Skip to main content
Erschienen in: Experimental Hematology & Oncology 1/2023

Open Access 01.12.2023 | Research

Targeting NRF2 uncovered an intrinsic susceptibility of acute myeloid leukemia cells to ferroptosis

verfasst von: Xin Liu, Shuxin Zhong, Kangjie Qiu, Xi Chen, Weiyue Wu, Jiamian Zheng, Yanwen Liu, Haolong Wu, Shiyun Fan, Dingrui Nie, Xianfeng Wang, Zhi Yu, Ziwei Liao, Mengjun Zhong, Yangqiu Li, Chengwu Zeng

Erschienen in: Experimental Hematology & Oncology | Ausgabe 1/2023

Abstract

Drug resistance and poor treatment response are major obstacles to the effective treatment of acute myeloid leukemia (AML). A deeper understanding of the mechanisms regulating drug resistance and response genes in AML is therefore urgently needed. Our previous research has highlighted the important role of nuclear factor E2-related factor 2 (NRF2) in AML, where it plays a critical role in detoxifying reactive oxygen species and influencing sensitivity to chemotherapy. In this study, we identify a core set of direct NRF2 targets that are involved in ferroptosis, a novel form of cell death. Of particular interest, we find that glutathione peroxidase 4 (GPX4) is a key ferroptosis gene that is consistently upregulated in AML, and high expression of GPX4 is associated with poor prognosis for AML patients. Importantly, simultaneous inhibition of NRF2 with ML385 and GPX4 with FIN56 or RSL3 synergistically targets AML cells, triggering ferroptosis. Treatment with ML385 + FIN56/RSL3 resulted in a marked reduction in NRF2 and GPX4 expression. Furthermore, NRF2 knockdown enhanced the sensitivity of AML cells to the ferroptosis inducers. Taken together, our results suggest that combination therapy targeting both NRF2 and GPX4 may represent a promising approach for the treatment of AML.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s40164-023-00411-4.
Xin Liu and Shuxin Zhong have contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Acute myeloid leukemia (AML) is the most common type of leukemia in adults, characterized by the uncontrolled proliferation of myeloid cells in the blood, bone marrow, and other tissues [14]. Despite recent advancements in treatment, AML patients with recurrent genetic abnormalities still have a poor prognosis, and relapsed or refractory AML poses a significant challenge[5]. Fortunately, in recent years, new drugs have emerged that provide promising options for relapsed or refractory AML treatment, and the search for novel targeted therapies offers hope for effective combination strategies against AML [6, 7].
Nuclear factor E2-related factor 2 (NRF2) is a crucial regulator in the anti-oxidative stress pathway [8]. However, recent studies have shown that enhanced reactive oxygen species (ROS) detoxification and other NRF2 functions can be pro-tumorigenic [9, 10]. Activating mutations in NRF2 and KEAP1 can promote tumor growth in several types of cancer [11, 12]. It was recently proposed that the NRF2-mediated pathway is a common survival pathway in AML [13]. In addition, studies have suggested that increased NRF2 expression mediated by oncogenes is a novel mechanism for activating the NRF2 antioxidant program in solid tumors [9]. Although many genes are known to be regulated by NRF2 in cancer cells, the direct targets of NRF2 are not yet fully understood [14, 15]. Therefore, a better understanding of the NRF2 pathway could pave the way for new therapeutic interventions in cancer treatment.
Ferroptosis is a newly identified form of non-apoptotic cell death that is characterized by iron-dependent activation of lipoxygenase and subsequent lipid peroxidation (LPO), and it has emerged as a critical factor in various types of cancers [1620], including leukemia [21]. Ferroptosis-induced cancer cell death presents a promising opportunity for anticancer therapy [14, 22]. One key regulator of cell survival and growth is NRF2, which induces multiple antioxidant genes, including glutathione peroxidase 4 (GPX4) [14]. GPX4 is a central defense enzyme that plays a critical role in catalyzing lethal lipid peroxide into nontoxic lipid alcohol to counteract LPO and ferroptosis [23, 24]. Interestingly, previous studies have highlighted the critical role of NRF2 in ROS detoxification and chemotherapy sensitivity in AML. However, the precise mechanisms underlying the regulation of ferroptosis by NRF2 in AML remained poorly understood until this study. In addition, the regulatory and functional roles of GPX4 in the treatment response of AML cells have not been fully explored.
In this study, we identified a novel mechanism by which NRF2 activation leads to overexpression of GPX4 in AML, resulting in chemoresistance and evasion of ferroptosis. By using various ferroptosis inducers and inhibitors, they have shown that NRF2-deficient AML cells are particularly sensitive to ferroptosis inducers and that NRF2-induced GPX4 supports AML cell survival against drug treatment. The study also demonstrates that combined inhibition of NRF2 and GPX4 can significantly reduce the expression of both genes, thereby facilitating ferroptosis. These findings provide important insights into the mechanism of chemoresistance in AML and suggest that targeting NRF2 and GPX4 may be a promising therapeutic strategy for the treatment of this disease.

Materials and methods

Clinical samples

Bone marrow (BM) samples were obtained from sixteen patients with newly diagnosed AML (characteristics provided in the Supplemental Table 1). Peripheral blood (PB) samples were obtained from eight healthy individuals (HIs). All samples were obtained with written informed consent in accordance with the ethical committee of the First Affiliated Hospitals of Jinan University.

Cell lines and reagents

AML cell lines HL60, MV4;11, KG1a, U937, HEL, Molm13, NB4, KG1 and Kasumi-1 were cultured in RPMI 1640 (Invitrogen) containing 10% fetal bovine serum (FBS). HEK293T cells were cultured in DMEM (Invitrogen) medium containing 10% FBS. All these cell lines were cultured in a humidified atmosphere containing 5% CO2 at 37 °C. Ferrostatin-1, ML385, FIN56, and RSL3 were purchased from Selleck Chemicals (Houston, TX, USA).

