Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,160)

Search Parameters:
Keywords = personalized oncology

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 2972 KB  
Review
AI-Driven Digital Pathology: Deep Learning and Multimodal Integration for Precision Oncology
by Hyun-Jong Jang and Sung Hak Lee
Int. J. Mol. Sci. 2026, 27(1), 379; https://doi.org/10.3390/ijms27010379 (registering DOI) - 29 Dec 2025
Abstract
Pathology is fundamental to precision oncology, offering molecular and morphologic insights that enable personalized diagnosis and treatment. Recently, deep learning has demonstrated substantial potential in digital pathology, effectively addressing a wide range of diagnostic, prognostic, and biomarker-prediction tasks. Although early approaches based on [...] Read more.
Pathology is fundamental to precision oncology, offering molecular and morphologic insights that enable personalized diagnosis and treatment. Recently, deep learning has demonstrated substantial potential in digital pathology, effectively addressing a wide range of diagnostic, prognostic, and biomarker-prediction tasks. Although early approaches based on convolutional neural networks had limited capacity to generalize across tasks and datasets, transformer-based foundation models have substantially advanced the field by enabling scalable representation learning, enhancing cross-cohort robustness, and supporting few- and even zero-shot inference across a wide range of pathology applications. Furthermore, the ability of foundation models to integrate heterogeneous data within a unified processing framework broadens the possibility of developing more generalizable models for medicine. These multimodal foundation models can accelerate the advancement of pathology-based precision oncology by enabling coherent interpretation of histopathology together with radiology, clinical text, and molecular data, thereby supporting more accurate diagnosis, prognostication, and therapeutic decision-making. In this review, we provide a concise overview of these advances and examine how foundation models are driving the ongoing evolution of pathology-based precision oncology. Full article
Show Figures

Figure 1

23 pages, 2214 KB  
Review
Role of Gut Microbiome in Oncogenesis and Oncotherapies
by Renuka Sri Sai Peddireddi, Sai Kiran Kuchana, Rohith Kode, Saketh Khammammettu, Aishwarya Koppanatham, Supriya Mattigiri, Harshavardhan Gobburi and Suresh K. Alahari
Cancers 2026, 18(1), 99; https://doi.org/10.3390/cancers18010099 (registering DOI) - 29 Dec 2025
Abstract
The gut microbiome has emerged as a key regulator of human health, influencing not only metabolism and immunity but also the development and treatment of cancer. Mounting evidence suggests that microbial dysbiosis contributes to oncogenesis by driving chronic inflammation, producing genotoxic metabolites, altering [...] Read more.
The gut microbiome has emerged as a key regulator of human health, influencing not only metabolism and immunity but also the development and treatment of cancer. Mounting evidence suggests that microbial dysbiosis contributes to oncogenesis by driving chronic inflammation, producing genotoxic metabolites, altering bile acid metabolism, and disrupting epithelial barrier integrity. At the same time, the gut microbiome significantly modulates the host response to oncotherapies including chemotherapy, radiotherapy, and especially immunotherapy, where microbial diversity and specific taxa determine treatment efficacy and toxicity. This review synthesizes current evidence on the role of the gut microbiome in both oncogenesis and oncotherapies, focusing on thirteen cancers with the strongest and most clinically relevant microbiome associations, colorectal cancer, gastric cancer, hepatocellular carcinoma, gallbladder cancer, esophageal cancer, pancreatic cancer, oral squamous cell carcinoma, cervical cancer, prostate cancer, breast cancer, lung cancer, brain cancer, and melanoma. These cancers were selected based on robust mechanistic data linking microbial alterations to tumor initiation, progression, and therapy modulation, as well as their global health burden and translational potential. In addition, we have provided mechanistic insights or clinical correlations between the microbiome and cancer outcomes. Across cancers, common microbial mechanisms included pro-inflammatory signaling (e.g., NF-κB and STAT3 pathways), DNA damage from bacterial toxins (e.g., colibactin, nitrosating species), and metabolite-driven tumor promotion (e.g., secondary bile acids, trimethylamine N-oxide). Conversely, beneficial commensals such as Faecalibacterium prausnitzii and Akkermansia muciniphila supported antitumor immunity and improved responses to immune checkpoint inhibitors. In conclusion, the gut microbiome functions as both a driver of malignancy and a modifiable determinant of therapeutic success. Integrating microbiome profiling and modulation strategies such as dietary interventions, probiotics, and fecal microbiota transplantation into oncology practice may pave the way for personalized and more effective cancer care. Full article
Show Figures

