homo sapiens CVCL_1267
MDA-MB-468 cells Breast adenocarcinoma Homo sapiens CVCL_0419
HCC1143 cells Breast ductal carcinoma Homo sapiens CVCL_1245
HCC1937 cells Breast ductal carcinoma Homo sapiens CVCL_0290
HCC70 cells Breast ductal carcinoma Homo sapiens CVCL_1270
HCC38 cells Breast ductal carcinoma Homo sapiens CVCL_1267
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
SUM159 cells Breast pleomorphic carcinoma Homo sapiens CVCL_5423
Hs-578T cells Invasive breast carcinoma Homo sapiens CVCL_0332
In Vivo Model
For mouse xenograft models, MDA-MB-231 cells (2 x 106 per mouse) expressing green fluorescent protein (GFP) luciferase were implanted bilaterally into the fat pads of the fourth inguinal mammary gland of 6-week-old female athymic nude-Foxn1nu mice. Twenty-three days later, mice were randomized into four groups (n = 10 mice per group) and treated with the following: (i) vehicle; (ii) OTX015, daily through oral gavage (25 mg/kg); OTX015 (100 mg/ml stock in DMSO) was diluted in vehicle solution containing 2% DMSO, 30% polyethylene glycol (PEG)300, and 5% Tween 80; (iii) SB225022, which was intraperitoneally administered 5 days a week, at 5 mg/kg prepared in PBS; or (iv) OTX015 + SB225022 combination.

    Click to Show/Hide
Response regulation BRD4 transcript and protein levels are highly enriched in triple-negative breast cancer tumors and cell lines. Cotargeting of BET (BRD2, BRD3, BRD4, B RDT) and the proteasome applied at low doses could be a promising therapeutic approach for TNBC.
Experiment 25 Reporting the Ferroptosis-centered Disease Response by This Target [46]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator Fatty acid-binding protein, adipocyte (FABP4) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
LL/2 (LLC1) cells Lung cancer Mus musculus CVCL_4358
HUVECs (Human umbilical vein endothelial cells)
In Vivo Model
For treatment with RSL3 (20 mg/kg), drug was administered in mice bearing LLC tumors by i.p. injection every other day for 2 weeks. For cisplatin, BALB/c mice bearing 4T1 tumors (50-100 mm3) were administered by i.p. injection of vehicle (0.7% DMSO in PBS) or cisplatin (7 mg/kg/week) for 3 weeks.

    Click to Show/Hide
Response regulation SCD1 and FABP4 were also found upregulated in recurrent human breast cancer samples and correlated with worse prognosis of cancer patients with different types of tumors. Mechanistically, SCD1 leads to fatty acid (FA) desaturation and FABP4 derived from TEM enhances lipid droplet (LD) in cancer cells, which cooperatively protect from oxidative stress-induced ferroptosis.
Experiment 26 Reporting the Ferroptosis-centered Disease Response by This Target [47]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator LINC00665 (IncRNA) Driver
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
Cell invasion
In Vitro Model MCF-10A cells Normal Homo sapiens CVCL_0598
MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Response regulation The LINC00665-miR-410-3p axis was identified as the most potential upstream ncRNA-related pathway of EMC2 in breast cancer. EMC2 levels were significantly positively correlated with tumor immune cell infiltration, immune cell biomarkers, and immune checkpoint expression.
Experiment 27 Reporting the Ferroptosis-centered Disease Response by This Target [48]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator CircRHOT1 (circRNA) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Apoptosis hsa04210
Cell Process Cell ferroptosis
Cell apoptosis
Cell proliferation
Cell migration
Cell invasion
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
T-47D cells Invasive breast carcinoma Homo sapiens CVCL_0553
In Vivo Model
The effect of levobupivacaine on tumor growth of breast cancer in vivo was assessed in the Balb/c nude mice (male, 4-week-old) (n = 5). About 1 x 107 cells MDA-MB-231 cells transfected with control shRNA or circRHOT1 shRNA were subcutaneously injected into the mice. After 5 days of injection, we measured tumor growth every 5 days. We sacrificed the mice after 30 days of injection, and tumors were scaled.

    Click to Show/Hide
Response regulation The depletion of circRHOT1 was able to repress the proliferation and induce the apoptosis of breast cancer cells. CircRHOT1 knockdown could remarkably inhibit the invasion and migration in the breast cancer cells. Mechanically, circRHOT1 contributed to malignant progression and attenuated ferroptosis in breast cancer by the miR-106a-5p/STAT3 axis.
Experiment 28 Reporting the Ferroptosis-centered Disease Response by This Target [48]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator Signal transducer and activator of transcription 3 (STAT3) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Apoptosis hsa04210
Cell Process Cell ferroptosis
Cell apoptosis
Cell proliferation
Cell migration
Cell invasion
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
T-47D cells Invasive breast carcinoma Homo sapiens CVCL_0553
In Vivo Model
The effect of levobupivacaine on tumor growth of breast cancer in vivo was assessed in the Balb/c nude mice (male, 4-week-old) (n = 5). About 1 x 107 cells MDA-MB-231 cells transfected with control shRNA or circRHOT1 shRNA were subcutaneously injected into the mice. After 5 days of injection, we measured tumor growth every 5 days. We sacrificed the mice after 30 days of injection, and tumors were scaled.

    Click to Show/Hide
Response regulation The depletion of circRHOT1 was able to repress the proliferation and induce the apoptosis of breast cancer cells. CircRHOT1 knockdown could remarkably inhibit the invasion and migration in the breast cancer cells. Mechanically, circRHOT1 contributed to malignant progression and attenuated ferroptosis in breast cancer by the miR-106a-5p/ STAT3 axis.
Experiment 29 Reporting the Ferroptosis-centered Disease Response by This Target [48]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator hsa-miR-106a-5p (miRNA) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Apoptosis hsa04210
Cell Process Cell ferroptosis
Cell apoptosis
Cell proliferation
Cell migration
Cell invasion
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
T-47D cells Invasive breast carcinoma Homo sapiens CVCL_0553
In Vivo Model
The effect of levobupivacaine on tumor growth of breast cancer in vivo was assessed in the Balb/c nude mice (male, 4-week-old) (n = 5). About 1 x 107 cells MDA-MB-231 cells transfected with control shRNA or circRHOT1 shRNA were subcutaneously injected into the mice. After 5 days of injection, we measured tumor growth every 5 days. We sacrificed the mice after 30 days of injection, and tumors were scaled.

