You are currently on the new version of our website. Access the old version .
Applied SciencesApplied Sciences
  • Editorial
  • Open Access

12 January 2026

Emerging Topics in Precision Medicine: Non-Invasive Innovations Shaping Cancer and Immunotherapy Progress

and
1
Department of Biochemistry, University of Nebraska, Lincoln, NE 68503, USA
2
Cancer Digital Twins, San Mateo, CA 94001, USA
*
Author to whom correspondence should be addressed.
This article belongs to the Special Issue Emerging Topics in Precision Medicine: Non-invasive Innovations Shaping Cancer and Immunotherapy Progress

1. Introduction

Cancer is a heterogeneous disease, resulting from genetic, epigenetic, signaling, and metabolic alterations, which are supported by a highly organized tumor microenvironment (TME) [1]. The dynamic interplay between cancer and TME allows tumors to adapt, evade therapy, and develop resistance, making treatment a persistent challenge [2]. While next-generation sequencing has yielded critical genetic insights, it has become increasingly evident that single-marker analyses and one-size-fits-all treatment approaches are insufficient to capture the multiscale complexity of cancer biology [3].
Conventional diagnostic approaches often rely on invasive and temporally limited tissue sampling, which restricts longitudinal monitoring and increases patient burden [4]. For instance, tissue biopsies offer only a static snapshot of a highly dynamic disease, are vulnerable to sampling bias driven by intratumoral heterogeneity, and are frequently impractical to repeat throughout the course of treatment [5]. Consequently, key biological shifts associated with tumor evolution, therapeutic pressure, and the emergence of resistance may go undetected until overt clinical progression occurs. These constraints limit real-time clinical decision-making and hinder the timely adaptation of therapeutic strategies to the evolving state of tumors. Due to these limitations, the oncology landscape has evolved to accelerate the development of minimally invasive and non-invasive methods [6]. The patient-derived data are increasingly leveraged through the integration of multimodal biomarkers, state-of-the-art imaging, and mechanistic as well as AI-driven computational models to uncover tumor complexity and guide precision therapies [7].
The first edition of this research topic, entitled “Emerging Topics in Precision Medicine: Non-invasive Innovations Shaping Cancer and Immunotherapy Progress,” collectively illustrates the ongoing shift toward integrative and non-invasive strategies in precision oncology. Rather than addressing isolated molecular events, the studies focus on the need to understand cancer as a dynamic system shaped by tumor-intrinsic programs, immune and stromal interactions, metabolic states, and therapeutic pressures.

2. An Overview of Published Articles

2.1. Non-Invasive Approaches

Cancer biomarkers have expanded beyond static genetic alterations to encompass functional, spatial, and systemic indicators that better reflect tumor dynamics and adaptability. Biomarkers are grouped as diagnostic, prognostic, predictive, and pharmacodynamic categories, reflecting their clinical utility rather than the underlying biology [8]. Diagnostic biomarkers enable disease detection and classification, prognostic biomarkers provide information on disease outcome independent of treatment, predictive biomarkers identify patients most likely to benefit from a specific therapy, and pharmacodynamic biomarkers monitor treatment response and target engagement [9]. However, this functional classification oversimplifies tumor biology, as many biomarkers participate in interconnected signaling, metabolic, and microenvironmental processes and may simultaneously fulfill multiple roles depending on context [10]. Three reviews in the present collection highlight how immune cell plasticity, stromal support, hypoxia, metabolism, and inflammatory signaling collectively shape tumor progression and therapeutic resistance. By emphasizing tumor–microenvironment crosstalk, these contributions reinforce the notion that durable clinical benefit will require strategies that disrupt not only malignant cells but also the supportive niches that sustain them [11].
To address this, the growing availability of large-scale data repositories enables proof-of-concept identification of novel biomarkers across spatial and temporal scales [12]. This is particularly important in accelerating early discovery, as the path for clinical approval remains long and complex [13]. Complementing this, in vitro experiments using 2D and 3D models support the mechanistic validation of candidate biomarkers, interrogation of signaling pathways, and assessment of drug sensitivity under defined conditions [14]. Together, in silico and in vitro strategies create an iterative pipeline improving the identification of promising biomarkers for further development [8,15].
Moreover, advanced imaging with AI further expanded the biomarker landscape beyond classical molecular assays [16]. Radiomics extracts high-dimensional quantitative features from medical imaging, capturing spatial heterogeneity, tumor shape, and texture that are not discernible by visual inspection [17]. In the present collection, combining artificial intelligence with PET/CT radiomics predicts immunotherapy response and tumor grade in advanced cutaneous squamous cell carcinoma. Additional molecular layers can also be integrated into imaging; for example, radiogenomics combines imaging-derived features with genomic and transcriptomic data, enabling non-invasive inference of molecular phenotypes and tumor evolution [18]. Longitudinal imaging monitoring is a powerful tool for patient stratification and predicting therapeutic response, capturing temporal changes in tumor size, shape, texture, and functional parameters [19].
Collectively, the integration of molecular biomarkers with radiomic, in silico, and in vitro approaches supports a more holistic, multiscale view of tumor biology. This convergence moves biomarker research from single-layer checkpoints toward dynamic indicators of disease state and therapeutic vulnerability, laying the groundwork for more precise and adaptive precision oncology strategies.

