Startseite Medizin Cancer cell mitochondria: the missing puzzle in predicting response to PD-1/PD-L1 inhibitors
Artikel Open Access

Cancer cell mitochondria: the missing puzzle in predicting response to PD-1/PD-L1 inhibitors

  • Nahid Nafissi und Farzad Taghizadeh-Hesary EMAIL logo
Veröffentlicht/Copyright: 19. November 2025
Oncologie
Aus der Zeitschrift Oncologie Band 27 Heft 6

Abstract

Immunotherapy with anti-programmed cell death protein 1 (PD-1) is a promising anticancer treatment. However, only a minority of patients benefit from anti-PD-1 antibodies. Therefore, identifying the corresponding predictive factors is crucial. Emerging studies have found that the mitochondrial status of immune cells determines tumor response to anti-PD-1 antibodies. Here, we propose a novel hypothesis, based on recent experimental evidence, introducing cancer cell mitochondrial content as a new predictive factor for response to anti-PD-1 immunotherapy. A recent experiment on triple-negative breast cancer cells demonstrated that chemotherapy-induced ATPase family AAA domain–containing protein 3A (ATAD3A) expression levels regulate programmed death ligand-1 (PD-L1) on cancer cells. Accordingly, high ATAD3A expression is associated with high PD-L1 expression, poor tumor response to immunotherapy, and poor overall survival. In contrast, clinical evidence shows that high PD-L1 expression is associated with better response to immunotherapy and improved survival. This discrepancy forms the basis of our hypothesis. We propose that ATAD3A overexpression promotes poor response to anti-PD-1 therapy not by upregulating PD-L1, but by supporting mitochondrial metabolism. ATAD3A is a mitochondrial protein that maintains mitochondrial function and structure under endoplasmic reticulum stress. Therefore, mitochondrial biogenesis may serve as a novel predictive factor for response to anti-PD-(L)1 antibodies.

Introduction

Cancer remains a major global health concern [1]. Immune checkpoint inhibitors targeting the programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) axis have transformed cancer therapy. PD-L1 expression on tumor cells – either cancer cells or their surrounding supporting cells in the tumor microenvironment – and its interaction with PD-1 on recruited anti-tumoral immune cells directly impairs immune reactivation [2], 3]. Recent evidence has revealed an additional mechanism by which PD-L1 overexpression on cancer cells indirectly suppresses the anti-tumor immune response. Cheng et al. [4] found that PD-L1 upregulation in triple-negative breast cancer (TNBC) cells can further inhibit immune activity by stimulating the expression of ecto-5′-nucleotidase (NT5E) on cancer cells. NT5E (also known as CD73) inhibits anti-tumor immunity by converting adenosine monophosphate (AMP) into adenosine, a potent immunosuppressor [5].

Clinical evidence indicates that only a minority of patients benefit from PD-1/PD-L1 inhibitors (PDi). A meta-analysis in patients with TNBC reported the overall response rate of just 12 % with anti-PD-L1 monotherapy in the PD-L1–positive population [6]. Current predictive factors, including PD-L1 expression level, tumor mutation burden, T-cell infiltration rate, tumor neoantigen load, and tumor-associated macrophage infiltration, provide valuable insights but do not fully capture the complexity of treatment response [7]. Consequently, there remains a crucial need to identify and validate additional predictive biomarkers to more effectively guide the clinical application of PDi [8].

Emerging evidence highlights immune cells’ mitochondrial biogenesis as a key determinant of response to PDi, with enhanced mitochondrial activation improving treatment outcomes [9], 10]. This commentary introduces a novel perspective: the mitochondrial status of cancer cells themselves may also be a critical factor in shaping tumor response to PDi. To this end, cancer cells actively enhance their mitochondrial content – both in quality and quantity – through multiple mechanisms (outlined in the following sections), thereby promoting PD-L1 expression and improving their resistance to intrinsic anti-tumor immune response on one hand, and enhancing their resistance to PDi on the other hand. The following sections clarify these intricate mechanisms.