Figure 1

17 pages, 2497 KB  
Article
Multimodal, Personalized Treatment of Pineal Region Tumors in Adulthood—A Single Center Study
by Tamás Mezei, János Báskay, Péter Pollner, Lukács Németh, Balázs Markia, Gábor Nagy, András Bajcsay and László Sipos
J. Clin. Med. 2026, 15(1), 248; https://doi.org/10.3390/jcm15010248 - 29 Dec 2025
Abstract
Background: Tumors of the pineal region account for less than 1% of supratentorial neoplasms in adults and represent a distinct neuro-oncological challenge. Their management requires a multidisciplinary and multimodal approach. Traditionally, direct surgical resection was considered the primary treatment modality. Recent advances in [...] Read more.
Background: Tumors of the pineal region account for less than 1% of supratentorial neoplasms in adults and represent a distinct neuro-oncological challenge. Their management requires a multidisciplinary and multimodal approach. Traditionally, direct surgical resection was considered the primary treatment modality. Recent advances in minimally invasive techniques and onco-radiotherapy have paved the way for safer and more personalized treatment strategies, in line with the principles of precision medicine. This study aims to present our institutional approach, which relies on a combination of endoscopic and radiotherapy-based techniques. Methods: A retrospective, single-center clinical study was conducted involving 28 adult patients who underwent endoscopic third ventriculostomy and biopsy of a pineal region tumor between January 2014 and March 2025. Descriptive statistics, permutation tests with bootstrap-derived confidence intervals, Fisher’s exact test, and Kaplan–Meier survival analysis were applied for data evaluation. Results: Endoscopic intervention resulted in clinical improvement in 78% of cases. A significant increase in performance status was observed in the postoperative period (<0.001) compared to preoperative results. Radiotherapy contributed to either tumor regression or disease stabilization. Conclusions: Based on our findings, the combination of endoscopic intervention and personalized radiotherapy represents a safe and effective treatment strategy, offering a compelling alternative to direct surgical resection, which is reserved as a second-line treatment. Full article
(This article belongs to the Section Oncology)
Show Figures

Figure 1

45 pages, 701 KB  
Conference Report
The Canadian Breast Cancer Symposium 2025: Meeting Report
by Christine Brezden-Masley, Katarzyna J. Jerzak, Nancy A. Nixon, Anne Koch, Amanda Roberts, Jean-François Boileau, May Lynn Quan, MJ DeCoteau and Tulin D. Cil
Curr. Oncol. 2026, 33(1), 15; https://doi.org/10.3390/curroncol33010015 - 27 Dec 2025
Viewed by 56
Abstract
The 2025 Canadian Breast Cancer Symposium (CBCS) brought together patients, clinicians and researchers from across Canada to discuss advances shaping personalized breast cancer care. Key updates in systemic therapy highlighted expanding treatment options, including CDK4/6 inhibitors, oral SERDs, PI3K/AKT-targeted therapies, and antibody–drug conjugates [...] Read more.
The 2025 Canadian Breast Cancer Symposium (CBCS) brought together patients, clinicians and researchers from across Canada to discuss advances shaping personalized breast cancer care. Key updates in systemic therapy highlighted expanding treatment options, including CDK4/6 inhibitors, oral SERDs, PI3K/AKT-targeted therapies, and antibody–drug conjugates across early and metastatic settings. Radiation oncology sessions emphasized treatment de-escalation, featuring evidence for ultra-hypofractionation, selective omission of nodal irradiation, and stereotactic strategies to manage oligoprogression. Surgical presentations focused on reducing morbidity through tailored axillary management and emerging techniques to prevent lymphedema. Advances in the management of central nervous system metastases underscored the growing synergy between stereotactic radiotherapy and CNS-active systemic therapies. Informed by patient testimony and advocacy perspectives, experts reflected on persistent gaps in diagnosis, access, and survivorship that shape priorities for future improvements. Together, these insights outline key directions that help to refine clinical practice and guide future research. Full article
(This article belongs to the Section Breast Cancer)
Show Figures