2.2. Minimally Invasive Approaches

Traditional diagnostic approaches, such as biopsies, are the gold standard for initial diagnosis, offering deeper tissue context [20]. However, these methods are invasive, risky, and provide a static snapshot of the tumor site, with results often influencing initial treatment decisions. While liquid biopsies (blood/fluid test) are minimally invasive, providing real-time monitoring of tumors, but they can miss rare mutations. While both can be complementary, liquid biopsies are increasingly transitioning from experimental tools to clinically relevant technologies, offering real-time, high-resolution insights into tumor burden, immune dynamics, and disease evolution [21]. Together, these innovations are reshaping cancer management toward more adaptive and patient-centered care [22].
A prominent theme across the collected works is the increasing relevance of minimally invasive biomarkers. Blood-based analytes, immune cell dynamics, and circulating tumor-derived signals are highlighted as powerful tools for assessing tumor burden, biological aggressiveness, and treatment response. For example, analyses of circulating cell-free DNA and immune cell populations were used to monitor tumor size and advanced pathological stages, while also providing actionable insights into immunotherapy response [23,24]. These approaches highlight the additional framework of liquid biopsy to complement or, in selected contexts, partially substitute tissue-based diagnostics, particularly for longitudinal disease monitoring.

3. Conclusions and Future Perspectives

The articles in this Special Issue reflect a maturation of the precision oncology field, moving beyond reductionist models toward systems-level perspectives that integrate molecular, cellular, and environmental dimensions of cancer biology. To translate these advances into durable clinical benefit, future research must prioritize longitudinal monitoring, integrative analytical frameworks, and rational combination strategies that account for tumor heterogeneity and adaptive resistance. By embracing complexity rather than attempting to oversimplify it, the field is better positioned to deliver truly personalized and effective cancer care.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Arora, H.L.; Sekar, G.; Phadnis, A.; Bahot, A.; Bomle, D.; Patel, V.; Pal, J.K.; Sarode, S.C.; Sharma, N.K. Emerging hallmarks and the rise of complexities and heterogeneity of tumor. Biochem. Biophys. Rep. 2025, 44, 102347. [Google Scholar] [CrossRef] [PubMed]
  2. Biray Avci, C.; Goker Bagca, B.; Nikanfar, M.; Takanlou, L.S.; Takanlou, M.S.; Nourazarian, A. Tumor microenvironment and cancer metastasis: Molecular mechanisms and therapeutic implications. Front. Pharmacol. 2024, 15, 1442888. [Google Scholar] [CrossRef]
  3. Zou, Y.; Zhao, Z.; Song, Y. An overview of multiomics: A powerful tool applied in cancer molecular subtyping for cancer therapy. Malig. Spectr. 2024, 1, 15–29. [Google Scholar] [CrossRef]
  4. Pashayan, N.; Pharoah, P.D.P. The challenge of early detection in cancer. Science 2020, 368, 589–590. [Google Scholar] [CrossRef]
  5. Neagu, A.-N.; Bruno, P.S.; Josan, C.-L.; Waterman, N.; Morrissiey, H.; Njoku, V.T.; Darie, C.C. In search of ideal solutions for cancer diagnosis: From conventional methods to protein biomarkers in liquid biopsy. Proteomes 2025, 13, 47. [Google Scholar] [CrossRef]
  6. Qiao, D.; Wang, R.C.; Wang, Z. Precision oncology: Current landscape, emerging trends, challenges, and future perspectives. Cells 2025, 14, 1804. [Google Scholar] [CrossRef]
  7. Puniya, B.L. Artificial-intelligence-driven Innovations in Mechanistic Computational Modeling and Digital Twins for Biomedical Applications. J. Mol. Biol. 2025, 437, 169181. [Google Scholar] [CrossRef]
  8. Aghamiri, S.S.; Amin, R. Digital Twin-Based Multiscale Models for Biomarker Discovery in Kinase and Phosphatase Tumorigenic Processes. Kinases Phosphatases 2025, 3, 18. [Google Scholar] [CrossRef]
  9. Carlomagno, N.; Incollingo, P.; Tammaro, V.; Peluso, G.; Rupealta, N.; Chiacchio, G.; Sandoval Sotelo, M.L.; Minieri, G.; Pisani, A.; Riccio, E.; et al. Diagnostic, predictive, prognostic, and therapeutic molecular biomarkers in third millennium: A breakthrough in gastric cancer. Biomed Res. Int. 2017, 2017, 7869802. [Google Scholar] [CrossRef] [PubMed]
