EDITORIAL article

Front. Immunol.

Sec. Cancer Immunity and Immunotherapy

Volume 16 - 2025 | doi: 10.3389/fimmu.2025.1631591

This article is part of the Research TopicClinical Implementation of Precision Oncology Data to Direct Individualized and Immunotherapy-Based Treatment StrategiesView all 18 articles

Editorial: Clinical Implementation of Precision Oncology Data to Direct Individualized and Immunotherapy-Based Treatment Strategies

Provisionally accepted
  • 1Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA, Columbus, United States
  • 2More than Moore’s Precision Medicine Solutions (MtM), Columbus, OH, 43230, USA, Columbus, United States
  • 3Private Health Management (PHM), Los Angeles, CA, 90024, USA, Los Angeles, United States
  • 4Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA, Notre Dame, United States
  • 5Harper Cancer Research Institute, South Bend, IN 46617, USA, South Bend, United States
  • 6Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, IN 46617, USA, South Bend, United States
  • 7Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA, Columbus, United States
  • 8Division of Hematology-Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA, Hershey, United States
  • 9Next-Generation Therapies Program, Penn State Cancer Institute, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA, Hershey, United States
  • 10Ignite Proteomics Inc., Golden, CO, 80401, USA, Golden, United States
  • 11Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, 20110, USA, Manassas, United States

The final, formatted version of the article will be published soon.