Subcellular PD-L1 distribution governed by ATAD3A

Experimental evidence

In an experimental study on TNBC, Xie et al. [11] demonstrated that the ATPase family AAA domain–containing protein 3A (ATAD3A)–PTEN-induced kinase 1 (PINK1)–mitophagy axis regulates the subcellular distribution of PD-L1, which in turn shapes tumor response to chemoimmunotherapy. They classified TNBC into two groups: tumors with mitochondrial PD-L1 accumulation after exposure to paclitaxel (named MITO signature), and tumors with cell membrane PD-L1 accumulation (CM signature). The MITO group exhibited greater infiltration of activated T cells (marked by IFN-γ expression), higher clinical response rates, and improved overall survival, whereas the CM group was characterized by exhausted PD-1–positive T cells, poor treatment response, and poor prognosis. Notably, MITO tumors had low ATAD3A expression, while CM tumors exhibited high ATAD3A expression. Mechanistically, high ATAD3A expression would inhibit PINK1-mediated anchoring of PD-L1 molecules to the mitochondrial membrane, resulting in PD-L1 accumulation on the cell membrane of cancer cells. While this work provides critical mechanistic insight, it does not adhere to the available clinical evidence, as outlined below.

Discrepancy with clinical evidence

Xie et al. concluded that high ATAD3A expression and high cell membrane-bound PD-L1 correlated with poor tumor response to chemoimmunotherapy [11]. This observation contrasts with findings from the phase III KEYNOTE-355 clinical trial on patients with TNBC. In that trial, patients with a combined positive score (CPS) ≥10 demonstrated longer overall survival compared to those with CPS<10 in the chemoimmunotherapy group (23 months vs. 14 months) [12]. This discrepancy suggests that the ATAD3A-PD-L1 axis alone cannot explain the improved prognosis of MITO-signature tumors to PDi, and additional parallel mechanisms may be involved. To better understand these mechanisms, it is helpful to briefly review ATAD3A and its functions in the cell.

ATAD3A as a mitochondrial integrity factor

ATAD3A is an inner mitochondrial membrane AAA+ ATPase that maintains mitochondrial DNA organization, cristae structure, and mitochondria-ER contact sites [13]. These functions preserve mitochondrial metabolism essential for cancer cell survival under endoplasmic stress [14], including endoplasmic stress led by paclitaxel [15]. Using this prelude, we propose an additional perspective of Xie et al.’s finding [11], as follows: in TNBC cells, paclitaxel exposure upregulates ATAD3A as a stress response, protecting mitochondria from damage. Therefore, tumors with high ATAD3A expression not only induce PD-L1 redistribution to the cell membrane but also preserve mitochondrial structures, providing various advantages that support cancer cells’ immune evasion, as outlined in the following section.

Mitochondria are vital for cancer cell immune evasion

Mitochondria: shared assets of cancer and immune cells

Functional mitochondria within cancer cells enhance their immune evasion capacity through several mechanisms: (I) promoting tumor microenvironment acidification via stabilization of hypoxia-inducible factor-1α (HIF-1α) and upregulation of carbonic anhydrase IX (CA-IX) [16], 17]; (II) facilitating competitive glucose uptake in cancer cells through HIF-1 stabilization, thereby limiting glucose availability for immune cell function [17], 18]; (III) suppressing major histocompatibility complex class I (MHC-I)-mediated antigen presentation through activating mitogen-activated protein kinase (MAPK) signaling pathways [19], 20]; (IV) driving T cell exhaustion through PD-L1–PD-1 signaling by impairing mitochondrial fission [21]; and (V) supporting galectin-9 expression and secretion by cancer cells, which activates immune checkpoints via T cell immunoglobulin and mucin domain-containing protein 3 (Tim3) axis [22]. On the other hand, mitochondrial activation in immune cells enhances their proliferation, memory function, cytotoxicity [23], and tumor recognition by downregulating PD-1 expression [24]. Thus, we may conclude that mitochondria are shared requirements for both cancer and immune cells, fueling a competition in which they either facilitate immune evasion or empower immunosurveillance. The following section outlines how cancer cells compete with immune cells over mitochondria.