Graphical abstract

30 pages, 1670 KB  
Review
Combining Fluorescence and Magnetic Resonance Imaging in Drug Discovery—A Review
by Barbara Smolak, Klaudia Dynarowicz, Dorota Bartusik-Aebisher, Gabriela Henrykowska, David Aebisher and Wiesław Guz
Pharmaceuticals 2026, 19(1), 56; https://doi.org/10.3390/ph19010056 - 26 Dec 2025
Viewed by 247
Abstract
Drug discovery is a complex and multi-stage process that requires advanced analytical technologies capable of accelerating preclinical evaluation and improving the precision of therapeutic design. The combination of fluorescence and magnetic resonance imaging (MRI) within multimodal imaging plays an increasingly important role in [...] Read more.
Drug discovery is a complex and multi-stage process that requires advanced analytical technologies capable of accelerating preclinical evaluation and improving the precision of therapeutic design. The combination of fluorescence and magnetic resonance imaging (MRI) within multimodal imaging plays an increasingly important role in modern pharmacokinetics, integrating the high molecular sensitivity of fluorescence with the non-invasive anatomical visualization offered by MRI. Fluorescence enables real-time monitoring of cellular processes, including drug–target interactions and molecular dynamics, whereas MRI provides detailed structural information on tissues without exposure to ionizing radiation. Hybrid probes—such as superparamagnetic iron oxide nanoparticles (SPIONs) functionalized with near-infrared (NIR) fluorophores or gadolinium-based complexes linked to optical dyes—enable simultaneous acquisition of molecular and anatomical data in a single examination. These multimodal systems are being explored in oncology, neurology, and cardiology, where they support improved visualization of tumor biology, amyloid pathology, and inflammatory processes in vascular disease. Although multimodal imaging shows great promise for enhancing pharmacokinetic and pharmacodynamic studies, several challenges remain, including the potential toxicity of heavy-metal-based contrast agents, limited tissue penetration of fluorescence signals, probe stability in vivo, and the complexity and cost of synthesis. Advances in nanotechnology, particularly biodegradable carriers and manganese-based MRI contrasts, together with the integration of artificial intelligence algorithms, are helping to address these limitations. In the future, fluorescence–MRI hybrid imaging may become an important tool in personalized medicine, supporting more precise therapy planning and reducing the likelihood of clinical failure. Full article
(This article belongs to the Special Issue Advances in Medicinal Chemistry: 2nd Edition)
Show Figures