Figures 32Precision oncology has become a core component of cancer care and can be broadly defined as the use of molecular profiling data from clinically approved testing to inform therapeutic decision making or to stratify patients based on predicted treatment responses or toxicities. Technologies such as next-generation sequencing (NGS), multigene expression assays, and companion diagnostics are now routinely used to identify actionable mutations and optimize therapy selection for individual cancer patients (1). Accordingly, clinical practice guidelines (e.g., National Comprehensive Cancer Network) recommend comprehensive biomarker testing for nearly all patients with advanced or metastatic cancer to help identify the most effective treatments (2). However, despite recent scientific advancements and guideline recommendations, clinical implementation of emerging molecular testing strategies has been suboptimal. While there are obstacles to establishing precision oncology workflows even in large healthcare networks (3), community oncology settings (where over 80% of cancer patients receive care in the USA) face more significant barriers to adoption. These include delays in tissue access, limited tissue availability, and lack of the infrastructure that is required for comprehensive molecular testing, sample pre-processing/shipment or storage, and ordering (4). Consequently, while individualized treatment strategies based on precision oncology analysis that considers molecular profiling and other clinical testing data are technically, logistically, and financially achievable, this approach is not comprehensively implemented currently.In addition to NGS-based methods to profile both somatic and germline alterations, the past decade has also seen the development and clinical approval of multiple technologies and pan-omic approaches to interrogate disease. These data provide robust information to identify clinically actionable targets and potentially predict treatment efficacy or sensitivity/resistance. The first tissueagnostic drug in oncology (pembrolizumab) was approved by the United States Food and Drug Administration (FDA) in 2017 for the treatment of microsatellite instability (MSI)-High or mismatch repair deficient (dMMR) solid tumors. Since then, the field of precision oncology continues to grow as more drugs are approved based on molecular-specific testing independent from tissue of origin or disease indication/diagnosis (5). When patients are matched to the optimal targeted treatment, outcomes often improve compared to traditional chemotherapy or standard of care strategies. In a prospective study of treatment-naïve and advanced solid tumors, patients treated based on molecular profiling matching and Molecular Tumor Board (MTB) recommendations had higher disease control rates, longer progression-free survival, and improved overall survival than those treated with standard of care therapy (6). The conclusions of this trial were that administering personalized treatment earlier in the disease course may improve outcomes and targeted therapy combinations are more effective than mono-therapeutic approaches.Since FDA approvals of cancer therapeutics will be more commonly tumor-agnostic moving forward, clinicians will require a better understanding of commercially available clinical tests to be confident in using the results to guide treatment decisions for their patients. However, the interpretation of data from comprehensive molecular testing and prioritization of testing modalities can be overwhelming, especially in a community oncology setting. Thus, clinician education and decision support are essential to bridge the gap between research and practice. Medical Science Liaisons (MSLs) are clinical scientists with expertise in molecular profiling who can educate physicians on emerging molecular testing and targeted therapeutics. Furthermore, multidisciplinary MTBs where oncologists, geneticists, pathologists, and other research specialists review complex cases to interpret genomic findings and discuss emerging therapeutic options can also be valuable for clinician education/exposure. Consequently, more comprehensive education initiatives, including MSLs and MTBs, are needed so that every oncologist is equipped with the knowledge and confidence to practice precision oncology. To begin to achieve this overarching goal, the objective of this Research Topic is to provide high-quality clinical evidence and summarize emerging molecular tests to help guide clinical adoption; publication of this collection of research may drive efficient translation of scientific discoveries into clinical care and ultimately improve patient outcomes by facilitating effective implementation of precision oncology-based treatment strategies.Developing precision medicine-guided immunotherapeutic strategies can improve patient outcomes by tailoring treatment to individual tumor biology and molecular profiling data. This approach was summarized in a mini-review by Chhabra that outlined the transformative impact of integrating molecular profiling, advanced diagnostics, and artificial intelligence (AI) into precision oncology practice, particularly in the context of immunotherapies. Collectively, these studies advance the field of precision oncology by identifying novel biomarkers, establishing predictive testing models, and evaluating novel therapeutic combinations that may facilitate better patient stratification and could be integrated into clinical care to improve patient outcomes. Thus, this Research Topic collection adds to the research demonstrating that integration of multi-dimensional patient and molecular profiling data is key to optimizing cancer prognostication and tailoring precision oncology treatment strategies.Recent scientific advances in genomics, molecular diagnostics, and drug sensitivity testing have significantly outpaced clinical implementation, which has translated into suboptimal incorporation into routine care. Even in the most scientifically and medically advanced countries, comprehensive molecular testing is not widespread clinically and standardized molecular testing strategies to stratify patients with precision oncology-based treatments are still lacking. In the USA specifically, studies have shown that despite a majority of oncologists accepting that the field of precision oncology is an important and emerging part of the clinical environment with great growth potential, significantly less reported regularly using comprehensive genomic testing to guide treatment (7,8). Challenges to clinical implementation of precision oncology include physician education and adoption, data analysis and interpretation, clinical trial access, and unknown costs associated with insurance coverage/reimbursement (9). As of 2025, the USA has a robust pipeline of targeted drugs and immunotherapies. Moreover, comprehensive genomic/molecular testing is more accessible than ever; by clinician education through MSLs and fostering structured collaborations through MTBs, the research-practice gap can be more effectively addressed. Importantly, access and equity should also be a focus so that precision oncology can be standard of care for all patients, including those in community hospitals and underserved areas.In conclusion, precision oncology is an emerging and under-practiced therapeutic approach that has demonstrated improved patient outcomes in limited studies by a more comprehensive understanding of cancer biology on a molecular level. Looking ahead, research discoveries will bring even more sophisticated precision medicine approaches to the clinic, including AI models and prediction algorithms, whole transcriptome RNA expression profiling, proteomic analyses, and functional precision oncology approaches (e.g., organoid drug sensitivity testing). The challenge moving forward will be to efficiently implement, scale, and optimize precision oncology utilization across all cancers and care settings.The authors declare that the editorial was written in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest or competing interest.Acquisition, analysis, or interpretation of data for the work: (NAY, JP, AF, NSY, EP). Drafting the work or revising it critically for important intellectual content: (NAY, JP, AF, NSY, EP). Provided approval for publication of the content: (NAY, JP, AF, NSY, EP). Agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved (NAY, JP, AF, NSY, EP).

Keywords: Precision Oncology1, immunotherapy2, individualized patient treatment3, tumoragnostic clinical/molecular testing4, molecular profiling5, Next-generation Sequencing -NGS (DNAseq/RNAseq)6, pan-omics7, drug sensitivity testing8 Editorial on the Research Topic: Clinical Implementation of Precision Oncology Data to Direct Individualized and Immunotherapy-Based Treatment Strategies

Received: 20 May 2025; Accepted: 26 May 2025.

Copyright: © 2025 Young, Prosperi, Freud, Yee and Petricoin. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

* Correspondence: Nicholas Adam Young, Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA, Columbus, United States

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.