bims-tumhet Biomed News
on Tumor heterogeneity
Issue of 2025–04–20
fourteen papers selected by
Sergio Marchini, Humanitas Research



  1. Int J Gynecol Cancer. 2024 Apr;pii: S1048-891X(24)01354-9. [Epub ahead of print]34(4): 550-558
       OBJECTIVE: In the PAOLA-1/ENGOT-ov25 trial (NCT02477644), adding maintenance olaparib to bevacizumab provided a substantial progression-free survival benefit in patients with newly diagnosed advanced ovarian cancer and homologous recombination deficiency (HRD)-positive tumors, irrespective of clinical risk. Subsequently, a clinically meaningful improvement in overall survival was reported with olaparib plus bevacizumab in the HRD-positive subgroup. We report updated progression-free survival and overall survival by clinical risk and HRD status.
    METHODS: Patients in clinical response after first-line platinum-based chemotherapy plus bevacizumab received maintenance olaparib (up to 24 months) plus bevacizumab (up to 15 months in total) or placebo plus bevacizumab. This post hoc analysis evaluated 5-year progression-free survival and mature overall survival in patients classified by clinical risk and HRD status.
    RESULTS: Of 806 randomized patients, 74% were higher-risk and 26% were lower-risk. In higher-risk HRD-positive patients, the hazard ratio (HR) for progression-free survival was 0.46 (95% confidence interval (95% CI) 0.34 to 0.61), with 5-year progression-free survival of 35% with olaparib plus bevacizumab versus 15% with bevacizumab alone; and the HR for overall survival was 0.70 (95% CI 0.50 to 1.00), with 5-year overall survival of 55% versus 42%, respectively. In lower-risk HRD-positive patients, the HR for progression-free survival was 0.26 (95% CI 0.15 to 0.45), with 5-year progression-free survival of 72% with olaparib plus bevacizumab versus 28% with bevacizumab alone; and the HR for overall survival was 0.31 (95% CI 0.14 to 0.66), with 5-year overall survival of 88% versus 61%, respectively. No benefit was seen in HRD-negative patients regardless of clinical risk.
    CONCLUSION: This post hoc analysis indicates that in patients with newly diagnosed advanced HRD-positive ovarian cancer, maintenance olaparib plus bevacizumab should not be limited to those considered at higher risk of disease progression. Five-year progression-free survival rates support long-term remission and suggest an increased potential for cure with particular benefit suggested in lower-risk HRD-positive patients.
    Keywords:  Ovarian Cancer
    DOI:  https://doi.org/10.1136/ijgc-2023-004995
  2. Int J Gynecol Cancer. 2024 Jun;pii: S1048-891X(24)01422-1. [Epub ahead of print]34(6): 906-918
       OBJECTIVES: Circulating tumor DNA (ctDNA) is emerging as a potential prognostic biomarker in multiple tumor types. However, despite the many studies available on small series of patients with ovarian cancer, a recent systematic review and meta-analysis is lacking. The objective of this study was to determine the association of ctDNA with progression-free-survival and overall survival in patients with epithelial ovarian cancer.
    METHODS: An electronic search was conducted using PubMed (MEDLINE), Embase, CENTRAL (Cochrane Library), and CINAHL-Complete from January 2000 to September 15, 2023. To be included in the analysis the studies had to meet the following pre-specified inclusion criteria: (1) evaluable ctDNA; (2) progression-free-survival and overall survival reported as hazard ratio (HR); and (3) the patient population had epithelial ovarian cancer at the time of ctDNA detection. We evaluated the association of ctDNA with progression-free survival and overall survival. Secondary outcomes focused on sub-group analysis of genomic alterations and international Federation of Gynecology and Obstetrics (FIGO) stage.
