Advances in Clinical and Experimental Medicine

Title abbreviation: Adv Clin Exp Med
5-Year IF – 2.0, IF – 1.9, JCI (2024) – 0.43
Scopus CiteScore – 4.3
Q1 in SJR 2024, SJR score – 0.598, H-index: 49 (SJR)
IC – 171.00; MNiSW – 70 pts
Initial editorial assessment and first decision within 24 h

ISSN 1899–5276 (print), ISSN 2451-2680 (online)
Periodicity – monthly

Download original text (EN)

Advances in Clinical and Experimental Medicine

2025, vol. 34, nr 5, May, p. 669–671

doi: 10.17219/acem/205024

Publication type: editorial

Language: English

License: Creative Commons Attribution 3.0 Unported (CC BY 3.0)

Download citation:

  • BIBTEX (JabRef, Mendeley)
  • RIS (Papers, Reference Manager, RefWorks, Zotero)

Cite as:


Zhang H. Revolutionizing cancer treatment: Navigating the intricate landscape of cellular signaling networks. Adv Clin Exp Med. 2025;34(5):669–671. doi:10.17219/acem/205024

Revolutionizing cancer treatment: Navigating the intricate landscape of cellular signaling networks

Hao Zhang1,A,B,C,D,E,F

1 Department of Neurosurgery, Second Affiliated Hospital, Chongqing Medical University, China

Graphical abstract


Graphical abstracts

Highlights


• Cancer-signaling networks and therapy resistance: Mapping pathway redundancy and tumor-microenvironment cross-talk pinpoints how cancers evade targeted treatments.
• Multi-omics + AI + spatial biology: Integrating genomics, proteomics, and deep-learning analytics deciphers tumor heterogeneity and predicts patient-specific drug response.
• Organoid and 3D-bioprinted tumor models: Next-gen preclinical platforms deliver physiologic testing grounds for network-targeted therapies – accelerating bench-to-bedside translation.
• Non-coding RNAs, epigenetic reprogramming, and TME dynamics: Emerging regulators that drive metastasis and therapy escape reveal fresh intervention points.
• Systems-biology and computational oncology convergence: Interdisciplinary modeling designs adaptive, precision cancer treatments that keep pace with tumor evolution.

Abstract

Cancer progression and therapeutic resistance are propelled by the remarkable plasticity of signaling networks, which dynamically rewire under selective pressures to maintain proliferation, enable immune evasion and promote metastasis. Despite advances in precision oncology, the dynamic crosstalk between tumor cells, non-coding genomes and the microenvironment continues to undermine treatment efficacy. This call for submissions, Revolutionizing Cancer Treatment: Navigating the Intricate Landscape of Cellular Signaling Networks, seeks cutting-edge research that dissects these adaptive mechanisms through innovative technologies – from single-cell multi-omics and spatial transcriptomics to AI-powered network modeling. We welcome studies leveraging physiomimetic models (e.g., organoids, 3D-bioprinted ecosystems) to decode tumor heterogeneity, as well as translational work targeting emergent vulnerabilities at the intersection of epigenetics, metabolic reprogramming and stromal interactions. By integrating systems biology with computational and experimental approaches, this collection aims to catalyze the design of adaptive therapies that outmaneuver cancer’s evolutionary resilience.

Key words: cancer treatment, cellular signaling networks, biomarkers

Introduction

Cancer, a disease of profound complexity, arises from the dysregulation of cellular signaling networks that orchestrate cell fate, immune evasion and metastatic dissemination.1 While chemotherapy, immunotherapy and targeted therapies have transformed oncology, the adaptability of tumors – fueled by redundant pathways, microenvironmental interactions and evolutionary pressures – remains a formidable barrier. This call for submissions, Revolutionizing Cancer Treatment: Navigating the Intricate Landscape of Cellular Signaling Networks, invites the scientific community to confront the complex challenges of cancer research through a systems-level lens – bridging molecular discovery with translational innovation.