Graphical abstract

13 pages, 342 KB  
Review
A Review of the Latest Evidence on Prognostic Factors in Locally Advanced and Metastatic Urothelial Carcinoma Treated with Immune Checkpoint Inhibitors
by Ion Cojocaru, Mădălin Guliciuc, Elena Cojocaru, Cristina Serban, Grigore Pascaru, Mihnea Bogdan Borz, Vlad Horia Schitcu, Andrei-Ionut Tise, Iulian Osoianu and Laura-Florentina Rebegea
Medicina 2026, 62(1), 46; https://doi.org/10.3390/medicina62010046 - 26 Dec 2025
Viewed by 98
Abstract
Background and Objectives: Urothelial carcinoma (UC) is one of the most prevalent and lethal cancers worldwide. Identifying and understanding the factors that influence treatment outcome is essential for improving therapeutic effectiveness and predicting patient response. The objective of this review is to [...] Read more.
Background and Objectives: Urothelial carcinoma (UC) is one of the most prevalent and lethal cancers worldwide. Identifying and understanding the factors that influence treatment outcome is essential for improving therapeutic effectiveness and predicting patient response. The objective of this review is to estimate how clinical, biochemical, molecular and therapeutic factors impact the prognosis of patients with advanced urothelial carcinoma (aUC) and metastatic urothelial carcinoma (mUC) treated with immune checkpoint inhibitors (ICIs). Methods: A review was performed using PubMed, Scopus and Web of Science databases. All articles were published from 2013 to 2025 focusing on prognostic factors in locally advanced and metastatic urothelial carcinoma treated with ICIs. Results: Clinical prognostic factors for patients treated with ICIs include poor Eastern Cooperative Oncology Group (ECOG) performance status and the presence of liver or bone metastases, both associated with poor outcomes. Low hemoglobin levels and several biochemical markers, such as high neutrophil-to-lymphocyte ratio (NLR), elevated systemic immune-inflammation index (SII) and low serum sodium are also associated with reduced survival. Programmed cell death-ligand 1 (PD-L1) expression shows predictive relevance for ICI response. Concomitant use of antibiotics or proton pump inhibitors (PPIs) may diminish immunotherapy effectiveness. Additionally, sarcopenia and high lactate dehydrogenase (LDH) levels correlate with poorer clinical outcomes. Conclusions: Prognostic outcomes in aUC and mUC are influenced by a complex interaction of clinical, biochemical and molecular factors. Integrative prognostic models are essential to the guidance of personalized immunotherapeutic strategies and the improvement of patient outcomes in aUC and mUC. Full article
37 pages, 1606 KB  
Review
Hybrid Nanocarriers for Cancer Therapy: Advancements in Co-Delivery of Gene Therapy and Immunotherapy
by Kulzhan Berikkhanova, Isah Inuwa, Abdulrahman Garba Jibo, Nurzhan Berikkhanov, Nurzhan Bikhanov, Yessenkhan Sultan and Ardak Omarbekov
Int. J. Mol. Sci. 2026, 27(1), 248; https://doi.org/10.3390/ijms27010248 (registering DOI) - 25 Dec 2025
Viewed by 152
Abstract
Over the years, cancer has continued to be a leading global health threat, prompting researchers to explore advanced therapies that go beyond traditional treatments like chemotherapy and radiotherapy. Among these advanced therapies, gene therapy and immunotherapy have shown significant promise in treating cancer [...] Read more.
Over the years, cancer has continued to be a leading global health threat, prompting researchers to explore advanced therapies that go beyond traditional treatments like chemotherapy and radiotherapy. Among these advanced therapies, gene therapy and immunotherapy have shown significant promise in treating cancer by either altering genetic makeup or stimulating the immune system. However, their clinical applications face significant obstacles such as poor drug delivery, rapid degradation, and immune system clearance. Hybrid nanocarriers have emerged as a transformative development in modern precision oncology, enabling the co-delivery of gene therapy and immunotherapy agents in a highly targeted manner to address the persistent limitations of traditional cancer treatments. This review focuses on hybrid nanocarrier systems specifically engineered for co-delivery applications and critically evaluates when and how these multifunctional platforms outperform conventional single-modality or non-hybrid formulations. We compare key hybrid architectures in terms of payload compatibility, pharmacokinetics, immune modulation, and translational readiness, and examine the influence of tumor microenvironmental characteristics on their therapeutic performance. Particular emphasis is placed on stimuli-responsive designs, biomimetic surface engineering, and artificial intelligence–assisted optimization as emerging strategies to enhance co-delivery efficacy. By synthesizing current evidence and identifying key scientific and manufacturing gaps, this review aims to provide a practical foundation for advancing hybrid nanocarriers from laboratory development to clinically meaningful, personalized cancer therapies. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

14 pages, 240 KB  
Review
Antibody-Based Therapeutics in Breast Cancer: Clinical and Translational Perspectives
by Anna Balata and Katarzyna Pogoda
Antibodies 2026, 15(1), 3; https://doi.org/10.3390/antib15010003 - 25 Dec 2025
Viewed by 165
Abstract
Breast cancer remains the most common malignancy and one of the leading causes of cancer-related death among women worldwide. Advances in antibody-based therapies have improved outcomes across all biological subtypes: HER2-positive, triple-negative, and luminal breast cancer. Monoclonal antibodies such as trastuzumab and pertuzumab [...] Read more.
Breast cancer remains the most common malignancy and one of the leading causes of cancer-related death among women worldwide. Advances in antibody-based therapies have improved outcomes across all biological subtypes: HER2-positive, triple-negative, and luminal breast cancer. Monoclonal antibodies such as trastuzumab and pertuzumab have established HER2-targeted therapy as a standard of care, while immune checkpoint inhibitors have introduced immunotherapy into the treatment of triple-negative breast cancer. The emergence of antibody–drug conjugates (ADCs), including trastuzumab deruxtecan, sacituzumab govitecan, and datopotamab deruxtecan, has further expanded the available therapeutic options. Bispecific antibodies represent a new generation of agents with the potential to overcome resistance and enhance immune activation. Despite impressive progress, important challenges remain, including resistance mechanisms and the management of treatment-related toxicities. This review summarizes the biological rationale, clinical evidence, resistance mechanisms, and safety profiles of therapies based on monoclonal antibodies, bispecific antibodies, and antibody–drug conjugates in breast cancer. The development of these treatment modalities fosters the implementation of personalized, immunologically informed treatment strategies that are redefining precision oncology in breast cancer. Full article
(This article belongs to the Section Antibody-Based Therapeutics)
35 pages, 1000 KB  
Review
From Mechanism to Medicine: Peptide-Based Approaches for Cancer Diagnosis and Therapy
by Maria João Gouveia, Joana Campanhã, Francisca Barbosa and Nuno Vale
Biomolecules 2026, 16(1), 27; https://doi.org/10.3390/biom16010027 - 24 Dec 2025
Viewed by 502
Abstract
Therapeutic peptides have rapidly evolved into multifunctional tools for precision oncology, offering molecular specificity and biocompatibility. Their roles in cancer therapy, however, are inherently overlapping. The same peptide can function as a targeting ligand, a cell-penetrating motif, a therapeutic effector, or a structural [...] Read more.
Therapeutic peptides have rapidly evolved into multifunctional tools for precision oncology, offering molecular specificity and biocompatibility. Their roles in cancer therapy, however, are inherently overlapping. The same peptide can function as a targeting ligand, a cell-penetrating motif, a therapeutic effector, or a structural component of peptide–drug conjugates (PDCs), nanoparticle (NP) systems, and radionuclide constructs. This functional convergence makes rigid classification challenging. In this review, we therefore organize peptide modalities according to their dominant therapeutic function while acknowledging the fluid boundaries between categories. Firstly, we outline the main functional classes of therapeutic peptides, covering their use as targeting ligands and their roles as active agents (i.e., receptor agonists/antagonists, intracellular protein–protein interaction modulators, etc.). Additionally, we summarize their application in peptide–drug conjugates (PDCs), peptide-guided radionuclides, and cancer vaccines, integrating key mechanistic principles and clinical evidence. Finally, we discuss the major translational barriers to clinical use and how they might be overcome. The developments in peptide engineering position them as adaptable, multifunctional platforms capable of improving precision, reducing toxicity, and advancing personalized cancer care. Full article
Show Figures