Mitochondria-enhancing strategies of cancer cells

Cancer cells employ multiple strategies to strengthen their mitochondrial content, some in competing with immune cells (Figure 1). These mechanisms can be categorized into three groups:

  1. Receiving mitochondria from surrounding cells: cancer cells can import mitochondria from the surrounding microenvironment, including tumor-infiltrating neurons [25], neighboring tumor cells [26], and cancer-associated fibroblasts [27];

  2. Improving mitochondrial content over immune cells: In addition, cancer cells can directly hijack functional mitochondria from T cells and NK cells through well-designed tunneling nanotubes [28]. Recent evidence demonstrates that cancer cells can export their own damaged mitochondria, besides mitophagy inhibitors, to impair T cell metabolism [29]. Additional mechanisms include induction of mitochondrial dysfunction in immune cells through PD-1/PD-L1 signaling [30] or by tumor-derived exosomes [31];

  3. Enhancing mitochondrial quality: cancer cells also upregulate mitochondria-enhancing regulators such as peroxisome proliferator–activated receptor gamma coactivator 1-alpha (PGC-1α) and mitofusin to optimize mitochondrial biogenesis and functionality [32], 33].

Figure 1: 
Strategies by which cancer cells compete with tumor-infiltrating lymphocytes to enhance their own mitochondrial content while undermining that of immune cells. (A) Cancer cells can acquire mitochondria from surrounding cancer-associated fibroblasts, tumor-infiltrating nerves, and neighboring cancer cells. (B) They further increase their mitochondrial pool by hijacking mitochondria from T cells, (C) while simultaneously impairing T cell function by transferring damaged mitochondria and releasing mitophagy-inhibiting factors. In addition, (D) cancer cells can damage immune cell mitochondria indirectly via exosome secretion or (E) directly through PD-1/PD-L1 interactions. (F) They can also improve the quality of their own mitochondria through the activation of PGC-1α and mitofusin-related pathways.
Figure 1:

Strategies by which cancer cells compete with tumor-infiltrating lymphocytes to enhance their own mitochondrial content while undermining that of immune cells. (A) Cancer cells can acquire mitochondria from surrounding cancer-associated fibroblasts, tumor-infiltrating nerves, and neighboring cancer cells. (B) They further increase their mitochondrial pool by hijacking mitochondria from T cells, (C) while simultaneously impairing T cell function by transferring damaged mitochondria and releasing mitophagy-inhibiting factors. In addition, (D) cancer cells can damage immune cell mitochondria indirectly via exosome secretion or (E) directly through PD-1/PD-L1 interactions. (F) They can also improve the quality of their own mitochondria through the activation of PGC-1α and mitofusin-related pathways.

Therefore, cancer cells pursue various strategies to improve their mitochondrial content both in quality and quantity. This characteristic has been demonstrated in rapidly proliferating cancer cells, which exhibit higher mitochondrial temperatures [34], 35]. This increase in temperature likely supports their rapid growth by enhancing the mitochondrial malate–aspartate shuttle (MAS) and the glycerol 3-phosphate shuttle (G3PS) [36]. When the mitochondrial functional reserve cannot keep pace with the high proliferation rate, backup metabolic pathways are activated, ultimately leading to MAS and G3PS saturation and increased lactate production – a hallmark of the Warburg effect [37].

Conclusions

While evidence shows that ATAD3A expression is a determinant of PD-L1 redistribution, we propose that ATAD3A also contributes to immune resistance by sustaining mitochondrial metabolism and suppressing tumor immunogenicity. Evaluating cancer cell mitochondrial function may therefore complement PD-L1 scoring as a predictive biomarker and guide strategies to improve the efficacy of anti–PD-1 immunotherapy.


Corresponding author: Farzad Taghizadeh-Hesary, MD, Breast Cancer Research Center, Iran University of Medical Sciences, Tehran, 14496-14535, Iran, E-mail:

  1. Research ethics: Not applicable.

  2. Informed consent: Not applicable.

  3. Author contributions: Farzad Taghizadeh-Hesary: Conceptualization, Investigation, Writing- Original draft preparation, Writing- Reviewing and Editing; Nahid Nafissi: Supervision, Writing- Reviewing and Editing.