The double-edged sword of signaling complexity: From pathways to networks

Carcinogenesis is not merely the result of driver mutations, but rather a systemic collapse of signaling homeostasis. Canonical pathways such as PI3K/AKT/mTOR,2 RAS/MAPK,3 and Wnt/β-catenin4 are frequently co-opted by tumors; however, their extensive crosstalk and functional redundancy often create escape routes that undermine therapeutic efficacy. For example, BRAF (B-Raf proto-oncogene, serine/threonine kinase) inhibitors in melanoma initially shrink tumors, only to see resistance emerge via NRAS (neuroblastoma RAS viral oncogene homolog) mutations or EGFR (epidermal growth factor receptor)-driven rewiring.5 Similarly, EGFR-targeted therapies in lung cancer fail when tumors activate AXL (AXL receptor tyrosine kinase) or MET (MET proto-oncogene, receptor tyrosine kinase) signaling.6, 7 Even immunotherapy, which reinvigorates cytotoxic T cells by blocking PD-1 (programmed cell death protein 1)/PD-L1 (programmed death-ligand 1), is thwarted by compensatory immunosuppressive networks involving TGF-β (transforming growth factor beta), adenosine or regulatory T cells within the tumor microenvironment (TME).8 The TME itself acts as a dynamic signaling hub. Cancer-associated fibroblasts (CAFs) secrete growth factors like FGF (fibroblast growth factor) and VEGF (vascular endothelial growth factor),9 while tumor-associated macrophages (TAMs) release interleukin 10 (IL-10) and CCL22,10 creating a pro-metastatic niche. Recent studies reveal that extracellular vesicles (EVs) shuttle oncogenic miRNAs (e.g., miR-122) between tumor and stromal cells, further entrenching resistance.11 These interactions underscore the need to map signaling networks as adaptive circuits rather than static pathways.

Bridging the gap: From multi-omic insights to AI, spatial biology and beyond

Advances in multi-omics – proteogenomics,12 spatial transcriptomics13 and metabolomics14 – have unmasked tumor heterogeneity and plasticity.15 Single-cell RNA sequencing has identified rare subpopulations with enhanced tumor metastasis ability.16 Furthermore, computational models now simulate network responses to perturbations, predicting resistance mechanisms and optimal drug sequences. Artificial intelligence (AI) and machine learning (ML) are accelerating discoveries. For instance, the deep learning model identified 8 core protein assemblies integrating multi-gene alterations to predict palbociclib response in breast cancer, outperforming single-gene biomarkers.17 Spatial transcriptomics, spatial proteomics and computational approaches have been combined to reveal that gliomas organize into spatially structured cellular states, with local microenvironments dominated by single states and specific state pairs consistently co-localizing across tumors. Hypoxia emerges as a key driver of long-range tissue architecture, shaping a layered global organization absent in non-hypoxic tumors like low-grade IDH-mutant gliomas.18 Patient-derived organoids19 and three-domesnional (3D) bioprinted TME20 models are also revolutionizing preclinical testing. These platforms recapitulate stromal interactions and drug penetration barriers, enabling high-throughput screening of network-targeted therapies.

Call for contributions

We invite contributions that explore the following emerging and advanced areas of cancer biology and therapy:

Novel regulators of oncogenic hubs: Non-coding RNAs, phase-separated condensates and post-translational modifiers (e.g., ubiquitin ligases).

TME-mediated network modulation: Role of exosomes, circadian rhythm disruptions and neural signaling in metastasis.

Resistance mechanisms: Epigenetic plasticity, adaptive kinome reprogramming and persister cell states.

AI/ML-driven discovery: Network-based drug repurposing and digital twins for personalized therapy.

Biomarkers: Circulating tumor DNA (ctDNA) for real-time network monitoring, and metabolic imaging signatures.

Conclusions

The future of cancer therapy lies in decoding the rules that govern signaling networks – not just the molecules. By integrating systems biology, AI, and spatially resolved technologies, we can design therapies that anticipate and disrupt cancer’s adaptive strategies. This call for submissions seeks to catalyze interdisciplinary collaboration, uniting basic researchers, computational biologists and clinicians to transform complexity into curability.