  10. Dabbagh Moghaddam, F.; Anvar, A.; Ilkhani, E.; Dadgar, D.; Rafiee, M.; Ranjbaran, N.; Mortazavi, P.; Ghoreishian, S.M.; Huh, Y.S.; Makvandi, P. Advances in engineering immune-tumor microenvironments on-a-chip: Integrative microfluidic platforms for immunotherapy and drug discovery. Mol. Cancer 2025, 24, 271. [Google Scholar] [CrossRef]
  11. Roma-Rodrigues, C.; Mendes, R.; Baptista, P.V.; Fernandes, A.R. Targeting tumor microenvironment for cancer therapy. Int. J. Mol. Sci. 2019, 20, 840. [Google Scholar] [CrossRef] [PubMed]
  12. Jeong, E.; Yoon, S. Current advances in comprehensive omics data mining for oncology and cancer research. Biochim. Biophys. Acta Rev. Cancer 2024, 1879, 189030. [Google Scholar] [CrossRef] [PubMed]
  13. Steyaert, S.; Pizurica, M.; Nagaraj, D.; Khandelwal, P.; Hernandez-Boussard, T.; Gentles, A.J.; Gevaert, O. Multimodal data fusion for cancer biomarker discovery with deep learning. Nat. Mach. Intell. 2023, 5, 351–362. [Google Scholar] [CrossRef]
  14. Guerrero-López, P.; Martín-Pardillos, A.; Bonet-Aleta, J.; Mosseri, A.; Hueso, J.L.; Santamaria, J.; Garcia-Aznar, J.M. 2D versus 3D tumor-on-chip models to study the impact of tumor organization on metabolic patterns in vitro. Sci. Rep. 2025, 15, 19506. [Google Scholar] [CrossRef]
  15. Abuwatfa, W.H.; Pitt, W.G.; Husseini, G.A. Scaffold-based 3D cell culture models in cancer research. J. Biomed. Sci. 2024, 31, 7. [Google Scholar] [CrossRef]
  16. Navarro-Garcia, D.; Marcos, A.; Beets-Tan, R.; Blomqvist, L.; Bodalal, Z.; Deandreis, D.; Crispin-Ortuzar, M.; Gallagher, F.; Giandini, T.; Graves, M.J.; et al. Real-world radiology data for artificial intelligence-driven cancer support systems and biomarker development. ESMO Real World Data Digit. Oncol. 2025, 8, 100120. [Google Scholar] [CrossRef]
  17. Lambin, P.; Leijenaar, R.T.H.; Deist, T.M.; Peerlings, J.; de Jong, E.E.C.; van Timmeren, J.; Sanduleanu, S.; Larue, R.T.H.M.; Even, A.J.G.; Jochems, A.; et al. Radiomics: The bridge between medical imaging and personalized medicine. Nat. Rev. Clin. Oncol. 2017, 14, 749–762. [Google Scholar] [CrossRef] [PubMed]
  18. He, W.; Huang, W.; Zhang, L.; Wu, X.; Zhang, S.; Zhang, B. Radiogenomics: Bridging the gap between imaging and genomics for precision oncology. MedComm 2024, 5, e722. [Google Scholar] [CrossRef]
  19. Jin, C.; Yu, H.; Ke, J.; Ding, P.; Yi, Y.; Jiang, X.; Duan, X.; Tang, J.; Chang, D.T.; Wu, X.; et al. Predicting treatment response from longitudinal images using multi-task deep learning. Nat. Commun. 2021, 12, 1851. [Google Scholar] [CrossRef]
  20. Alwahaibi, N.; Alwahaibi, M. Liver biopsy in the modern era: From traditional techniques to artificial intelligence and multi-omics integration. Front. Med. 2025, 12, 1678753. [Google Scholar] [CrossRef]
  21. Bayle, A.; Peyraud, F.; Belcaid, L.; Brunet, M.; Aldea, M.; Clodion, R.; Dubos, P.; Vasseur, D.; Nicotra, C.; Geraud, A.; et al. Liquid versus tissue biopsy for detecting actionable alterations according to the ESMO Scale for Clinical Actionability of molecular Targets in patients with advanced cancer: A study from the French National Center for Precision Medicine (PRISM). Ann. Oncol. 2022, 33, 1328–1331. [Google Scholar] [CrossRef] [PubMed]
  22. Rodríguez, J.; Avila, J.; Rolfo, C.; Ruíz-Patiño, A.; Russo, A.; Ricaurte, L.; Ordóñez-Reyes, C.; Arrieta, O.; Zatarain-Barrón, Z.L.; Recondo, G.; et al. When tissue is an issue the liquid biopsy is nonissue: A review. Oncol. Ther. 2021, 9, 89–110. [Google Scholar] [CrossRef] [PubMed]
  23. Ranganathan, K.; Kurian, N.S.; Goswami, H.M.; Rishi, K.D.; Veldore, V.H. Exploring the clinical utility of liquid biopsy with cfDNA in cancer: A systematic review. J. Liq. Biopsy 2024, 5, 100150. [Google Scholar] [CrossRef]
  24. Dalloul, Z.; Briend, J.G.; Diawara, M.; Taylor, C.; Ouellette, R.J. Leveraging liquid biopsy to uncover resistance mechanisms and guide personalized immunotherapy. Transl. Oncol. 2025, 59, 102445. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Article Metrics

Citations

Article Access Statistics

Multiple requests from the same IP address are counted as one view.