  4. Use of Large Language Models, AI and Machine Learning Tools: None declared.

  5. Conflict of interest: The authors declare no conflicts of interest to report regarding the present study.

  6. Research funding: The authors received no specific funding for this study.

  7. Data availability: Not applicable.

References

1. Bray, F, Laversanne, M, Sung, H, Ferlay, J, Siegel, RL, Soerjomataram, I, et al.. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2024;74:229–63. https://doi.org/10.3322/caac.21834.Suche in Google Scholar PubMed

2. Kawasaki, K, Noma, K, Kato, T, Ohara, T, Tanabe, S, Takeda, Y, et al.. PD-L1-expressing cancer-associated fibroblasts induce tumor immunosuppression and contribute to poor clinical outcome in esophageal cancer. Cancer Immunol Immunother 2023;72:3787–802. https://doi.org/10.1007/s00262-023-03531-2.Suche in Google Scholar PubMed PubMed Central

3. Wang, L, Han, H, Ma, J, Feng, Y, Han, Z, Maharaj, V, et al.. Identification of hypoxia-immune-related signatures for predicting immune efficacy in triple-negative breast cancer. Oncologie 2024;26:433–44. https://doi.org/10.1515/oncologie-2023-0539.Suche in Google Scholar

4. Cheng, C, Shi, C, Wu, S, Wu, W, Li, J, Gao, S, et al.. Tumor-expressing PD-L1 regulates NT5E expression through MAPK/ERK pathway in triple-negative breast cancer. Oncol Res 2025;33:1633–48. https://doi.org/10.32604/or.2025.061637.Suche in Google Scholar PubMed PubMed Central

5. Zhang, C, Wang, K, Wang, H. Adenosine in cancer immunotherapy: taking off on a new plane. Biochim Biophys Acta Rev Cancer 2023;1878:189005. https://doi.org/10.1016/j.bbcan.2023.189005.Suche in Google Scholar PubMed

6. Xiao, BY, Lin, GH, Zhao, YX, Wang, BC. The efficacy and safety of PD-1/PD-L1 inhibitors in breast cancer: a systematic review and meta-analysis. Transl Cancer Res 2020;9:3804–18. https://doi.org/10.21037/tcr-19-3020.Suche in Google Scholar PubMed PubMed Central

7. Mao, Y, Xie, H, Lv, M, Yang, Q, Shuang, Z, Gao, F, et al.. The landscape of objective response rate of anti-PD-1/L1 monotherapy across 31 types of cancer: a system review and novel biomarker investigating. Cancer Immunol Immunother 2023;72:2483–98. https://doi.org/10.1007/s00262-023-03441-3.Suche in Google Scholar PubMed PubMed Central

8. Bie, F, Tian, H, Sun, N, Zang, R, Zhang, M, Song, P, et al.. Research progress of Anti-PD-1/PD-L1 immunotherapy related mechanisms and predictive biomarkers in NSCLC. Front Oncol 2022;12:769124. https://doi.org/10.3389/fonc.2022.769124.Suche in Google Scholar PubMed PubMed Central

9. Chamoto, K, Chowdhury, PS, Kumar, A, Sonomura, K, Matsuda, F, Fagarasan, S, et al.. Mitochondrial activation chemicals synergize with surface receptor PD-1 blockade for T cell-dependent antitumor activity. Proc Natl Acad Sci U S A 2017;114:E761–E770. https://doi.org/10.1073/pnas.1620433114.Suche in Google Scholar PubMed PubMed Central

10. Houshyari, M, Taghizadeh-Hesary, F. Is mitochondrial metabolism a new predictive biomarker for antiprogrammed cell death Protein-1 immunotherapy? JCO Oncol Pract 2023;19:123–4. https://doi.org/10.1200/op.22.00733.Suche in Google Scholar PubMed