    RESULTS: A total of 26 studies reporting on 1696 patients with epithelial ovarian cancer were included. The overall concordance rate between plasma-based and tissue-based analyses was approximately 62%. We found that a high level of ctDNA in epithelial ovarian cancer was associated with worse progression-free survival (HR 5.31, 95% CI 2.14 to 13.17, p<0.001) and overall survival (HR 2.98, 95% CI 1.86 to 4.76, p<0.0001). The sub-group analysis showed a greater than threefold increase in the risk of relapse in patients with positive HOXA9 meth-ctDNA (HR 3.84, 95% CI 1.57 to 9.41, p=0.003).
    CONCLUSIONS: ctDNA was significantly associated with worse progression-free survival and overall survival in patients with epithelial ovarian cancer. Further prospective studies are needed.
    PROSPERO REGISTRATION NUMBER: CRD42023469390.
    Keywords:  Carcinoma, Ovarian Epithelial; Gynecologic Surgical Procedures; Gynecology; Neoplasm Recurrence, Local; Ovarian Neoplasms
    DOI:  https://doi.org/10.1136/ijgc-2024-005313
  3. STAR Protoc. 2025 Apr 11. pii: S2666-1667(25)00163-7. [Epub ahead of print]6(2): 103757
      Fragmentation patterns of plasma cell-free DNA (cfDNA) are promising biomarkers in cancer diagnosis. Here, we present a protocol to enrich tumor-derived cfDNA molecules through end selection. We describe steps for installing software, aligning plasma cfDNA data to the reference genome, and performing end selection on cfDNA. We then detail procedures for building cancer diagnostic models with artificial intelligence. Overall, we provide commands to align cfDNA whole-genome sequencing data starting from raw reads, then extract fragmentomic features, and finally build diagnostic models with performance evaluations. For complete information on the generation and use of this protocol, please refer to Ju et al.1.
    Keywords:  Cancer; Genomics; Sequence analysis
    DOI:  https://doi.org/10.1016/j.xpro.2025.103757
  4. Genome Res. 2025 Apr 14. 35(4): 572-582
      In this mini-review, we explore the advancements in genome-wide DNA methylation profiling, tracing the evolution from traditional methods such as methylation arrays and whole-genome bisulfite sequencing to the cutting-edge single-molecule profiling enabled by long-read sequencing (LRS) technologies. We highlight how LRS is transforming clinical and translational research, particularly by its ability to simultaneously measure genetic and epigenetic information, providing a more comprehensive understanding of complex disease mechanisms. We discuss current challenges and future directions in the field, emphasizing the need for innovative computational tools and robust, reproducible approaches to fully harness the capabilities of LRS in molecular diagnostics.
    DOI:  https://doi.org/10.1101/gr.278407.123
  5. Cancer Cell. 2025 Apr 14. pii: S1535-6108(25)00119-9. [Epub ahead of print]43(4): 708-727
      Artificial intelligence (AI) is increasingly being utilized in cancer research as a computational strategy for analyzing multiomics datasets. Advances in single-cell and spatial profiling technologies have contributed significantly to our understanding of tumor biology, and AI methodologies are now being applied to accelerate translational efforts, including target discovery, biomarker identification, patient stratification, and therapeutic response prediction. Despite these advancements, the integration of AI into clinical workflows remains limited, presenting both challenges and opportunities. This review discusses AI applications in multiomics analysis and translational oncology, emphasizing their role in advancing biological discoveries and informing clinical decision-making. Key areas of focus include cellular heterogeneity, tumor microenvironment interactions, and AI-aided diagnostics. Challenges such as reproducibility, interpretability of AI models, and clinical integration are explored, with attention to strategies for addressing these hurdles. Together, these developments underscore the potential of AI and multiomics to enhance precision oncology and contribute to advancements in cancer care.
    Keywords:  AI-aided diagnostics; artificial intelligence; cancer; machine learning; multiomics; translational oncology
    DOI:  https://doi.org/10.1016/j.ccell.2025.03.018
  6. Clin Cancer Res. 2025 Apr 16.
      Circulating tumor DNA (ctDNA) analysis is now a widely used diagnostic approach in the management of patients with solid tumors, including gene fusion-driven lung cancer. However, the accurate detection of genetic rearrangements in ctDNA presents considerable technical challenges. Consequently, studies that assess and compare methodologies for detection are vital.