    Click to Show/Hide
Response regulation The depletion of circRHOT1 was able to repress the proliferation and induce the apoptosis of breast cancer cells. CircRHOT1 knockdown could remarkably inhibit the invasion and migration in the breast cancer cells. Mechanically, circRHOT1 contributed to malignant progression and attenuated ferroptosis in breast cancer by the miR-106a-5p/STAT3 axis.
Experiment 30 Reporting the Ferroptosis-centered Disease Response by This Target [47]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator hsa-miR-410-3p (miRNA) Suppressor
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
Cell invasion
In Vitro Model MCF-10A cells Normal Homo sapiens CVCL_0598
MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Response regulation The LINC00665- miR-410-3p axis was identified as the most potential upstream ncRNA-related pathway of EMC2 in breast cancer. EMC2 levels were significantly positively correlated with tumor immune cell infiltration, immune cell biomarkers, and immune checkpoint expression.
Experiment 31 Reporting the Ferroptosis-centered Disease Response by This Target [47]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator ER membrane protein complex subunit 2 (EMC2) Driver
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
Cell invasion
In Vitro Model MCF-10A cells Normal Homo sapiens CVCL_0598
MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Response regulation The LINC00665-miR-410-3p axis was identified as the most potential upstream ncRNA-related pathway of EMC2 in breast cancer. EMC2 levels were significantly positively correlated with tumor immune cell infiltration, immune cell biomarkers, and immune checkpoint expression.
Experiment 32 Reporting the Ferroptosis-centered Disease Response by This Target [45]
Responsed Disease Triple-negative breast cancer [ICD-11: 2C60]
Responsed Regulator Nuclear protein 1 (NUPR1) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
Cell migration
Cell invasion
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
ZR-75-30 cells Breast carcinoma Homo sapiens CVCL_1661
T-47D cells Invasive breast carcinoma Homo sapiens CVCL_0553
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
MCF-10A cells Normal Homo sapiens CVCL_0598
Response regulation WTAP knockdown promoted ferroptosis to suppress triple-negative breast cancer (TNBC) cell malignant behaviors, which were abrogated by NUPR1 overexpression. WTAP upregulated LCN2 by regulation of NUPR1 m6A modification, thereby suppressing ferroptosis to contribute to accelerate TNBC progression.
Experiment 33 Reporting the Ferroptosis-centered Disease Response by This Target [45]
Responsed Disease Triple-negative breast cancer [ICD-11: 2C60]
Responsed Regulator Neutrophil gelatinase-associated lipocalin (LCN2) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
Cell migration
Cell invasion
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
ZR-75-30 cells Breast carcinoma Homo sapiens CVCL_1661
T-47D cells Invasive breast carcinoma Homo sapiens CVCL_0553
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
MCF-10A cells Normal Homo sapiens CVCL_0598
Response regulation WTAP knockdown promoted ferroptosis to suppress triple-negative breast cancer (TNBC) cell malignant behaviors, which were abrogated by NUPR1 overexpression. WTAP upregulated LCN2 by regulation of NUPR1 m6A modification, thereby suppressing ferroptosis to contribute to accelerate TNBC progression.
Experiment 34 Reporting the Ferroptosis-centered Disease Response by This Target [49]
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator Zinc transporter SLC39A7 (SLC39A7) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
HT-1080 cells Fibrosarcoma Homo sapiens CVCL_0317
RCC4 cells Clear cell renal cell carcinoma Homo sapiens CVCL_0498
Response regulation Zinc is also essential for ferroptosis in breast cancer and renal cancer cells. The study identified SLC39A7, encoding ZIP7 that controls zinc transport from endoplasmic reticulum (ER) to cytosol, as a novel genetic determinant of ferroptosis.
Transferrin receptor protein 1 (TFRC)
In total 3 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [14]
Target for Ferroptosis Marker/Suppressor/Driver
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Sulfasalazine Investigative
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
T-47D cells Invasive breast carcinoma Homo sapiens CVCL_0553
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
Response regulation Sulfasalazine (SAS) upregulated TFRC and DMT1. Knockdown of the ER increased TFRC expression in breast cancer cells. In conclusion SAS could trigger ferroptosis in breast cancer cells, especially in cells with low ER expression.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [15]
Target for Ferroptosis Marker/Suppressor/Driver
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator Plasmanylethanolamine desaturase (PEDS1) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Autophagy hsa04140
Cell Process Cell ferroptosis
Cell autophagy
Cell proliferation
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
In Vivo Model
Twenty BALB/c nude female mice (4- to 6-week-old) were purchased from Charles River Laboratories (Beijing, China) to establish tumorigenesis (5 mice in each). We injected MCF-7 or MDA-MB-231 cells (4 x 106) transfected with the stable knockdown and over-expressing TMEM189 into the flanks of mice to construct tumorigenesis models, respectively. Meanwhile, the sh-Con and empty vector were served as the control groups for each model.

    Click to Show/Hide
Response regulation TMEM189 (PEDS1) could inhibit autophagy to mediate ferroptosis in breast cancer cells. Moreover, TMEM189 ablation strongly up-regulated LC3BII and transferrin receptor 1 (TfR1) expression levels in breast cancer cells, whereas down-regulated p62 and GPX4.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [16]
Target for Ferroptosis Marker/Suppressor/Driver
Responsed Disease Breast adenocarcinoma [ICD-11: 2C60]
Responsed Regulator YY1-associated protein 1 (YY1AP1) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Hippo signaling pathway hsa04390
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model mEFs (Mouse embryonic fibroblasts)
NF639 (Mouse breast epithelial cells)
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
BT-474 cells Invasive breast carcinoma Homo sapiens CVCL_0179
H1650-ER1 cells Minimally invasive lung adenocarcinoma Homo sapiens CVCL_4V01
In Vivo Model
Six- to eight-week-old female athymic nu/nu mice were purchased from Envigo (East Millstone, NJ, USA). For s.c. tumour models, mice were injected in the right flank with 1 x 107 shNT-GPX4 iKO MSTO-211H cells or shMerlin-GPX4 iKO MSTO-211H cells suspended in 150 uL Matrigel. Tumours were measured with callipers every 3 days. When tumours reached a mean volume of 100 mm3, mice with similarly sized tumours were grouped into four treatment groups. For control or knockout cohorts, mice were given intraperitoneal (i.p.) injections of 0.9% sterile saline or Doxycycline (100 mg/kg body weight) for two days. At the same time, mice were provided with either a normal diet or Doxycycline diet for control or knockout cohorts, respectively.