Establishment of stable cell lines

Specific oligonucleotides directed against human NRF2 were designed and cloned into the pLKO.1-puro plasmid according to the protocol recommended by Addgene, and empty pLKO.1-puro plasmid served as control. Lentiviruses carrying pLKO.1-puro were prepared by the transfection of standard packaging vectors into HEK293T cells using lipofectamine 3000 (Life Technologies, USA). Viral supernatant was harvested 24 and 48 h after transfection. A total of 2 × 105 MV4;11 cells were co-cultured with virus supernatant plus 8 µg/mL polybrene (Sigma-Aldrich, St Louis, USA). Then 5 µg/mL puromycin was applied to select positively infected cells for at least 5 days. The shRNA sequences are listed in Supplemental Material 1. For ectopic expression of NRF2, lentiviruses overexpressing NRF2 (LV-NRF2) and empty viruses (LV-NC) were purchased from Genechem (Shanghai, China). A total of 5 µg/mL puromycin was used to select positively infected cells for at least 5 days.

Detection of cell cycle and cell death

For analysis of cell death, cells were harvested, and the Annexin-V-APC/PI Detection Kit (MultiSciences, Shanghai, China) was used following the manufacturer’s protocols and performed as previously described [25, 26]. For cell cycle assay, the Cell Cycle Staining Kit (MultiSciences, Shanghai, China) was used according to the manufacturer’s instructions. Cells were collected by CytoFLEX, and data were analyzed by Flowjo software.

Quantitative real-time PCR analysis

Total RNA from cells were extracted using TRIzol reagent (Invitrogen, CA, USA) following the manufacturer’s instructions and was reversed transcribed into cDNA using High-Capacity cDNA Reverse Transcription Kits (Applied Biosystems, CA, USA). Quantitative real-time PCR (qRT-PCR) was performed with SYBR Green (Tiangen, China) in a total volume of 20 µl using the following program: 95°C for 5 min followed by 40 cycles of 95℃ for 30s, 60℃ for 30s, and 72℃ for 30s. Relative target gene expression levels were normalized to the mRNA level of β-actin. The primers for qRT-PCR are shown in Supplemental Material 1.

Western blotting

Cells were collected and lysed in 1х SDS loading buffer containing protease inhibitors. Protein lysates were separated using 12% SDS-PAGE and subsequently transferred to polyvinylidene fluoride membrane (Millipore, USA) following by blocking with QuickBlock™ blocking buffer (Beyotime, China) and incubation with antibodies directed against GPX4 (1:1000, 52,455, CST, MA, USA) or NRF2 (1:800, sc-365,949, Santa cruz) overnight at 4℃. Anti-β-actin antibody (1:5,000, Beyotime, China) served as control. After incubation with a secondary antibody, ECL (Beyotime, China) was used to amplify the binding antibody signal, and images were obtained with a UVITEC photo documenter. The full gel images in the paper figures are displayed in Figure S1.

Small interfering RNA (siRNA) transfection

The Neon® Transfection System (Invitrogen) was used to transfect 100 pmol oligonucleotides into MV4;11 cells in a total volume of 10 µl [27, 28]. In brief, 2 × 105 cells were suspended in a volume of 10 µl with 100 pmol oligonucleotides and transfected three times in each 12-well plate. After electroporation, cells were cultured in RPMI 1640 medium supplemented with 10% FBS at 37 °C and 5% CO2 for 24 h, and subsequently divided into two groups with or without drug treatment. The cells were then collected for protein level detection and evaluation of cell death. Supplemental Material 1 lists the siRNA sequences targeting GPX4.

Measurement of lipid peroxidation (LPO)

To detect LPO, C11-BODIPY 581/591 (Abclonal, China) was employed following the manufacturer’s instructions. In brief, cells were labeled with C11-BODIPY (2.5 µM) in 400 µL RPMI 1640 medium and incubated at 37 °C and 5% CO2 in a cell incubator away from light for 60 min. The cells were then washed twice with PBS, resuspended in 200 µL of fresh PBS, and analyzed using flow cytometry.

RNA-seq analysis

RNA from LV-NC and LV-NRF2 MV4;11 cells was extracted using TRIzol reagent. Amplified cDNA was sequentially converted into short-read sequencing libraries using the VAHTS Universal V6 RNA-seq Library Prep Kit for Illumina® Hisat2 (version: 2.0.4). Sequencing was performed by Novogene Co. Ltd., Tianjin Biotechnology Corporation. The expression level of each gene was calculated as fragments per kilobase of exon model per million mapped reads (FPKM).

Statistical analysis

Unless otherwise stated in the figure legends, all data are presented as the mean ± SD of three independent experiments. Statistical analysis was conducted using SPSS 22.0 software on data from independent biological replicates. Student’s t-test was used to determine the significance of differences between two groups. One-way ANOVA followed by either Bonferroni or Dunnett’s post hoc test was applied to compare three or more groups, as specified in the figure legends. A p value < 0.05 was considered significant. *, p < 0.05, **, p < 0.01, ***, p < 0.001.