11. Xie, XQ, Yang, Y, Wang, Q, Liu, HF, Fang, XY, Li, CL, et al.. Targeting ATAD3A-PINK1-mitophagy axis overcomes chemoimmunotherapy resistance by redirecting PD-L1 to mitochondria. Cell Res 2023;33:215–28. https://doi.org/10.1038/s41422-022-00766-z.Suche in Google Scholar PubMed PubMed Central

12. Cortes, J, Cescon, DW, Rugo, HS, Nowecki, Z, Im, SA, Yusof, MM, et al.. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet 2020;396:1817–28.10.1016/S0140-6736(20)32531-9Suche in Google Scholar PubMed

13. Chen, L, Li, Y, Zambidis, A, Papadopoulos, V. ATAD3A: a key regulator of mitochondria-associated diseases. Int J Mol Sci 2023;24:12511. https://doi.org/10.3390/ijms241512511.Suche in Google Scholar PubMed PubMed Central

14. Brar, KK, Hughes, DT, Morris, JL, Subramanian, K, Krishna, S, Gao, F, et al.. PERK-ATAD3A interaction provides a subcellular safe haven for protein synthesis during ER stress. Science 2024;385:eadp7114. https://doi.org/10.1126/science.adp7114.Suche in Google Scholar PubMed

15. Wang, J, Fan, P, Shen, P, Fan, C, Zhao, P, Yao, S, et al.. XBP1s activates METTL3/METTL14 for ER-phagy and paclitaxel sensitivity regulation in breast cancer. Cancer Lett 2024;596:216846. https://doi.org/10.1016/j.canlet.2024.216846.Suche in Google Scholar PubMed

16. Chen, S, Wu, Z. Targeting tumor microenvironments with gold nanoparticles for enhanced photothermal therapy. Oncologie 2024;26:899–912. https://doi.org/10.1515/oncologie-2024-0320.Suche in Google Scholar

17. Van Gisbergen, MW, Offermans, K, Voets, AM, Lieuwes, NG, Biemans, R, Hoffmann, RF, et al.. Mitochondrial dysfunction inhibits hypoxia-induced HIF-1α stabilization and expression of its downstream targets. Front Oncol 2020;10:770. https://doi.org/10.3389/fonc.2020.00770.Suche in Google Scholar PubMed PubMed Central

18. Chen, C, Pore, N, Behrooz, A, Ismail-Beigi, F, Maity, A. Regulation of glut1 mRNA by hypoxia-inducible Factor-1. J Biol Chem 2001;276:9519–25. https://doi.org/10.1074/jbc.m010144200.Suche in Google Scholar

19. Inoue, M, Mimura, K, Izawa, S, Shiraishi, K, Inoue, A, Shiba, S, et al.. Expression of MHC class I on breast cancer cells correlates inversely with HER2 expression. OncoImmunology 2012;1:1104–10. https://doi.org/10.4161/onci.21056.Suche in Google Scholar PubMed PubMed Central

20. Mandal, JP, Shiue, CN, Chen, YC, Lee, MC, Yang, HH, Chang, HH, et al.. PKCδ mediates mitochondrial ROS generation and oxidation of HSP60 to relieve RKIP inhibition on MAPK pathway for HCC progression. Free Radic Biol Med 2021;163:69–87. https://doi.org/10.1016/j.freeradbiomed.2020.12.003.Suche in Google Scholar PubMed

21. Simula, L, Antonucci, Y, Scarpelli, G, Cancila, V, Colamatteo, A, Manni, S, et al.. PD-1-induced T cell exhaustion is controlled by a Drp1-dependent mechanism. Mol Oncol 2022;16:188–205. https://doi.org/10.1002/1878-0261.13103.Suche in Google Scholar PubMed PubMed Central

22. Sakhnevych, SS, Yasinska, IM, Fasler-Kan, E, Sumbayev, VV. Mitochondrial defunctionalization supresses Tim-3-Galectin-9 secretory pathway in human colorectal cancer cells and thus can possibly affect tumor immune escape. Front Pharmacol 2019;10:342. https://doi.org/10.3389/fphar.2019.00342.Suche in Google Scholar PubMed PubMed Central