    Click to Show/Hide
Response regulation In epithelial cells, such interactions mediated by E-cadherin suppress ferroptosis by activating the intracellular NF2 (also known as merlin) and Hippo signalling pathway in Breast adenocarcinoma. Antagonizing this signalling axis allows the proto-oncogenic transcriptional co-activator YAP to promote ferroptosis by upregulating several ferroptosis modulators, including ACSL4 and TFRC.
Solute carrier family 40 member 1 (SLC40A1)
In total 5 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [17]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Siramesine Terminated
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
SK-BR-3 cells Breast adenocarcinoma Homo sapiens CVCL_0033
MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
ZR-75-1 cells Invasive breast carcinoma Homo sapiens CVCL_0588
Response regulation Overexpression FPN resulted in decreased ROS and cell death whereas knockdown of FPN increased cell death after siramesine and lapatinib treatment. This indicates a novel induction of ferroptosis through altered iron regulation by treating breast cancer cells with a lysosome disruptor and a tyrosine kinase inhibitor.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [10]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Etoposide Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
MCF-10A cells Normal Homo sapiens CVCL_0598
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Response regulation The combined treatment of etoposide and erastin synergistically induced oxidative stress and lipid peroxidation, while suppressing glutathione peroxidase activity in breast cancer cells. More importantly, the combination treatment synergistically increased iron accumulation, which was associated with altered expression of IREB2/FPN1. Additionally, ferroptosis-regulating proteins ACSF2 and GPX4 were altered more potently by the combination treatment, compared to untreated cells and erastin treatment alone (p<0.05).
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [17]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Lapatinib Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
SK-BR-3 cells Breast adenocarcinoma Homo sapiens CVCL_0033
MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
ZR-75-1 cells Invasive breast carcinoma Homo sapiens CVCL_0588
Response regulation Overexpression FPN resulted in decreased ROS and cell death whereas knockdown of FPN increased cell death after siramesine and lapatinib treatment. This indicates a novel induction of ferroptosis through altered iron regulation by treating breast cancer cells with a lysosome disruptor and a tyrosine kinase inhibitor.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [18]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Breast adenocarcinoma [ICD-11: 2C60]
Responsed Regulator Metal regulatory transcription factor 1 (MTF1) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
RCC4 cells Clear cell renal cell carcinoma Homo sapiens CVCL_0498
HEK-293T cells Normal Homo sapiens CVCL_0063
HT-1080 cells Fibrosarcoma Homo sapiens CVCL_0317
Response regulation ATM inhibition enhanced the nuclear translocation of metal-regulatory transcription factor 1 (MTF1), responsible for regulating expression of Ferritin/FPN1 and ferroptosis protection in breast adenocarcinoma.
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target [18]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Breast adenocarcinoma [ICD-11: 2C60]
Responsed Regulator Serine-protein kinase ATM (ATM) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
RCC4 cells Clear cell renal cell carcinoma Homo sapiens CVCL_0498
HEK-293T cells Normal Homo sapiens CVCL_0063
HT-1080 cells Fibrosarcoma Homo sapiens CVCL_0317
Response regulation ATM inhibition enhanced the nuclear translocation of metal-regulatory transcription factor 1 (MTF1), responsible for regulating expression of Ferritin/FPN1 and ferroptosis protection in breast adenocarcinoma.
Nuclear factor erythroid 2-related factor 2 (NFE2L2)
In total 3 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [22]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Breast adenocarcinoma [ICD-11: 2C60]
Responsed Regulator Cyclic AMP-dependent transcription factor ATF-2 (ATF2) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
HeLa cells Endocervical adenocarcinoma Homo sapiens CVCL_0030
A-549 cells Lung adenocarcinoma Homo sapiens CVCL_0023
In Vivo Model
Athymic nu/nu mice (5 to 6-week-old female) were acquired from the Silaike Experimental Animal Co. Ltd (Shanghai, China). Mice was randomly divided into different groups (n = 6 per group), 1 x 106 MB-231 cells with stable overexpression of ATF2 and control cells were subcutaneously inoculated on the right flanks. Seven days later, mice were injected intraperitoneally with PBS containingdimethyl sulfoxide(DMSO) and JQ1 (50 mg/kg) once every 2-4 days for 8 times, and the tumor volumes and body weight of mice were monitored and recorded every 23 days.

    Click to Show/Hide
Response regulation ATF2 inhibited BETi-induced ferroptosis by increasing NRF2 expression. Altogether, ATF2 suppressed ani-tumor effects of BETi in a negative feedback manner by attenuating ferroptosis. BETi combined with ATF2 or NRF2 inhibitor might be a novel strategy for treatment of Breast cancer.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [23]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Breast ductal carcinoma [ICD-11: 2C60]
Responsed Regulator Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN (PTEN) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Ubiquitin mediated proteolysis hsa04120
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HCC1419 cells Breast ductal carcinoma Homo sapiens CVCL_1251
HCC1395 cells Breast ductal carcinoma Homo sapiens CVCL_1249
NCI-H226 cells Pleural epithelioid mesothelioma Homo sapiens CVCL_1544
NCI-H446 cells Lung small cell carcinoma Homo sapiens CVCL_1562
ZR-75-1 cells Invasive breast carcinoma Homo sapiens CVCL_0588
HCC1937 cells Breast ductal carcinoma Homo sapiens CVCL_0290
HCC1187 cells Breast ductal carcinoma Homo sapiens CVCL_1247
HCC1806 cells Breast squamous cell carcinoma Homo sapiens CVCL_1258
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
NCI-H520 cells Lung squamous cell carcinoma Homo sapiens CVCL_1566
mEFs (Mouse embryonic fibroblasts)
In Vivo Model
A cross between 12 week old, B6.129S4 Ptenfl/fl mice obtained from Jackson Laboratory was set up and 14 days later both male and female embryos were harvested from the pregnant females. Highly vascularized sections of the embryos were removed - head, extremities, and liver. The remainder was minced via a scalpel and resuspended in 0.25% trypsin using a pipet. The cells were then incubated for 10 min in an incubator at 37 with 5% CO2 before being further resuspended into single cell suspension. The cells were then spun down at 1500 RPM for 5 min to remove the trypsin, after which they were resuspended in 10 mL of fresh media and transferred to a 10 cm dish.