Results

NRF2 target gene analysis reveals its role in ferroptosis

Our recent studies have demonstrated that NRF2 promotes the expression of key molecules involved in AML development and drug sensitivity. To identify potential therapeutic targets for AML, we performed a broad screening of direct NRF2 target genes (Fig. 1a). Our hypothesis was that NRF2 target genes would contain ChIP-Seq binding peaks in their vicinity and show altered expression levels in RNA-seq studies. To generate a list of NRF2 target genes, we initially utilized several ChIP-Seq profiling datasets (ENCSR707IUN_1, GSM2394418, GSM2423706, GSM2423560) and identified 349 unique overlapping genes. These represent a high-confidence set of NRF2-bound genomic regions (Fig. 1b). The majority of the identified binding sites were found near potential NRF2 target genes, suggesting that these genes could be regulated by NRF2. We further performed KEGG pathway analysis on these potential NRF2 target genes and found that they were significantly enriched in several terms, including ferroptosis and glutathione metabolism, both of which are closely related to ferroptosis (Fig. 1c, Supplemental Material 2). Next, we conducted RNA-seq to identify NRF2 target genes in AML. We identified over 3,000 upregulated genes in NRF2-overexpressing MV4;11 cells (log2 fold change ≥ 0.25). Our results revealed the upregulation of numerous antioxidative and ferroptosis genes in response to NRF2 overexpression. Subsequently, we identified 70 unique genes overlapping between ChIP-Seq and RNA-seq (Fig. 1d), and compiled them in Supplemental Material 3. Consistent with the previous findings, we found that the NRF2 targets are mainly involved in the processes of ferroptosis and glutathione metabolism (Fig. 1e, Supplemental Material 4). Both NRF2 and ferroptosis have been shown to be linked to cancer therapy resistance. It is intriguing to speculate that these NRF2 targets may act in concert to function as anti-ferroptosis mediators upon drug treatment in AML.
Using qRT-PCR analysis, we validated that several of these genes are indeed transcriptional targets of NRF2 in AML cells (Fig. 1f-g). Among the selected genes, including SLC7A11, GCLC, GPX4, FTH1, HMOX1, GCLM, and FTL, GPX4 mRNA was upregulated upon lentivirus-mediated NRF2 overexpression and downregulated upon shRNA-mediated NRF2 (shNRF2) knockdown. Furthermore, GPX4 was also upregulated in response to NRF2 activation by arsenic trioxide (ATO) and cytarabine (Ara-C) treatment (Figure S2). Given the known activation of NRF2 in AML and its resistance to therapies, we propose that GPX4 may be one of the key targets that confer an anti-ferroptosis role to NRF2.

NRF2-regulated GPX4 was increased and associated with poor prognosis in AML

To confirm the regulatory role of NRF2 on GPX4 in AML, we used western blotting to detect changes in GPX4 expression after manipulating NRF2 expression. As shown in Fig. 2a and b, overexpression of NRF2 led to increased GPX4 expression, while knockdown of NRF2 resulted in decreased GPX4 expression. Moreover, we observed higher expression of NRF2 and GPX4 in the BM of AML patients compared with HIs (Fig. 2c). Among several AML cell lines, we found high expression of GPX4 in MV4;11 and Kasumi-1 cells (Fig. 2c, right panel), which were subsequently used in our experiments. We also analyzed the expression of NRF2 and GPX4 in 15 AML patients and found a positive correlation between the two (Fig. 2d). Furthermore, survival analysis of AML patients included in The Cancer Genome Atlas (TCGA) dataset showed that both NRF2 and GPX4 expression were significantly associated with poor overall survival (OS) (Fig. 2e and Figure S3a-b), indicating that their increased expression is associated with poor prognosis in AML. In addition, the NRF2 inhibitor ML385 reduced GPX4 protein levels in a concentration-dependent manner (Fig. 2f). We also found that inhibiting NRF2 with ML385 inhibited the viability of various AML cell lines (Fig. 2g), suggesting that NRF2 and GPX4 could represent promising targets for the treatment of AML cancer cells.

NRF2 inhibition (ML385) cooperates with GPX4 inhibition (FIN56/RSL3) to reduce the viability and induce the cell death of AML cells

It has been reported that RSL3 and FIN56 can specifically inhibit GPX4 to induce the ferroptosis of tumor cells [14, 29, 30]. To investigate the synergistic effects of NRF2 and GPX4 inhibition in AML cells, we first explored the use of these specific GPX4 inhibitors to induce ferroptosis in AML cells. Our experiments showed that treatment with GPX4 inhibitors resulted in a dose-dependent suppression of GPX4 expression in MV4;11 cells (Fig. 3a). We also found that RSL3 inhibited the viability of various AML cell lines in a concentration-dependent manner, while having little effect on peripheral blood mononuclear cells (PBMCs) from HIs, indicating its relative safety (Fig. 3b). Next, AML cell lines MV4;11, Kasumi-1, and KG1α were treated with NRF2 inhibitor ML385 alone, a GPX4 inhibitor (FIN56 or RSL3) alone, or ML385 combined with a GPX4 inhibitor. We found that combined administration of ML385 and GPX4 inhibitor markedly inhibits the proliferation of AML cell lines compared with either inhibitor alone, and the combination index (CI) indicating that these two inhibitors have synergistic effects (Fig. 3c and d and S4a).
Further studies revealed that treatment with ML385 and a GPX4 inhibitor significantly increases the cell death of MV4;11 and Kasumi-1 cells (Fig. 4a, b). In line with these findings, it was observed that ML385 acted in collaboration with Erastin, another ferroptosis inducer, to induce cell death in MV4;11 cells (Figure S4b). Furthermore, our study has uncovered that RSL3 enhances the effectiveness of the primary drugs used to treat AML like Ara-C (Figure S4c). Additionally, we have observed that treatment with a combination of ML385 and GPX4 inhibitors results in the arrest of the cell cycle of MV4;11 cells in the G0/G1 phase, as demonstrated in Fig. 4c. We further examined if the co-administration of ML385 and a GPX4 inhibitor would augment the cytotoxic effects in cells obtained from newly diagnosed AML patients. Comparable to the findings in AML cell lines, the combined use of ML385 and FIN56/RSL3 inhibited the viability and stimulated the cell death of primary AML cells, as depicted in Fig. 4d and e. In summary, these outcomes imply that the combination of inhibiting NRF2 and GPX4 produces a more robust cytotoxic effect on AML cells.