    DOI:  https://doi.org/10.1158/1078-0432.CCR-25-0432
  7. Ann Oncol. 2025 Apr 15. pii: S0923-7534(25)00158-9. [Epub ahead of print]
       PURPOSE: Targeted therapy based on the molecular characterization of tumors has been among the most remarkable advances in cancer medicine. Here, we report the impact of almost 10 years of comprehensive genomic profiling in more than 2000 patients with advanced solid tumors at the Phase 1 unit, Department of Oncology, Rigshospitalet, Copenhagen, Denmark.
    MATERIALS AND METHODS: A prospective, single-center, single-arm open-label study (NCT02290522) was conducted, enrolling patients with advanced cancer referred to a Phase I Unit. Fresh tumor tissue was obtained for whole genome or exome sequencing (germline and somatic), RNA sequencing and SNP array. In cases where fresh tumor tissue was unavailable, archived formalin-fixed paraffin-embedded tumor tissue or circulating tumor DNA extracted from plasma were obtained for targeted panels. Genomic reports were reviewed and discussed by a multidisciplinary tumor board and actionable alterations were classified according to the ESMO scale for the clinical actionability of molecular targets (ESCAT). When possible, patients were treated with regimen matched to the genomic profile.
    RESULTS: A total of 2147 patients with advanced cancer and exhausted treatment options were enrolled from April 2013 to December 2021. Genomic profiles were obtained in 1866 patients (87%). At least one actionable target was identified in 1062 patients (57%) with a total of 1614 actionable alterations including high RNA CEACAM5 expression (N=559, 35%), homologous recombination deficiency (HRD) alteration (N=314, 20%) and tumor mutational burden-high (N=84, 5%). Overall, 256 patients (24% of the patients with an actionable target) were treated with targeted therapy and among these 14 patients were treated with more than one line of targeted therapy. In total, 274 targeted treatment regimens were initiated, and 259 treatments were evaluable with an overall response (OR) rate of 25% (CI95%: 0.20 - 0.30). ESCAT I/II was associated with improved OR, along with better progression-free survival (PFS) and overall survival (OS).
    CONCLUSION: This large-scale study demonstrates that genomic profiling is efficient to identify actionable targets and to match patients to targeted therapies.
    Keywords:  ESCAT; Genomic profile; Phase 1; Targeted therapy
    DOI:  https://doi.org/10.1016/j.annonc.2025.04.004
  8. Mol Diagn Ther. 2025 Apr 16.
      Cell-free DNA (cfDNA) is changing the face of liquid biopsy as a minimally invasive tool for disease detection and monitoring, with its main applications in oncology and prenatal testing, and rising roles in transplant patient monitoring. However, the processes of cfDNA biogenesis, fragmentation, and clearance are complex and require further investigation. Evidence suggests that cfDNA production relates to mechanisms of cell death and DNA repair, both of which further influence fragment size and its applicability as a biomarker. An emerging domain, cfDNA fragmentomics is being explored for advancing the field of diagnostics using non-mutational signatures such as fragment size ratios and methylation patterns. Thus, this review examines structural diversity in cfDNA with various fragment sizes. In examining these cfDNA subsets, we discuss their distinct biological origins and potential clinical utility. Development of sequencing methodologies has broadened the application of cfDNA in diagnosing cancers and organ-specific pathologies, as well as directing personalized therapies. This has been achieved by identifying and uncovering different subsets of cfDNA in biofluids using different methodologies and biofluids. Different cfDNA subsets provide important insights regarding genomic and epigenetic features, enhancing the understanding of gene regulation, tissue-specific functions, and disease progression. Advancement of these key areas further asserts increasing clinical relevance for the use of cfDNA as a biomarker. Continued exploration of cfDNA subsets is expected to drive further innovation in liquid biopsy and its integration into routine clinical practice.