    Click to Show/Hide
Response regulation Loss of PTEN activates AKT kinase to inhibit GSK3, increasing NF-E2 p45-related factor 2 (NRF2) along with transcription of one of its known target genes encoding xCT in breast cancers.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [24]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Mammary carcinoma [ICD-11: 2C60]
Responsed Regulator Tyrosine-protein kinase receptor TYRO3 (TYRO3) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
PI3K-Akt signaling pathway hsa04151
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model 4T1 cells Mammary carcinoma Mus musculus CVCL_0125
HEK-293T cells Normal Homo sapiens CVCL_0063
B16-F10 cells Melanoma Mus musculus CVCL_0159
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
THP-1 cells Childhood acute monocytic leukemia Homo sapiens CVCL_0006
Py8119 cells Breast carcinoma Mus musculus CVCL_AQ09
In Vivo Model
Six-week-old BALB/c mice were purchased from The Jackson Laboratory. Mouse 4T1 cells (5 x 104 cells) in 50 uL of 50% Matrigel (47743-720, Corning) were injected into the mammary fat pad. Three days after inoculation, 100 ug mouse anti-PD-1 antibody (BE0146, Bio X Cell) or IgG control (BE0089, Bio X Cell) was injected intraperitoneally twice a week for a total of 5 injections. For the TYRO3 inhibitor and antimPD-1 combination treatment, mice were also treated daily with vehicle control (90% polyethylene glycol 400 and 10% DMSO) or LDC1267 (20 mg/kg, S7638, Selleck Chemical) for 10 days by intraperitoneal injection.

    Click to Show/Hide
Response regulation The study proposed a model in which high expression of TYRO3 or TYRO3 activation by its ligands on apoptotic cells triggers the downstream AKT/NRF2 pathway, followed by the transcription of genes that inhibit ferroptosis in Mammary carcinoma.
Natural resistance-associated macrophage protein 2 (SLC11A2)
In total 1 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [14]
Target for Ferroptosis Driver
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Sulfasalazine Investigative
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
T-47D cells Invasive breast carcinoma Homo sapiens CVCL_0553
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
Response regulation Sulfasalazine (SAS) upregulated TFRC and DMT1. Knockdown of the ER increased TFRC expression in breast cancer cells. In conclusion SAS could trigger ferroptosis in breast cancer cells, especially in cells with low ER expression.
Long-chain-fatty-acid--CoA ligase 4 (ACSL4)
In total 4 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [8]
Target for Ferroptosis Driver
Responsed Disease Triple-negative breast cancer [ICD-11: 2C60]
Responsed Drug Polyphyllin III Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Hs-578T cells Invasive breast carcinoma Homo sapiens CVCL_0332
MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
T-47D cells Invasive breast carcinoma Homo sapiens CVCL_0553
HBL-100 cells Normal Homo sapiens CVCL_4362
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
MDA-MB-453 cells Breast adenocarcinoma Homo sapiens CVCL_0418
In Vivo Model
MDA-MB-231 xenografts were established in 5 week-old BALB/C nude mice (Shanghai SLAC Laboratory Animal Corporation) by inoculating 1 x 106 cells mixed with Matrigel (BD Biosciences) at a 1:1 ratio into the abdominal mammary fat pad. When the tumor reached 50-100 mm3, the mice were assigned randomly into different treatment groups (DMSO, PPIII, SAS, and PPIII + SAS groups), and each group consisted of 5 mice. PPIII (5 mg/kg/day) and SAS (200 mg/kg/day) were dissolved in dimethyl sulfoxide (DMSO), diluted in PBS, and then intraperitoneally injected into mice at a dose of 10 ml/kg/d once a day.

    Click to Show/Hide
Response regulation Polyphyllin III, which is a major saponin extracted fromParis polyphyllarhizomes, exerted its proliferation-inhibitory effect on MDA-MB-231 triple-negative breast cancer cells mainly through ACSL4-mediated lipid peroxidation elevation and ferroptosis induction. Polyphyllin III treatment also induced KLF4-mediated protective upregulation of xCT(SLC7A11), which is the negative regulator of ferroptosis.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [25]
Target for Ferroptosis Driver
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Robustaflavone 7,5'-dimethyl ether Investigative
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
Response regulation Molecular docking study indicated that robustaflavone 7,5'-dimethyl ether (8) is a potential strong inhibitor, which induce ferroptosis via down-regulating the expression level of ACSL4 proteins in human breast cancer MCF-7 cells.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [16]
Target for Ferroptosis Driver
Responsed Disease Breast adenocarcinoma [ICD-11: 2C60]
Responsed Regulator YY1-associated protein 1 (YY1AP1) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Hippo signaling pathway hsa04390
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model mEFs (Mouse embryonic fibroblasts)
NF639 (Mouse breast epithelial cells)
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
BT-474 cells Invasive breast carcinoma Homo sapiens CVCL_0179
H1650-ER1 cells Minimally invasive lung adenocarcinoma Homo sapiens CVCL_4V01
In Vivo Model
Six- to eight-week-old female athymic nu/nu mice were purchased from Envigo (East Millstone, NJ, USA). For s.c. tumour models, mice were injected in the right flank with 1 x 107 shNT-GPX4 iKO MSTO-211H cells or shMerlin-GPX4 iKO MSTO-211H cells suspended in 150 uL Matrigel. Tumours were measured with callipers every 3 days. When tumours reached a mean volume of 100 mm3, mice with similarly sized tumours were grouped into four treatment groups. For control or knockout cohorts, mice were given intraperitoneal (i.p.) injections of 0.9% sterile saline or Doxycycline (100 mg/kg body weight) for two days. At the same time, mice were provided with either a normal diet or Doxycycline diet for control or knockout cohorts, respectively.