ML385 cooperates with FIN56/RSL3 to promote ferroptosis of AML cells

Previous studies have shown that GPX4 plays a crucial role in ferroptosis and chemoresistance in human cancers. Activation of the NRF2 pathway plays a dominant role in protecting human cancer cells against cell death. To investigate potential synergistic effects of ML385 and GPX4 inhibitors on ferroptosis, we assessed the combined impact of these agents on lipid peroxidation levels using the C11-BODIPY probe. As depicted in Fig. 5a and b, co-treatment with ML385 and GPX4 inhibitors markedly elevated lipid-based ROS accumulation in MV4;11 and Kasumi-1 cells. Consistent with these results, simultaneous treatment with ML385 and RSL3 led to the highest cell death compared to ML385 or RSL3 alone in MV4;11 cells (Fig. 5c). Furthermore, we confirmed that the enhanced cytotoxic effects of ML385 and RSL3 in AML cell lines was blocked by ferrostatin-1 (Fer-1, an inhibitor of ferroptosis) (Fig. 5d). The above data suggested that ML385 may act synergistically with FIN56/RSL3 to induce ferroptosis via the NRF2/GPX4 pathway.

Synergistic effects of ML385 and FIN56 in downregulating NRF2 and GPX4 expression

To further investigate the synergistic effects of NRF2 and GPX4 inhibition, MV4;11 and Kasumi-1 cells were treated with ML385 or GPX4 inhibitor alone or ML385 combined with GPX4 inhibitor, and the expression of NRF2 and GPX4 was detected by western blotting. As shown in Fig. 6a-b, the GPX4 inhibitor combined with the NRF2 inhibitor resulted in the lowest expression of GPX4 and NRF2. At the same time, transfection of shNRF2 sharply increased the sensitivity of MV4;11 cells to GPX4 inhibitors (Fig. 6c). We next measured the level of intracellular lipid ROS in shNRF2-transfected MV4;11 cells to confirm whether knockdown of NRF2 could increase the oxidation of lipids. As expected, down-regulation of NRF2 promoted GPX4 inhibitors-induced lipid oxidation (Fig. 6d). To investigate the impact of NRF2 expression on AML cells treated with GPX4 inhibitors, we overexpressed NRF2 in MV4;11 cells and treated them with RSL3 at the indicated dose. Our results showed that NRF2 overexpression partially restored the cytotoxic effects of RSL3 on MV4;11 cells, as depicted in Fig. 6e. Additionally, we employed two small interfering RNAs (siRNAs) to target GPX4 and explored its effects on the reactivity of AML cells to ferroptosis (Fig. 6f). Our findings indicated that transfection of siRNA targeting GPX4 significantly increased the sensitivity of MV4;11 cells to RSL3 and FIN56, as demonstrated in Fig. 6g. Overall, our results suggest that targeting NRF2 and GPX4 may enhance the sensitivity of AML cells to ferroptosis.

Discussion

In this study, we made three significant observations. First, the NRF2 protein induced high constitutive GPX4 levels in AML. Second, GPX4 is one of the most important NRF2 target genes in AML. Third, NRF2/GPX4 attenuates ferroptosis in AML.
Previously, we established that NRF2 plays a vital role in the detoxification of ROS and the resistance to chemotherapy in AML [31]. As such, the primary aim of this study was to investigate the detailed mechanisms of NRF2 in chemoresistance. The NRF2 signaling pathway is notably activated in AML [13, 32], and our prior studies have revealed that the transcriptional activation of the NRF2 signaling pathway leads to high expression of miR-125b in AML [31]. Recent research has also suggested that NRF2 transcription promotes GPX4 expression. In agreement with these findings, we have demonstrated that NRF2 can promote an increase in GPX4 mRNA and protein expression. Our data indicate that GPX4 is one of the most important target genes of NRF2 in AML.
Elevated GPX4 levels are frequently observed in many types of cancer, implying that it may be protumorigenic. Our study has found that GPX4 is highly expressed in AML cells, and elevated GPX4 levels correlate with poor prognosis in AML patients. This suggests that GPX4 may play a critical role in the development and treatment of AML. GPX4 functions as a central defensive enzyme against ferroptosis, and previous studies have indicated that NRF2 may regulate GPX4 expression and be related to ferroptosis in solid cancers [12, 33]. Similarly, we found that GPX4 is directly regulated by NRF2 and that it is resistant to ferroptosis. Combined inhibition of NRF2 with ML385 and GPX4 with RSL3/FIN56 synergistically targets AML cells. Consistent with these findings, ML385 cooperated with another ferroptosis inducer, Erastin, to induce cell death in MV4;11 cells. NRF2 knockdown increased the sensitivity of AML cells to FIN56 and RSL3. At the molecular level, upregulation of GPX4 by NRF2 was found to reduce the cytotoxic effects of these ferroptosis inducers. Still, this resistance could be reversed by adding an NRF2 inhibitor in combination. Similarly, we found significant concurrent down-regulation of NRF2 and GPX4 in response to ML385 + FIN56/RSL3 treatment. Thus, our data suggest that dysregulation of NRF2/GPX4 signaling may be a potential barrier to ferroptosis inducers.
Chemotherapy-induced cell death is often associated with the generation of ROS [3436]. High levels of ROS can cause oxidative stress and lead to the death of cancer cells [3740]. However, in AML, chemotherapy resistance is associated with the activation of NRF2. Our previous study showed that chemotherapy increased NRF2 levels in AML cells, and NRF2 activation contributes to chemotherapy resistance [31]. Recently, it has been suggested that ferroptosis is closely linked to the sensitivity of cancer cells to chemotherapy, and induction of ferroptosis may overcome chemotherapy resistance [41]. In fact, several studies have demonstrated that the ferroptosis inducer Erastin can enhance the anticancer activity of Ara-C and doxorubicin [42]. In this study, we also found that RSL3, a GPX4 inhibitor and ferroptosis inducer, enhanced the anticancer activity of Ara-C in AML cells, leading to increased cell death. Our findings suggest that targeting the NRF2/GPX4 pathway and inducing ferroptosis may be an effective strategy for enhancing chemo-sensitivity in AML patients, and may lead to the development of novel combination therapies.