    DOI:  https://doi.org/10.1007/s40291-025-00773-x
  9. Cancer Cell. 2025 Apr 14. pii: S1535-6108(25)00127-8. [Epub ahead of print]43(4): 577-580
      This special issue of Cancer Cell is dedicated to "bridging the gap between foundational cancer biology and clinical oncology." We asked scientists and clinicians to give us a brief "state of the field," discussing the integration of new technologies, the importance of patient-centric approaches, the need for innovative clinical trial designs, and how fostering teamwork and inclusive research can help us bridge the gap between cancer research and effective clinical applications.
    DOI:  https://doi.org/10.1016/j.ccell.2025.03.026
  10. Clin Cancer Res. 2025 Apr 14.
       PURPOSE: Epithelial ovarian cancer (EOC) is a lethal malignancy. CA125, the "best" available marker for detecting EOC, has insufficient sensitivity and specificity for earlier-stage disease and is not a meaningful screening tool, motivating the search for further biomarkers. Cancer biomarker discovery is enhanced by "omics" technologies. Discovery studies for EOC biomarkers should be conducted in pre-diagnosis blood samples from prospective cohorts to maximize likelihood of identifying markers that can detect disease before usual diagnosis and in earlier disease stage, while reducing methodologic biases.
    EXPERIMENTAL DESIGN: Individual cohorts with pre-diagnosis blood samples have insufficient sample size for such studies. thus, we established "PREDICT" ("Prospective Early Detection Consortium for Ovarian Cancer")-a collaboration of nine prospective studies-to assemble a sufficient number of EOC cases with blood samples collected ≤18 months before diagnosis plus controls. The 457 cases and 1,687 controls have circulating CA125 measured using a clinical assay.
    RESULTS: The discrimination capacity for single CA125 measurements in samples collected <6 months prior to diagnosis was high (area under the curve (AUC), PREDICT overall=0.92; range across cohorts of non-pregnant individuals=0.89-0.98), and declined with extended time between blood collection and diagnosis. Between-cohort variability in CA125 levels and predictive performance was observed.
    CONCLUSIONS: Ongoing investigations in PREDICT are evaluating the early detection potential of tumor-associated autoantibodies and microRNAs, using CA125 as a benchmark. PREDICT is a well-characterized resource for identifying and validating detection markers for EOC that may then be used in multi-modal screening, as a complement to CA125 and combined with imaging.
    DOI:  https://doi.org/10.1158/1078-0432.CCR-24-1845
  11. Cytopathology. 2025 Apr 11.
      The advent of personalised and precision medicine has radically modified the management and the clinical outcome of cancer patients. However, the expanding number of predictive, prognostic, and diagnostic biomarkers has raised the need for simple, noninvasive, quicker, but equally efficient tests for molecular profiling. In this complex scenario, the adoption of liquid biopsy, particularly circulating tumour DNA (ctDNA), has been a real godsend for many cancer patients who would otherwise have been denied the benefits of targeted treatments. Undeniably, ctDNA analysis has several advantages over conventional tissue-based analysis. One advantage is that it can guide treatment decision making, especially when tissue samples are scarce or totally unavailable. Indeed, a simple blood test can inform clinicians on patients' response or resistance to targeted therapies, help them monitor minimal residual disease (MRD) after surgical resections, and facilitate them with early cancer detection and interception. Finally, an equally important advantage is that ctDNA analysis can help decipher temporal and spatial tumour heterogeneity, a mechanism highly responsible for therapeutic resistance. In this review, we gathered and analysed current evidence on the clinical usefulness of ctDNA analysis in solid tumours.