    Click to Show/Hide
Response regulation In epithelial cells, such interactions mediated by E-cadherin suppress ferroptosis by activating the intracellular NF2 (also known as merlin) and Hippo signalling pathway in breast adenocarcinoma. Antagonizing this signalling axis allows the proto-oncogenic transcriptional co-activator YAP to promote ferroptosis by upregulating several ferroptosis modulators, including ACSL4 and TFRC.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [26]
Target for Ferroptosis Driver
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator hsa-miR-5096 (miRNA) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MDA-MB-468 cells Breast adenocarcinoma Homo sapiens CVCL_0419
MDA-MB-453 cells Breast adenocarcinoma Homo sapiens CVCL_0418
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
SK-BR-3 cells Breast adenocarcinoma Homo sapiens CVCL_0033
T-47D cells Invasive breast carcinoma Homo sapiens CVCL_0553
MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
ZR-75-1 cells Invasive breast carcinoma Homo sapiens CVCL_0588
MCF-10A cells Normal Homo sapiens CVCL_0598
In Vivo Model
Mating was setup 2 days prior to injection day and zebrafish embryos were collected and incubated in E3 embryo medium (5 mM NaCl, 0.17 mM KCl, 10 mM HEPES, 0.33 mM MgSO4·7H2O, 0.33 mM CaCl2·6H2O, and 0.00001% methylene blue) containing 0.2 mM N-phenyl-thiourea (PTU) (catalog no: P7629, Sigma-Aldrich). Two days post-fertilization, the chorion was removed manually using fine forceps, and the embryos were anesthetized using E3 medium containing 200 mg/L Ethyl 3-aminobenzoate methanesulfonate (Tricaine) (catalog no: A5040, Sigma-Aldrich). Anesthetized embryos were mounted in 0.7% low melting agarose containing 200 ug/ml of Tricaine and were microinjected with 500 cells in the yolk sac using Nanoject III (catalog no: 3-000-207; Drummond Scientific Company, PA, USA). At 1 day post-injection (dpi), embryos with similar graft size were selected and imaged using both bright field and RFP channels and incubated in E3-PTU medium containing 5 ug/ml doxycycline at 34 until further imaging. At 4 dpi, embryos were anesthetized and imaged again using both bright field and RFP channels using Olympus IX-73 microscope. Cells that migrated outside the yolk sac (injection site) were represented by a notable fluorescent dot and considered a metastatic event; these were counted manually for all embryos in each experimental group.

    Click to Show/Hide
Response regulation The present study demonstrated that miR-5096 targets and downregulates SLC7A11, thereby providing a mechanistic basis for ferroptosis in human breast cancer cells. In addition, miR-5096 induced cell death via ferroptosis, characterized by mitochondrial shrinkage with partial loss of cristae with simultaneous changes in ACSL4, ROS, lipid ROS, OH-, reactive iron, GSH, and MMP levels.
Long-chain-fatty-acid--CoA ligase 1 (ACSL1)
In total 1 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [27]
Target for Ferroptosis Driver
Responsed Disease Breast adenocarcinoma [ICD-11: 2C60]
Responsed Drug alpha-Eleostearic acid Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MDA-MB-468 cells Breast adenocarcinoma Homo sapiens CVCL_0419
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
BT-20 cells Invasive breast carcinoma of no special type Homo sapiens CVCL_0178
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
HCC38 cells Breast ductal carcinoma Homo sapiens CVCL_1267
HCC1806 cells Breast squamous cell carcinoma Homo sapiens CVCL_1258
HCC1187 cells Breast ductal carcinoma Homo sapiens CVCL_1247
HCC1143 cells Breast ductal carcinoma Homo sapiens CVCL_1245
HCC70 cells Breast ductal carcinoma Homo sapiens CVCL_1270
Hs-578T cells Invasive breast carcinoma Homo sapiens CVCL_0332
MCF-10A cells Normal Homo sapiens CVCL_0598
MCF-12A cells Normal Homo sapiens CVCL_3744
In Vivo Model
Mice were housed in a dedicated laboratory animal facility with 12-h light:dark cycle, at 70F+/-2 degrees, and 40-70% relative humidity. Orthotopic xenografts were generated by implanting 2.5 million MDA-MB-231 cells in 100 uL phosphate-buffered saline (PBS) mixed with 100 uL growth factor-reduced Matrigel (Corning) bilaterally into the fourth inguinal fat pad of four- to six-week-old female NOD.

    Click to Show/Hide
Response regulation a-eleostearic acid (ESA)-triggered ferroptosis is mediated by acyl-CoA synthetase long-chain isoform 1 (ACSL1), which promotes ESA incorporation into neutral lipids including triacylglycerols in Breast adenocarcinoma.
Iron-responsive element-binding protein 2 (IREB2)
In total 1 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [10]
Target for Ferroptosis Driver
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Drug Etoposide Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MCF-7 cells Breast carcinoma Homo sapiens CVCL_0031
MCF-10A cells Normal Homo sapiens CVCL_0598
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
Response regulation The combined treatment of etoposide and erastin synergistically induced oxidative stress and lipid peroxidation, while suppressing glutathione peroxidase activity in breast cancer cells. More importantly, the combination treatment synergistically increased iron accumulation, which was associated with altered expression of IREB2/FPN1. Additionally, ferroptosis-regulating proteins ACSF2 and GPX4 were altered more potently by the combination treatment, compared to untreated cells and erastin treatment alone (p<0.05).
Heme oxygenase 1 (HMOX1)
In total 1 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [28]
Target for Ferroptosis Driver/Suppressor
Responsed Disease Triple-negative breast cancer [ICD-11: 2C60]
Responsed Drug Shuganning injection Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model SK-BR-3 cells Breast adenocarcinoma Homo sapiens CVCL_0033
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
MDA-MB-468 cells Breast adenocarcinoma Homo sapiens CVCL_0419
BT-549 cells Invasive breast carcinoma Homo sapiens CVCL_1092
MCF-10A cells Normal Homo sapiens CVCL_0598
786-O cells Renal cell carcinoma Homo sapiens CVCL_1051
A2780 cells Ovarian endometrioid adenocarcinoma Homo sapiens CVCL_0134
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
Hep-G2 cells Hepatoblastoma Homo sapiens CVCL_0027
A-549 cells Lung adenocarcinoma Homo sapiens CVCL_0023
L-02 cells Endocervical adenocarcinoma Homo sapiens CVCL_6926
WPMY-1 cells Normal Homo sapiens CVCL_3814
U-87MG cells Glioblastoma Homo sapiens CVCL_0022
HT-1080 cells Fibrosarcoma Homo sapiens CVCL_0317
In Vivo Model
MDA-MB-231 cells resuspended in PBS (2 x 106/100 m1) were injected subcutaneously into bothhind limbsof 4-6 weeks old femalenude mice. A week later, SGNI was administrated byintraperitoneal injectionat a dose of 112.5 mg/kg/3 d.