Conclusions

Our investigation has revealed that the expression of GPX4 in AML is likely attributed to the activity of NRF2 in AML cells. Furthermore, we have demonstrated that the NRF2/GPX4 axis plays a crucial part in ferroptosis and the antileukemia response. Based on these findings, our study provides supporting evidence and a basis for the targeting of NRF2/GPX4 as a strategy for developing therapeutics to improve chemo-sensitivity in AML.

Acknowledgements

We express our gratitude to the Medical Experimental Center of the School of Medicine, Jinan University for their support. We would also like to acknowledge the resources provided by TCGA dataset (htps://portal,gdc.cancer,gov) and Cistrome Data Browser (http://​cistrome.​org/​db/​#/​) (ENCSR707IUN_1, GSM2394418, GSM2423706, GSM2423560) for their valuable datasets and analysis.

Declarations

Ethics approval

This study was approved by the ethics committee of the affiliated hospitals of Jinan University. Written informed consent was obtained from all patients.

Competing interests

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Deschler B, Lübbert M. Acute myeloid leukemia: epidemiology and etiology. Cancer. 2006;107(9):2099–107.CrossRefPubMed Deschler B, Lübbert M. Acute myeloid leukemia: epidemiology and etiology. Cancer. 2006;107(9):2099–107.CrossRefPubMed
2.
Zurück zum Zitat Yao Y, Li F, Huang J, Jin J, Wang H. Leukemia stem cell-bone marrow microenvironment interplay in acute myeloid leukemia development. Exp Hematol Oncol. 2021;10(1):39.CrossRefPubMedPubMedCentral Yao Y, Li F, Huang J, Jin J, Wang H. Leukemia stem cell-bone marrow microenvironment interplay in acute myeloid leukemia development. Exp Hematol Oncol. 2021;10(1):39.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Keykhaei M, Masinaei M, Mohammadi E, Azadnajafabad S, Rezaei N, Saeedi Moghaddam S, et al. A global, regional, and national survey on burden and quality of Care Index (QCI) of hematologic malignancies; global burden of disease systematic analysis 1990–2017. Exp Hematol Oncol. 2021;10(1):11.CrossRefPubMedPubMedCentral Keykhaei M, Masinaei M, Mohammadi E, Azadnajafabad S, Rezaei N, Saeedi Moghaddam S, et al. A global, regional, and national survey on burden and quality of Care Index (QCI) of hematologic malignancies; global burden of disease systematic analysis 1990–2017. Exp Hematol Oncol. 2021;10(1):11.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Yi M, Li A, Zhou L, Chu Q, Song Y, Wu K. The global burden and attributable risk factor analysis of acute myeloid leukemia in 195 countries and territories from 1990 to 2017: estimates based on the global burden of disease study 2017. J Hematol Oncol. 2020;13(1):72.CrossRefPubMedPubMedCentral Yi M, Li A, Zhou L, Chu Q, Song Y, Wu K. The global burden and attributable risk factor analysis of acute myeloid leukemia in 195 countries and territories from 1990 to 2017: estimates based on the global burden of disease study 2017. J Hematol Oncol. 2020;13(1):72.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Cao X, Dai H, Cui Q, Li Z, Shen W, Pan J, et al. CD7-directed CAR T-cell therapy: a potential immunotherapy strategy for relapsed/refractory acute myeloid leukemia. Exp Hematol Oncol. 2022;11(1):67.CrossRefPubMedPubMedCentral Cao X, Dai H, Cui Q, Li Z, Shen W, Pan J, et al. CD7-directed CAR T-cell therapy: a potential immunotherapy strategy for relapsed/refractory acute myeloid leukemia. Exp Hematol Oncol. 2022;11(1):67.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Short NJ, Konopleva M, Kadia TM, Borthakur G, Ravandi F, DiNardo CD, et al. Advances in the treatment of Acute myeloid leukemia: New Drugs and New Challenges. Cancer Discov. 2020;10(4):506–25.CrossRefPubMed Short NJ, Konopleva M, Kadia TM, Borthakur G, Ravandi F, DiNardo CD, et al. Advances in the treatment of Acute myeloid leukemia: New Drugs and New Challenges. Cancer Discov. 2020;10(4):506–25.CrossRefPubMed
7.
Zurück zum Zitat Newell LF, Cook RJ. Advances in acute myeloid leukemia. BMJ. 2021;375n2026. Newell LF, Cook RJ. Advances in acute myeloid leukemia. BMJ. 2021;375n2026.
8.
Zurück zum Zitat Yang J, Mo J, Dai J, Ye C, Cen W, Zheng X, et al. Cetuximab promotes RSL3-induced ferroptosis by suppressing the Nrf2/HO-1 signalling pathway in KRAS mutant colorectal cancer. Cell Death Dis. 2021;12(11):1079.CrossRefPubMedPubMedCentral Yang J, Mo J, Dai J, Ye C, Cen W, Zheng X, et al. Cetuximab promotes RSL3-induced ferroptosis by suppressing the Nrf2/HO-1 signalling pathway in KRAS mutant colorectal cancer. Cell Death Dis. 2021;12(11):1079.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat DeNicola GM, Karreth FA, Humpton TJ, Gopinathan A, Wei C, Frese K, et al. Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature. 2011;475(7354):106–9.CrossRefPubMedPubMedCentral DeNicola GM, Karreth FA, Humpton TJ, Gopinathan A, Wei C, Frese K, et al. Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature. 2011;475(7354):106–9.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Hayes JD, McMahon M. NRF2 and KEAP1 mutations: permanent activation of an adaptive response in cancer. Trends Biochem Sci. 2009;34(4):176–88.CrossRefPubMed Hayes JD, McMahon M. NRF2 and KEAP1 mutations: permanent activation of an adaptive response in cancer. Trends Biochem Sci. 2009;34(4):176–88.CrossRefPubMed