23. Desdín-Micó, G, Soto-Heredero, G, Mittelbrunn, M. Mitochondrial activity in T cells. Mitochondrion 2018;41:51–7. https://doi.org/10.1016/j.mito.2017.10.006.Suche in Google Scholar PubMed

24. Wu, H, Zhao, X, Hochrein, SM, Eckstein, M, Gubert, GF, Knöpper, K, et al.. Mitochondrial dysfunction promotes the transition of precursor to terminally exhausted T cells through HIF-1α-mediated glycolytic reprogramming. Nat Commun 2023;14:6858. https://doi.org/10.1038/s41467-023-42634-3.Suche in Google Scholar PubMed PubMed Central

25. Hoover, G, Gilbert, S, Curley, O, Obellianne, C, Lin, MT, Hixson, W, et al.. Nerve-to-cancer transfer of mitochondria during cancer metastasis. Nature 2025;644:252–62. https://doi.org/10.1038/s41586-025-09176-8.Suche in Google Scholar PubMed PubMed Central

26. Lu, J, Zheng, X, Li, F, Yu, Y, Chen, Z, Liu, Z, et al.. Tunneling nanotubes promote intercellular mitochondria transfer followed by increased invasiveness in bladder cancer cells. Oncotarget 2017;8:15539–52. https://doi.org/10.18632/oncotarget.14695.Suche in Google Scholar PubMed PubMed Central

27. Goliwas, KF, Libring, S, Berestesky, E, Gholizadeh, S, Schwager, SC, Frost, AR, et al.. Mitochondrial transfer from cancer-associated fibroblasts increases migration in aggressive breast cancer. J Cell Sci 2023;136:jcs260419. https://doi.org/10.1242/jcs.260419.Suche in Google Scholar PubMed PubMed Central

28. Saha, T, Dash, C, Jayabalan, R, Khiste, S, Kulkarni, A, Kurmi, K, et al.. Intercellular nanotubes mediate mitochondrial trafficking between cancer and immune cells. Nat Nanotechnol 2022;17:98–106. https://doi.org/10.1038/s41565-021-01000-4.Suche in Google Scholar PubMed PubMed Central

29. Ikeda, H, Kawase, K, Nishi, T, Watanabe, T, Takenaga, K, Inozume, T, et al.. Immune evasion through mitochondrial transfer in the tumour microenvironment. Nature 2025;638:225–36. https://doi.org/10.1038/s41586-024-08439-0.Suche in Google Scholar PubMed PubMed Central

30. Ogando, J, Sáez, ME, Santos, J, Nuevo-Tapioles, C, Gut, M, Esteve-Codina, A, et al.. PD-1 signaling affects cristae morphology and leads to mitochondrial dysfunction in human CD8+ T lymphocytes. J Immunother Cancer 2019;7:151. https://doi.org/10.1186/s40425-019-0628-7.Suche in Google Scholar PubMed PubMed Central

31. Mondal, SK, Haas, D, Han, J, Whiteside, TL. Small EV in plasma of triple negative breast cancer patients induce intrinsic apoptosis in activated T cells. Commun Biol 2023;6:815. https://doi.org/10.1038/s42003-023-05169-3.Suche in Google Scholar PubMed PubMed Central

32. Andrzejewski, S, Klimcakova, E, Johnson, RM, Tabariès, S, Annis, MG, McGuirk, S, et al.. PGC-1α promotes breast cancer metastasis and confers bioenergetic flexibility against metabolic drugs. Cell Metab 2017;26:778–87.e5. https://doi.org/10.1016/j.cmet.2017.09.006.Suche in Google Scholar PubMed

33. Purohit, PK, Edwards, R, Tokatlidis, K, Saini, N. MiR-195 regulates mitochondrial function by targeting mitofusin-2 in breast cancer cells. RNA Biol 2019;16:918–29. https://doi.org/10.1080/15476286.2019.1600999.Suche in Google Scholar PubMed PubMed Central