    Click to Show/Hide
Response regulation Shuganning injection (SGNI) induced a ferroptotic cell death of Triple-negative breast cancer (TNBC) cells. Mechanistically, SGNI induced ferroptosis was dependent on HO-1, which promotes intracellular labile iron pool accumulation, and was alleviated by HO-1 knockdown and inhibition by tin protoporphyrin IX.
Ferroptosis suppressor protein 1 (AIFM2)
In total 4 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [29]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator CircGFRA1 (circRNA) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
Cell infiltration
Cell migration
In Vitro Model SK-BR-3 cells Breast adenocarcinoma Homo sapiens CVCL_0033
BT-474 cells Invasive breast carcinoma Homo sapiens CVCL_0179
In Vivo Model
Female BALB/c nude mice aged 4 weeks were administered subcutaneously with 2 x 106 cells (five mice/group). After that, the mice were given intratumoural inoculation of 40 uL si-circGFRA1 or si-NC every 4 days. To detect lung metastases, we intravenously inoculated 1 x 105 cells into the tail veins of mice (six mice/group). After the elapse of 8 weeks, we anaesthetized the mice, harvested their lungs and visually counted the metastatic nodules in the lungs, followed by validation via H&E staining and counting under a microscope.

    Click to Show/Hide
Response regulation Knockdown of circGFRA1 could attenuate HER-2-positive HER-2-positive breast cancer progression by inhibiting the proliferation, infiltration and migratory ability of HER-2-positive BC cells. Through ceRNA mechanism, circGFRA1 could bind to miR-1228 and alleviate inhibitory activity of miR-1228 on targeted gene AIFM2.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [29]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Responsed Regulator hsa-miR-1228-3p (miRNA) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
Cell infiltration
Cell migration
In Vitro Model SK-BR-3 cells Breast adenocarcinoma Homo sapiens CVCL_0033
BT-474 cells Invasive breast carcinoma Homo sapiens CVCL_0179
In Vivo Model
Female BALB/c nude mice aged 4 weeks were administered subcutaneously with 2 x 106 cells (five mice/group). After that, the mice were given intratumoural inoculation of 40 uL si-circGFRA1 or si-NC every 4 days. To detect lung metastases, we intravenously inoculated 1 x 105 cells into the tail veins of mice (six mice/group). After the elapse of 8 weeks, we anaesthetized the mice, harvested their lungs and visually counted the metastatic nodules in the lungs, followed by validation via H&E staining and counting under a microscope.

    Click to Show/Hide
Response regulation Knockdown of circGFRA1 could attenuate HER-2-positive HER-2-positive breast cancer progression by inhibiting the proliferation, infiltration and migratory ability of HER-2-positive BC cells. Through ceRNA mechanism, circGFRA1 could bind to miR-1228 and alleviate inhibitory activity of miR-1228 on targeted gene AIFM2.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [30]
Target for Ferroptosis Suppressor
Responsed Disease Triple-negative breast cancer [ICD-11: 2C60]
Responsed Regulator Merlin (NF2) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model hMPs (Human macrophages)
In Vivo Model
C57BL/6 mice (female, 6-8 weeks old, 20-30 g weight) and SPF-grade SD rats (female, 180-230 g weight) were used to detect the toxicity of nanoparticles. Different cells (5 x 106) cells were grafted in the left flank; 5 days after engraftation, the stimulated TAMs (1 x 106) were injected into NSG mice through the tail vein. Different treatments were given and recorded as day 0.

    Click to Show/Hide
Response regulation The NF2-YAP signaling axis modulated the expression of ferroptosis suppressor protein 1 (FSP1) and CD24 in CD24 high cells. This system achieved dual antitumor effects, ultimately promoting cell death and thus inhibiting triple-negative breast cancer (TNBC) tumor growth, with some tumors even disappearing.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [30]
Target for Ferroptosis Suppressor
Responsed Disease Triple-negative breast cancer [ICD-11: 2C60]
Responsed Regulator Transcriptional coactivator YAP1 (YAP1) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model hMPs (Human macrophages)
In Vivo Model
C57BL/6 mice (female, 6-8 weeks old, 20-30 g weight) and SPF-grade SD rats (female, 180-230 g weight) were used to detect the toxicity of nanoparticles. Different cells (5 x 106) cells were grafted in the left flank; 5 days after engraftation, the stimulated TAMs (1 x 106) were injected into NSG mice through the tail vein. Different treatments were given and recorded as day 0.

    Click to Show/Hide
Response regulation The NF2- YAP signaling axis modulated the expression of ferroptosis suppressor protein 1 (FSP1) and CD24 in CD24 high cells. This system achieved dual antitumor effects, ultimately promoting cell death and thus inhibiting triple-negative breast cancer (TNBC) tumor growth, with some tumors even disappearing.
Stearoyl-CoA desaturase (SCD)
In total 1 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [46]
Target for Ferroptosis Suppressor
Responsed Disease Breast cancer [ICD-11: 2C60-2C65]
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
LL/2 (LLC1) cells Lung cancer Mus musculus CVCL_4358
HUVECs (Human umbilical vein endothelial cells)
In Vivo Model
For treatment with RSL3 (20 mg/kg), drug was administered in mice bearing LLC tumors by i.p. injection every other day for 2 weeks. For cisplatin, BALB/c mice bearing 4T1 tumors (50-100 mm3) were administered by i.p. injection of vehicle (0.7% DMSO in PBS) or cisplatin (7 mg/kg/week) for 3 weeks.