12.
Zurück zum Zitat Han Y, Gao X, Wu N, Jin Y, Zhou H, Wang W, et al. Long noncoding RNA LINC00239 inhibits ferroptosis in colorectal cancer by binding to Keap1 to stabilize Nrf2. Cell Death Dis. 2022;13(8):742.CrossRefPubMedPubMedCentral Han Y, Gao X, Wu N, Jin Y, Zhou H, Wang W, et al. Long noncoding RNA LINC00239 inhibits ferroptosis in colorectal cancer by binding to Keap1 to stabilize Nrf2. Cell Death Dis. 2022;13(8):742.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Rushworth SA, Zaitseva L, Murray MY, Shah NM, Bowles KM, MacEwan DJ. The high Nrf2 expression in human acute myeloid leukemia is driven by NF-kappaB and underlies its chemo-resistance. Blood. 2012;120(26):5188–98.CrossRefPubMed Rushworth SA, Zaitseva L, Murray MY, Shah NM, Bowles KM, MacEwan DJ. The high Nrf2 expression in human acute myeloid leukemia is driven by NF-kappaB and underlies its chemo-resistance. Blood. 2012;120(26):5188–98.CrossRefPubMed
14.
Zurück zum Zitat Wang Q, Bin C, Xue Q, Gao Q, Huang A, Wang K, et al. GSTZ1 sensitizes hepatocellular carcinoma cells to sorafenib-induced ferroptosis via inhibition of NRF2/GPX4 axis. Cell Death Dis. 2021;12(5):426.CrossRefPubMedPubMedCentral Wang Q, Bin C, Xue Q, Gao Q, Huang A, Wang K, et al. GSTZ1 sensitizes hepatocellular carcinoma cells to sorafenib-induced ferroptosis via inhibition of NRF2/GPX4 axis. Cell Death Dis. 2021;12(5):426.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Dong H, Xia Y, Jin S, Xue C, Wang Y, Hu R, et al. Nrf2 attenuates ferroptosis-mediated IIR-ALI by modulating TERT and SLC7A11. Cell Death Dis. 2021;12(11):1027.CrossRefPubMedPubMedCentral Dong H, Xia Y, Jin S, Xue C, Wang Y, Hu R, et al. Nrf2 attenuates ferroptosis-mediated IIR-ALI by modulating TERT and SLC7A11. Cell Death Dis. 2021;12(11):1027.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Nie D, Chen C, Li Y, Zeng C. Disulfiram, an aldehyde dehydrogenase inhibitor, works as a potent drug against sepsis and cancer via NETosis, pyroptosis, apoptosis, ferroptosis, and cuproptosis. Blood Sci. 2022;04(03):152–4.CrossRef Nie D, Chen C, Li Y, Zeng C. Disulfiram, an aldehyde dehydrogenase inhibitor, works as a potent drug against sepsis and cancer via NETosis, pyroptosis, apoptosis, ferroptosis, and cuproptosis. Blood Sci. 2022;04(03):152–4.CrossRef
17.
Zurück zum Zitat Wang C, Zeng J, Li LJ, Xue M, He SL. Cdc25A inhibits autophagy-mediated ferroptosis by upregulating ErbB2 through PKM2 dephosphorylation in cervical cancer cells. Cell Death Dis. 2021;12(11):1055.CrossRefPubMedPubMedCentral Wang C, Zeng J, Li LJ, Xue M, He SL. Cdc25A inhibits autophagy-mediated ferroptosis by upregulating ErbB2 through PKM2 dephosphorylation in cervical cancer cells. Cell Death Dis. 2021;12(11):1055.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Zhang H, Wang M, He Y, Deng T, Liu R, Wang W, et al. Chemotoxicity-induced exosomal lncFERO regulates ferroptosis and stemness in gastric cancer stem cells. Cell Death Dis. 2021;12(12):1116.CrossRefPubMedPubMedCentral Zhang H, Wang M, He Y, Deng T, Liu R, Wang W, et al. Chemotoxicity-induced exosomal lncFERO regulates ferroptosis and stemness in gastric cancer stem cells. Cell Death Dis. 2021;12(12):1116.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Xie Y, Wang B, Zhao Y, Tao Z, Wang Y, Chen G, et al. Mammary adipocytes protect triple-negative breast cancer cells from ferroptosis. J Hematol Oncol. 2022;15(1):72.CrossRefPubMedPubMedCentral Xie Y, Wang B, Zhao Y, Tao Z, Wang Y, Chen G, et al. Mammary adipocytes protect triple-negative breast cancer cells from ferroptosis. J Hematol Oncol. 2022;15(1):72.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Tong X, Tang R, Xiao M, Xu J, Wang W, Zhang B, et al. Targeting cell death pathways for cancer therapy: recent developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research. J Hematol Oncol. 2022;15(1):174.CrossRefPubMedPubMedCentral Tong X, Tang R, Xiao M, Xu J, Wang W, Zhang B, et al. Targeting cell death pathways for cancer therapy: recent developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research. J Hematol Oncol. 2022;15(1):174.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Thayyullathil F, Cheratta AR, Alakkal A, Subburayan K, Pallichankandy S, Hannun YA, et al. Acid sphingomyelinase-dependent autophagic degradation of GPX4 is critical for the execution of ferroptosis. Cell Death Dis. 2021;12(1):26.CrossRefPubMedPubMedCentral Thayyullathil F, Cheratta AR, Alakkal A, Subburayan K, Pallichankandy S, Hannun YA, et al. Acid sphingomyelinase-dependent autophagic degradation of GPX4 is critical for the execution of ferroptosis. Cell Death Dis. 2021;12(1):26.