34. Zhang, X, Hu, Y. Mitochondrial thermogenesis in cancer cells. Oncologie 2023;25:591–603. https://doi.org/10.1515/oncologie-2023-0298.Suche in Google Scholar

35. Zhu, Q, Sun, Y, Fu, M, Bian, M, Zhu, X, Wang, K, et al.. Ultrasensitive small-molecule fluorescent thermometer reveals hot mitochondria in surgically resected human tumors. ACS Sens 2023;8:51–60. https://doi.org/10.1021/acssensors.2c01563.Suche in Google Scholar PubMed

36. Wang, Y, Stancliffe, E, Fowle-Grider, R, Wang, R, Wang, C, Schwaiger-Haber, M, et al.. Saturation of the mitochondrial NADH shuttles drives aerobic glycolysis in proliferating cells. Mol Cell 2022;82:3270–83.e9. https://doi.org/10.1016/j.molcel.2022.07.007.Suche in Google Scholar PubMed PubMed Central

37. Jaiswal, A, Singh, R. A negative feedback loop underlies the Warburg effect. npj Syst Biol Appl 2024;10:55. https://doi.org/10.1038/s41540-024-00377-x.Suche in Google Scholar PubMed PubMed Central

Received: 2025-09-26
Accepted: 2025-11-06
Published Online: 2025-11-19

© 2025 the author(s), published by De Gruyter on behalf of Tech Science Press (TSP)

This work is licensed under the Creative Commons Attribution 4.0 International License.

Artikel in diesem Heft

  1. Frontmatter
  2. Review Articles
  3. Liquid biopsy – a promising and effective method for surveying non-small cell lung cancer minimal residual diseases and anti-cancer drug response after treatment
  4. Current application status of proton beam therapy for gastrointestinal tumors
  5. Research progress on the regulation of cuproptosis-related genes by non-coding RNAs in tumors
  6. Deep learning in hepatic oncology imaging: a narrative review of computed tomography applications
  7. Synergistic approaches: a narrative mini-review of radiotherapy and immunotherapy in the treatment of lung cancer
  8. Research Articles
  9. Intravesical prostatic protrusion as a predictor of acute urinary retention following stereotactic body radiation therapy for localised prostate cancer: a retrospective study
  10. The differential effect of glutamine supplementation on the orthotopic and subcutaneous growth of two syngeneic murine models of glioma
  11. Intermittent afatinib treatment suppresses the growth of resistant T790M-H1975 cells in non-small cell lung cancer (NSCLC) co-culture
  12. Prognostic stratification of colorectal cancer by immune profiling reveals SPP1 as a key indicator for tumor immune status
  13. The activity of base excision repair is positively correlated with the infiltration of CD4+ T cells in melanoma
  14. Integrated analysis of immunity and ferroptosis related tumor microenvironment in a novel risk score model for lung adenocarcinoma prognosis
  15. Retrospective analysis of risk factors for early recurrence after hepatocellular carcinoma resection
  16. The ENST00000539930 transcript predicts sensitivity to PARP inhibitors and clinical prognosis in cancers
  17. VTA1 and breast cancer: a potential indicator for diagnostic and prognostic evaluation
  18. USP24 stabilizes VDAC2 via deubiquitination to promote apoptosis and ferroptosis in clear cell renal cell carcinoma (ccRCC)
  19. Clinicopathological characteristics, prognosis, and therapeutic implications in breast cancer with pathologically confirmed bone marrow metastases: an observational retrospective study
  20. Short Commentaries
  21. Cancer cell mitochondria: the missing puzzle in predicting response to PD-1/PD-L1 inhibitors
  22. From mitochondrial cristae pathobiology to metabolic reprogramming in cancer: the α and ω of Malignancies?
  23. Article Commentary
  24. Stopping SOAT1 sparks an immune attack on liver cancer: a metabolic-immune axis in hepatocellular carcinoma
Heruntergeladen am 21.12.2025 von https://www.degruyterbrill.com/document/doi/10.1515/oncologie-2025-0420/html
Button zum nach oben scrollen