    Click to Show/Hide
Response regulation SCD1 and FABP4 were also found upregulated in recurrent human breast cancer samples and correlated with worse prognosis of cancer patients with different types of tumors. Mechanistically, SCD1 leads to fatty acid (FA) desaturation and FABP4 derived from TEM enhances lipid droplet (LD) in cancer cells, which cooperatively protect from oxidative stress-induced ferroptosis.
References
Ref 1 Identification of a new natural biflavonoids against breast cancer cells induced ferroptosis via the mitochondrial pathway. Bioorg Chem. 2021 Apr;109:104744. doi: 10.1016/j.bioorg.2021.104744. Epub 2021 Feb 17.
Ref 2 Ferritinophagy is required for the induction of ferroptosis by the bromodomain protein BRD4 inhibitor (+)-JQ1 in cancer cells. Cell Death Dis. 2019 Apr 15;10(5):331. doi: 10.1038/s41419-019-1564-7.
Ref 3 Ketamine suppresses proliferation and induces ferroptosis and apoptosis of breast cancer cells by targeting KAT5/GPX4 axis. Biochem Biophys Res Commun. 2021 Dec 31;585:111-116. doi: 10.1016/j.bbrc.2021.11.029. Epub 2021 Nov 12.
Ref 4 Metformin induces ferroptosis by targeting miR-324-3p/GPX4 axis in breast cancer. Acta Biochim Biophys Sin (Shanghai). 2021 Mar 2;53(3):333-341. doi: 10.1093/abbs/gmaa180.
Ref 5 Simvastatin induced ferroptosis for triple-negative breast cancer therapy. J Nanobiotechnology. 2021 Oct 9;19(1):311. doi: 10.1186/s12951-021-01058-1.
Ref 6 Lidocaine Promoted Ferroptosis by Targeting miR-382-5p /SLC7A11 Axis in Ovarian and Breast Cancer. Front Pharmacol. 2021 May 26;12:681223. doi: 10.3389/fphar.2021.681223. eCollection 2021.
Ref 7 Metformin induces Ferroptosis by inhibiting UFMylation of SLC7A11 in breast cancer. J Exp Clin Cancer Res. 2021 Jun 23;40(1):206. doi: 10.1186/s13046-021-02012-7.
Ref 8 Polyphyllin -Induced Ferroptosis in MDA-MB-231 Triple-Negative Breast Cancer Cells can Be Protected Against by KLF4-Mediated Upregulation of xCT. Front Pharmacol. 2021 May 10;12:670224. doi: 10.3389/fphar.2021.670224. eCollection 2021.
Ref 9 DDR2 upregulation confers ferroptosis susceptibility of recurrent breast tumors through the Hippo pathway. Oncogene. 2021 Mar;40(11):2018-2034. doi: 10.1038/s41388-021-01676-x. Epub 2021 Feb 18.
Ref 10 Etoposide in combination with erastin synergistically altered iron homeostasis and induced ferroptotic cell death through regulating IREB2/FPN1 expression in estrogen receptor positive-breast cancer cells. Life Sci. 2023 Jan 1;312:121222. doi: 10.1016/j.lfs.2022.121222. Epub 2022 Nov 25.
Ref 11 NR5A2 synergizes with NCOA3 to induce breast cancer resistance to BET inhibitor by upregulating NRF2 to attenuate ferroptosis. Biochem Biophys Res Commun. 2020 Sep 17;530(2):402-409. doi: 10.1016/j.bbrc.2020.05.069. Epub 2020 Jun 11.
Ref 12 Isoliquiritin modulates ferroptosis via NF-B signaling inhibition and alleviates doxorubicin resistance in breast cancer. Immunopharmacol Immunotoxicol. 2023 Dec;45(4):443-454. doi: 10.1080/08923973.2023.2165943. Epub 2023 Jan 17.
Ref 13 Metformin may induce ferroptosis by inhibiting autophagy via lncRNA H19 in breast cancer. FEBS Open Bio. 2022 Jan;12(1):146-153. doi: 10.1002/2211-5463.13314. Epub 2021 Oct 27.
Ref 14 Sulfasalazineinduced ferroptosis in breast cancer cells is reduced by the inhibitory effect of estrogen receptor on the transferrin receptor. Oncol Rep. 2019 Aug;42(2):826-838. doi: 10.3892/or.2019.7189. Epub 2019 Jun 6.
Ref 15 TMEM189 promotes breast cancer through inhibition of autophagy-regulated ferroptosis. Biochem Biophys Res Commun. 2022 Sep 24;622:37-44. doi: 10.1016/j.bbrc.2022.06.024. Epub 2022 Jun 12.
Ref 16 Intercellular interaction dictates cancer cell ferroptosis via NF2-YAP signalling. Nature. 2019 Aug;572(7769):402-406. doi: 10.1038/s41586-019-1426-6. Epub 2019 Jul 24.
Ref 17 Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells. Cell Death Dis. 2016 Jul 21;7(7):e2307. doi: 10.1038/cddis.2016.208.
Ref 18 Kinome screen of ferroptosis reveals a novel role of ATM in regulating iron metabolism. Cell Death Differ. 2020 Mar;27(3):1008-1022. doi: 10.1038/s41418-019-0393-7. Epub 2019 Jul 18.
Ref 19 A novel anticancer property of Lycium barbarum polysaccharide in triggering ferroptosis of breast cancer cells. J Zhejiang Univ Sci B. 2022 Apr 15;23(4):286-299. doi: 10.1631/jzus.B2100748.
Ref 20 Tubastatin A potently inhibits GPX4 activity to potentiate cancer radiotherapy through boosting ferroptosis. Redox Biol. 2023 Jun;62:102677. doi: 10.1016/j.redox.2023.102677. Epub 2023 Mar 17.
Ref 21 RUNX1-IT1 favors breast cancer carcinogenesis through regulation of IGF2BP1/GPX4 axis. Discov Oncol. 2023 Apr 10;14(1):42. doi: 10.1007/s12672-023-00652-z.
Ref 22 ATF2 inhibits ani-tumor effects of BET inhibitor in a negative feedback manner by attenuating ferroptosis. Biochem Biophys Res Commun. 2021 Jun 18;558:216-223. doi: 10.1016/j.bbrc.2020.08.113. Epub 2020 Sep 30.
Ref 23 AKT activation because of PTEN loss upregulates xCT via GSK3/NRF2, leading to inhibition of ferroptosis in PTEN-mutant tumor cells. Cell Rep. 2023 May 30;42(5):112536. doi: 10.1016/j.celrep.2023.112536. Epub 2023 May 20.
Ref 24 TYRO3 induces anti-PD-1/PD-L1 therapy resistance by limiting innate immunity and tumoral ferroptosis. J Clin Invest. 2021 Apr 15;131(8):e139434. doi: 10.1172/JCI139434.