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Ding Y, Chen X, Liu C, Ge W, Wang Q, Hao X, et al. Identification of a small molecule as inducer of ferroptosis and apoptosis through ubiquitination of GPX4 in triple negative breast cancer cells. J Hematol Oncol. 2021;14(1):19.CrossRefPubMedPubMedCentral Ding Y, Chen X, Liu C, Ge W, Wang Q, Hao X, et al. Identification of a small molecule as inducer of ferroptosis and apoptosis through ubiquitination of GPX4 in triple negative breast cancer cells. J Hematol Oncol. 2021;14(1):19.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Zhong M, Gao R, Zhao R, Huang Y, Chen C, Li K, et al. BET bromodomain inhibition rescues PD-1-mediated T-cell exhaustion in acute myeloid leukemia. Cell Death Dis. 2022;13(8):671.CrossRefPubMedPubMedCentral Zhong M, Gao R, Zhao R, Huang Y, Chen C, Li K, et al. BET bromodomain inhibition rescues PD-1-mediated T-cell exhaustion in acute myeloid leukemia. Cell Death Dis. 2022;13(8):671.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Chen C, Nie D, Huang Y, Yu X, Chen Z, Zhong M, et al. Anticancer effects of disulfiram in T-cell malignancies through NPL4-mediated ubiquitin-proteasome pathway. J Leukoc Biol. 2022;112(4):919–29.CrossRefPubMed Chen C, Nie D, Huang Y, Yu X, Chen Z, Zhong M, et al. Anticancer effects of disulfiram in T-cell malignancies through NPL4-mediated ubiquitin-proteasome pathway. J Leukoc Biol. 2022;112(4):919–29.CrossRefPubMed
27.
Zurück zum Zitat Li K, Chen C, Gao R, Yu X, Huang Y, Chen Z, et al. Inhibition of BCL11B induces downregulation of PTK7 and results in growth retardation and apoptosis in T-cell acute lymphoblastic leukemia. Biomark Res. 2021;9(1):17.CrossRefPubMedPubMedCentral Li K, Chen C, Gao R, Yu X, Huang Y, Chen Z, et al. Inhibition of BCL11B induces downregulation of PTK7 and results in growth retardation and apoptosis in T-cell acute lymphoblastic leukemia. Biomark Res. 2021;9(1):17.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Zeng C, Liu S, Lu S, Yu X, Lai J, Wu Y, et al. The c-Myc-regulated lncRNA NEAT1 and paraspeckles modulate imatinib-induced apoptosis in CML cells. Mol Cancer. 2018;17(1):130.CrossRefPubMedPubMedCentral Zeng C, Liu S, Lu S, Yu X, Lai J, Wu Y, et al. The c-Myc-regulated lncRNA NEAT1 and paraspeckles modulate imatinib-induced apoptosis in CML cells. Mol Cancer. 2018;17(1):130.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156(1–2):317–31.CrossRefPubMedPubMedCentral Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156(1–2):317–31.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Shimada K, Skouta R, Kaplan A, Yang WS, Hayano M, Dixon SJ, et al. Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat Chem Biol. 2016;12(7):497–503.CrossRefPubMedPubMedCentral Shimada K, Skouta R, Kaplan A, Yang WS, Hayano M, Dixon SJ, et al. Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat Chem Biol. 2016;12(7):497–503.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Yu X, Mansouri A, Liu Z, Gao R, Li K, Chen C, et al. NRF2 activation induced by PML-RARalpha promotes microRNA 125b-1 expression and confers resistance to chemotherapy in acute promyelocytic leukemia. Clin Transl Med. 2021;11(5):e418.CrossRefPubMedPubMedCentral Yu X, Mansouri A, Liu Z, Gao R, Li K, Chen C, et al. NRF2 activation induced by PML-RARalpha promotes microRNA 125b-1 expression and confers resistance to chemotherapy in acute promyelocytic leukemia. Clin Transl Med. 2021;11(5):e418.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Rushworth SA, Bowles KM, MacEwan DJ. High basal nuclear levels of Nrf2 in acute myeloid leukemia reduces sensitivity to proteasome inhibitors. Cancer Res. 2011;71(5):1999–2009.CrossRefPubMed Rushworth SA, Bowles KM, MacEwan DJ. High basal nuclear levels of Nrf2 in acute myeloid leukemia reduces sensitivity to proteasome inhibitors. Cancer Res. 2011;71(5):1999–2009.CrossRefPubMed
33.
Zurück zum Zitat Salazar M, Rojo AI, Velasco D, de Sagarra RM, Cuadrado A. Glycogen synthase kinase-3beta inhibits the xenobiotic and antioxidant cell response by direct phosphorylation and nuclear exclusion of the transcription factor Nrf2. J Biol Chem. 2006;281(21):14841–51.CrossRefPubMed Salazar M, Rojo AI, Velasco D, de Sagarra RM, Cuadrado A. Glycogen synthase kinase-3beta inhibits the xenobiotic and antioxidant cell response by direct phosphorylation and nuclear exclusion of the transcription factor Nrf2. J Biol Chem. 2006;281(21):14841–51.CrossRefPubMed
34.