References (20)

  1. Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. Cell. 2011;144(5):646–674. doi:10.1016/j.cell.2011.02.013
  2. Glaviano A, Foo ASC, Lam HY, et al. PI3K/AKT/mTOR signaling transduction pathway and targeted therapies in cancer. Mol Cancer. 2023;22(1):138. doi:10.1186/s12943-023-01827-6
  3. Bahar ME, Kim HJ, Kim DR. Targeting the RAS/RAF/MAPK pathway for cancer therapy: From mechanism to clinical studies. Sig Transduct Target Ther. 2023;8(1):455. doi:10.1038/s41392-023-01705-z
  4. Liu J, Xiao Q, Xiao J, et al. Wnt/β-catenin signalling: Function, biological mechanisms, and therapeutic opportunities. Sig Transduct Target Ther. 2022;7(1):3. doi:10.1038/s41392-021-00762-6
  5. Nazarian R, Shi H, Wang Q, et al. Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature. 2010;468(7326):973–977. doi:10.1038/nature09626
  6. Engelman JA, Zejnullahu K, Mitsudomi T, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316(5827):1039–1043. doi:10.1126/science.1141478
  7. Zhang Z, Lee JC, Lin L, et al. Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat Genet. 2012;44(8):852–860. doi:10.1038/ng.2330
  8. Binnewies M, Roberts EW, Kersten K, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018;24(5):541–550. doi:10.1038/s41591-018-0014-x
  9. Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer. 2016;16(9):582–598. doi:10.1038/nrc.2016.73
  10. Mantovani A, Marchesi F, Malesci A, Laghi L, Allavena P. Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol. 2017;14(7):399–416. doi:10.1038/nrclinonc.2016.217
  11. Fong MY, Zhou W, Liu L, et al. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat Cell Biol. 2015;17(2):183–194. doi:10.1038/ncb3094
  12. Petralia F, Ma W, Yaron TM, et al. Pan-cancer proteogenomics characterization of tumor immunity. Cell. 2024;187(5):1255–1277.e27. doi:10.1016/j.cell.2024.01.027
  13. Jin Y, Zuo Y, Li G, et al. Advances in spatial transcriptomics and its applications in cancer research. Mol Cancer. 2024;23(1):129. doi:10.1186/s12943-024-02040-9
  14. Kumar A, Misra BB. Challenges and opportunities in cancer metabolomics. Proteomics. 2019;19(21–22):1900042. doi:10.1002/pmic.201900042
  15. Sanchez-Vega F, Mina M, Armenia J, et al. Oncogenic signaling pathways in The Cancer Genome Atlas. Cell. 2018;173(2):321–337.e10. doi:10.1016/j.cell.2018.03.035
  16. Puram SV, Tirosh I, Parikh AS, et al. Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell. 2017;171(7):1611–1624.e24. doi:10.1016/j.cell.2017.10.044
  17. Park S, Silva E, Singhal A, et al. A deep learning model of tumor cell architecture elucidates response and resistance to CDK4/6 inhibitors. Nat Cancer. 2024;5(7):996–1009. doi:10.1038/s43018-024-00740-1
  18. Greenwald AC, Darnell NG, Hoefflin R, et al. Integrative spatial analysis reveals a multi-layered organization of glioblastoma. Cell. 2024;187(10):2485–2501.e26. doi:10.1016/j.cell.2024.03.029
  19. Driehuis E, Kretzschmar K, Clevers H. Establishment of patient-derived cancer organoids for drug-screening applications. Nat Protoc. 2020;15(10):3380–3409. doi:10.1038/s41596-020-0379-4
  20. Shukla P, Yeleswarapu S, Heinrich MA, Prakash J, Pati F. Mimicking tumor microenvironment by 3D bioprinting: 3D cancer modeling. Biofabrication. 2022;14(3):032002. doi:10.1088/1758-5090/ac6d11