    Keywords:  NGS; ctDNA; liquid biopsy; molecular oncology; molecular pathology
    DOI:  https://doi.org/10.1111/cyt.13485
  12. Int J Mol Sci. 2025 Apr 03. pii: 3333. [Epub ahead of print]26(7):
      Restrictions resulting from the COVID-19 pandemic abruptly reversed the slow decline of the diagnosis and mortality rates of gastric cancer (GC). This scenario highlights the importance of developing cost-effective methods for mass screening and evaluation of treatment response. In this study, we evaluated a non-invasive method based on the circulating methylated cell-free DNA (cfDNA) of Reprimo (RPRM), a tumor suppressor gene associated with the development of GC. Methylated RPRM cfDNA was analyzed in three de-identified cohorts: Cohort 1 comprised 81 participants with GC and 137 healthy donors (HDs); Cohort 2 comprised 27 participants with GC undergoing gastrectomy and/or chemotherapy analyzed at the beginning and after three months of treatment; and Cohort 3 comprised 1105 population-based participants in a secondary prevention program who underwent esophagogastroduodenal (EGD) endoscopy. This cohort includes 180 normal participants, 845 participants with premalignant conditions (692 with chronic atrophic gastritis [AG] and 153 with gastric intestinal metaplasia/low-grade dysplasia [GIM/LGD]), 21 with high-grade dysplasia/early GC [HGD/eGC], and 59 with advanced GC [aGC]). A nested case-control substudy was performed using a combination of methylated RPRM cfDNA and pepsinogens (PG)-I/II ratio. The dense CpG island of the promoter region of the RPRM gene was bisulfite sequenced and analyzed to develop a fluorescence-based real-time PCR assay (MethyLight). This assay allows the determination of the absolute number of copies of methylated RPRM cfDNA. A targeted sequence of PCR amplicon products confirmed the gastric origin of the plasma-isolated samples. In Cohort 1, the mean value of GCs (32,240.00 copies/mL) was higher than that of the HD controls (139.00 copies/mL) (p < 0.0001). After dividing this cohort into training-validation subcohorts, we identified an area under the curve of 0.764 (95% confidence interval (CI) = 0.683-0.845) in the training group. This resulted in a cut-off value of 87.37 copies/mL (sensitivity 70.0% and specificity 80.2%). The validation subcohort predicted a sensitivity of 66.67% and a specificity of 83.33%. In Cohort 2 (monitoring treatment response), RPRM levels significantly decreased in responders (p = 0.0042) compared to non-responders. In Cohort 3 (population-based participants), 18.9% %, 24.1%, 30.7%, 47.0%, and 71.2% of normal, AG, GIM/LGD, HGD/eGC, and aGC participants tested positive for methylated RPRM cfDNA, respectively. Overall sensitivity and specificity in distinguishing normal/premalignant conditions vs. GC were 65.0% (95% CI 53.52% to 75.33%) and 75.9% (95% CI 73.16% to 78.49%), respectively, with an accuracy of 75.11% (95% CI 72.45% to 77.64%). Logistic regression analyses revealed an OR of 1.85 (95% CI 1.11-3.07, p = 0.02) and an odds ratio (OR) of 3.9 (95% CI 1.53-9.93, p = 0.004) for the risk of developing GIM/LGD and HGD/eGC, respectively. The combined methylated RPRM cfDNA and PG-I/II ratio reached a sensitivity of 78.9% (95% CI 54.43% to 93.95%) and specificity of 63.04% (95% CI 52.34% to 72.88%) for detecting HGD/eGC vs. three to six age- and sex-matched participants with premalignant conditions. Our results demonstrate that methylated RPRM cfDNA should be considered a direct biomarker for the non-invasive detection of GC and a predictive biomarker for treatment response.
    Keywords:  biomarkers; cancer prevention; cancer screening; cfDNA; gastric cancer; liquid biopsy; methylated RPRM; non-invasive diagnosis
    DOI:  https://doi.org/10.3390/ijms26073333
  13. Int J Gynecol Cancer. 2024 Apr;pii: S1048-891X(24)01374-4. [Epub ahead of print]34(4): 478-479
      
    Keywords:  ovarian cancer
    DOI:  https://doi.org/10.1136/ijgc-2024-005400