Ref 25 Cytotoxic effects of the biflavonoids isolated from Selaginella trichoclada on MCF-7 cells and its potential mechanism. Bioorg Med Chem Lett. 2022 Jan 15;56:128486. doi: 10.1016/j.bmcl.2021.128486. Epub 2021 Dec 4.
Ref 26 SLC7A11/ xCT is a target of miR-5096 and its restoration partially rescues miR-5096-mediated ferroptosis and anti-tumor effects in human breast cancer cells. Cancer Lett. 2021 Dec 1;522:211-224. doi: 10.1016/j.canlet.2021.09.033. Epub 2021 Sep 24.
Ref 27 Ferroptotic cell death triggered by conjugated linolenic acids is mediated by ACSL1. Nat Commun. 2021 Apr 14;12(1):2244. doi: 10.1038/s41467-021-22471-y.
Ref 28 Shuganning injection, a traditional Chinese patent medicine, induces ferroptosis and suppresses tumor growth in triple-negative breast cancer cells. Phytomedicine. 2021 May;85:153551. doi: 10.1016/j.phymed.2021.153551. Epub 2021 Mar 18.
Ref 29 CircGFRA1 facilitates the malignant progression of HER-2-positive breast cancer via acting as a sponge of miR-1228 and enhancing AIFM2 expression. J Cell Mol Med. 2021 Nov;25(21):10248-10256. doi: 10.1111/jcmm.16963. Epub 2021 Oct 20.
Ref 30 Targeted Intervention of NF2-YAP Signaling Axis in CD24-Overexpressing Cells Contributes to Encouraging Therapeutic Effects in TNBC. ACS Nano. 2022 Apr 26;16(4):5807-5819. doi: 10.1021/acsnano.1c10921. Epub 2022 Apr 14.
Ref 31 xCT Inhibition Increases Sensitivity to Vorinostat in a ROS-Dependent Manner. Cancers (Basel). 2020 Mar 30;12(4):827. doi: 10.3390/cancers12040827.
Ref 32 A novel circular RNA confers trastuzumab resistance in human epidermal growth factor receptor 2-positive breast cancer through regulating ferroptosis. Environ Toxicol. 2022 Jul;37(7):1597-1607. doi: 10.1002/tox.23509. Epub 2022 Mar 2.
Ref 33 Functional interactions of the cystine/glutamate antiporter, CD44v and MUC1-C oncoprotein in triple-negative breast cancer cells. Oncotarget. 2016 Mar 15;7(11):11756-69. doi: 10.18632/oncotarget.7598.
Ref 34 The Dual Regulation Effects of ESR1/NEDD4L on SLC7A11 in Breast Cancer Under Ionizing Radiation. Front Cell Dev Biol. 2022 Feb 16;9:772380. doi: 10.3389/fcell.2021.772380. eCollection 2021.
Ref 35 A compendium of kinetic modulatory profiles identifies ferroptosis regulators. Nat Chem Biol. 2021 Jun;17(6):665-674. doi: 10.1038/s41589-021-00751-4. Epub 2021 Mar 8.
Ref 36 Salinomycin kills cancer stem cells by sequestering iron in lysosomes. Nat Chem. 2017 Oct;9(10):1025-1033. doi: 10.1038/nchem.2778. Epub 2017 May 16.
Ref 37 Ferroptosis and autophagy induced cell death occur independently after siramesine and lapatinib treatment in breast cancer cells. PLoS One. 2017 Aug 21;12(8):e0182921. doi: 10.1371/journal.pone.0182921. eCollection 2017.
Ref 38 Diverse compounds from pleuromutilin lead to a thioredoxin inhibitor and inducer of ferroptosis. Nat Chem. 2019 Jun;11(6):521-532. doi: 10.1038/s41557-019-0261-6. Epub 2019 May 13.
Ref 39 Herceptin induces ferroptosis and mitochondrial dysfunction in H9c2cells. Int J Mol Med. 2022 Feb;49(2):17. doi: 10.3892/ijmm.2021.5072. Epub 2021 Dec 22.
Ref 40 Neoadjuvant neratinib promotes ferroptosis and inhibits brain metastasis in a novel syngeneic model of spontaneous HER2(+ve) breast cancer metastasis. Breast Cancer Res. 2019 Aug 13;21(1):94. doi: 10.1186/s13058-019-1177-1.
Ref 41 Piperlongumine rapidly induces the death of human pancreatic cancer cells mainly through the induction of ferroptosis. Int J Oncol. 2018 Mar;52(3):1011-1022. doi: 10.3892/ijo.2018.4259. Epub 2018 Jan 31.
Ref 42 The alkaloid, soyauxinium chloride, displays remarkable cytotoxic effects towards a panel of cancer cells, inducing apoptosis, ferroptosis and necroptosis. Chem Biol Interact. 2021 Jan 5;333:109334. doi: 10.1016/j.cbi.2020.109334. Epub 2020 Nov 24.
Ref 43 Synthetic lethal combination targeting BET uncovered intrinsic susceptibility of TNBC to ferroptosis. Sci Adv. 2020 Aug 21;6(34):eaba8968. doi: 10.1126/sciadv.aba8968. Print 2020 Aug.
Ref 44 Prominin2 Drives Ferroptosis Resistance by Stimulating Iron Export. Dev Cell. 2019 Dec 2;51(5):575-586.e4. doi: 10.1016/j.devcel.2019.10.007. Epub 2019 Nov 14.
Ref 45 WTAP Mediates NUPR1 Regulation of LCN2 Through m(6)A Modification to Influence Ferroptosis, Thereby Promoting Breast Cancer Proliferation, Migration and Invasion. Biochem Genet. 2023 Jul 21. doi: 10.1007/s10528-023-10423-8. Online ahead of print.
Ref 46 Tumor resistance to ferroptosis driven by Stearoyl-CoA Desaturase-1 (SCD1) in cancer cells and Fatty Acid Biding Protein-4 (FABP4) in tumor microenvironment promote tumor recurrence. Redox Biol. 2021 Jul;43:102006. doi: 10.1016/j.redox.2021.102006. Epub 2021 May 14.
Ref 47 The ncRNA-Mediated Overexpression of Ferroptosis-Related Gene EMC2 Correlates With Poor Prognosis and Tumor Immune Infiltration in Breast Cancer. Front Oncol. 2021 Dec 8;11:777037. doi: 10.3389/fonc.2021.777037. eCollection 2021.
Ref 48 Circular RNA RHOT1 promotes progression and inhibits ferroptosis via mir-106a-5p/STAT3 axis in breast cancer. Aging (Albany NY). 2021 Mar 3;13(6):8115-8126. doi: 10.18632/aging.202608. Epub 2021 Mar 3.
Ref 49 Zinc transporter ZIP7 is a novel determinant of ferroptosis. Cell Death Dis. 2021 Feb 19;12(2):198. doi: 10.1038/s41419-021-03482-5.