Zurück zum Zitat Wang X, Dawod A, Nachliely M, Harrison JS, Danilenko M, Studzinski GP. Differentiation agents increase the potential AraC therapy of AML by reactivating cell death pathways without enhancing ROS generation. J Cell Physiol. 2020;235(1):573–86.CrossRefPubMed Wang X, Dawod A, Nachliely M, Harrison JS, Danilenko M, Studzinski GP. Differentiation agents increase the potential AraC therapy of AML by reactivating cell death pathways without enhancing ROS generation. J Cell Physiol. 2020;235(1):573–86.CrossRefPubMed
35.
Zurück zum Zitat You BR, Park WH. MG132, a proteasome inhibitor-induced calf pulmonary arterial endothelial cell growth and death, are changed by MAPK inhibitors. Drug Chem Toxicol. 2011;34(1):45–52.CrossRefPubMed You BR, Park WH. MG132, a proteasome inhibitor-induced calf pulmonary arterial endothelial cell growth and death, are changed by MAPK inhibitors. Drug Chem Toxicol. 2011;34(1):45–52.CrossRefPubMed
36.
Zurück zum Zitat You BR, Park WH. Arsenic trioxide induces human pulmonary fibroblast cell death via increasing ROS levels and GSH depletion. Oncol Rep. 2012;28(2):749–57.CrossRefPubMed You BR, Park WH. Arsenic trioxide induces human pulmonary fibroblast cell death via increasing ROS levels and GSH depletion. Oncol Rep. 2012;28(2):749–57.CrossRefPubMed
37.
Zurück zum Zitat Cao Y, Wang J, Tian H, Fu GH. Mitochondrial ROS accumulation inhibiting JAK2/STAT3 pathway is a critical modulator of CYT997-induced autophagy and apoptosis in gastric cancer. J Exp Clin Cancer Res. 2020;39(1):119.CrossRefPubMedPubMedCentral Cao Y, Wang J, Tian H, Fu GH. Mitochondrial ROS accumulation inhibiting JAK2/STAT3 pathway is a critical modulator of CYT997-induced autophagy and apoptosis in gastric cancer. J Exp Clin Cancer Res. 2020;39(1):119.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Aggarwal V, Tuli HS, Varol A, Thakral F, Yerer MB, Sak K et al. Role of Reactive Oxygen Species in Cancer Progression: Molecular Mechanisms and Recent Advancements. Biomolecules. 2019;9(11). Aggarwal V, Tuli HS, Varol A, Thakral F, Yerer MB, Sak K et al. Role of Reactive Oxygen Species in Cancer Progression: Molecular Mechanisms and Recent Advancements. Biomolecules. 2019;9(11).
39.
Zurück zum Zitat Cottini F, Hideshima T, Suzuki R, Tai YT, Bianchini G, Richardson PG, et al. Synthetic Lethal Approaches exploiting DNA damage in aggressive myeloma. Cancer Discov. 2015;5(9):972–87.CrossRefPubMedPubMedCentral Cottini F, Hideshima T, Suzuki R, Tai YT, Bianchini G, Richardson PG, et al. Synthetic Lethal Approaches exploiting DNA damage in aggressive myeloma. Cancer Discov. 2015;5(9):972–87.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Raj L, Ide T, Gurkar AU, Foley M, Schenone M, Li X, et al. Selective killing of cancer cells by a small molecule targeting the stress response to ROS. Nature. 2011;475(7355):231–4.CrossRefPubMedPubMedCentral Raj L, Ide T, Gurkar AU, Foley M, Schenone M, Li X, et al. Selective killing of cancer cells by a small molecule targeting the stress response to ROS. Nature. 2011;475(7355):231–4.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Zheng J, Sato M, Mishima E, Sato H, Proneth B, Conrad M. Sorafenib fails to trigger ferroptosis across a wide range of cancer cell lines. Cell Death Dis. 2021;12(7):698.CrossRefPubMedPubMedCentral Zheng J, Sato M, Mishima E, Sato H, Proneth B, Conrad M. Sorafenib fails to trigger ferroptosis across a wide range of cancer cell lines. Cell Death Dis. 2021;12(7):698.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Yu Y, Xie Y, Cao L, Yang L, Yang M, Lotze MT, et al. The ferroptosis inducer erastin enhances sensitivity of acute myeloid leukemia cells to chemotherapeutic agents. Mol Cell Oncol. 2015;2(4):e1054549.CrossRefPubMedPubMedCentral Yu Y, Xie Y, Cao L, Yang L, Yang M, Lotze MT, et al. The ferroptosis inducer erastin enhances sensitivity of acute myeloid leukemia cells to chemotherapeutic agents. Mol Cell Oncol. 2015;2(4):e1054549.CrossRefPubMedPubMedCentral
Metadaten
Titel
Targeting NRF2 uncovered an intrinsic susceptibility of acute myeloid leukemia cells to ferroptosis
verfasst von
Xin Liu
Shuxin Zhong
Kangjie Qiu
Xi Chen
Weiyue Wu
Jiamian Zheng
Yanwen Liu
Haolong Wu
Shiyun Fan
Dingrui Nie
Xianfeng Wang
Zhi Yu
Ziwei Liao
Mengjun Zhong
Yangqiu Li
Chengwu Zeng
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Experimental Hematology & Oncology / Ausgabe 1/2023
Elektronische ISSN: 2162-3619
DOI
https://doi.org/10.1186/s40164-023-00411-4

Weitere Artikel der Ausgabe 1/2023

Experimental Hematology & Oncology 1/2023 Zur Ausgabe

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Perioperative Checkpointhemmer-Therapie verbessert NSCLC-Prognose

28.05.2024 NSCLC Nachrichten

Eine perioperative Therapie mit Nivolumab reduziert das Risiko für Rezidive und Todesfälle bei operablem NSCLC im Vergleich zu einer alleinigen neoadjuvanten Chemotherapie um über 40%. Darauf deuten die Resultate der Phase-3-Studie CheckMate 77T.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.