Figure 1

14 pages, 527 KB  
Review
Circulating Tumor Cells in Glioblastoma
by Robert H. Eibl and Markus Schneemann
Cancers 2026, 18(1), 10; https://doi.org/10.3390/cancers18010010 - 19 Dec 2025
Viewed by 270
Abstract
Glioblastoma multiforme (GBM) remains a devastating brain tumor with poor prognosis, traditionally viewed as non-metastatic. The recent detection of circulating tumor cells (CTCs) in glioblastoma challenges this long-held view and opens new opportunities for liquid biopsy in neuro-oncology. This review summarizes current understanding [...] Read more.
Glioblastoma multiforme (GBM) remains a devastating brain tumor with poor prognosis, traditionally viewed as non-metastatic. The recent detection of circulating tumor cells (CTCs) in glioblastoma challenges this long-held view and opens new opportunities for liquid biopsy in neuro-oncology. This review summarizes current understanding of glioblastoma CTCs, emphasizing their unique properties, detection technologies, and differences compared to CTCs in extracranial cancers. Key challenges include their rarity, the absence of epithelial markers, and the presence of the blood–brain barrier. Despite the need for specialized enrichment approaches, CTC analysis in glioblastoma can offer helpful information regarding tumor heterogeneity, treatment response, and minimal residual disease. We discuss emerging clinical studies leveraging CTCs for early relapse detection and therapy monitoring. Integrating CTC phenotyping with molecular and functional characterization may enhance future personalized treatment strategies in glioblastoma. Refined CTC methodologies combined with other liquid biopsy modalities may transform glioblastoma management, improving patient outcomes through less invasive, dynamic tumor surveillance. Full article
Show Figures

Figure 1

23 pages, 1797 KB  
Review
Beyond Precision: Ambiomic Survivorship in Childhood and AYA Cancer
by Juan Antonio Ortega-García, Omar Shakeel, Nicole M. Wood, Antonio Pérez-Martínez, Jose Luís Fuster-Soler and Mark D. Miller
Cancers 2026, 18(1), 7; https://doi.org/10.3390/cancers18010007 - 19 Dec 2025
Viewed by 414
Abstract
Background: Survival among children and adolescents and young adults (AYA) with cancer has improved substantially over recent decades; however, dominant survivorship models remain reactive—activated post-treatment and anchored to static exposure- and organ-based screening. This design underuses the anticipatory window at diagnosis and overlooks [...] Read more.
Background: Survival among children and adolescents and young adults (AYA) with cancer has improved substantially over recent decades; however, dominant survivorship models remain reactive—activated post-treatment and anchored to static exposure- and organ-based screening. This design underuses the anticipatory window at diagnosis and overlooks environmental and social determinants that modulate outcomes across the life course. Methods: We narratively reviewed international frameworks including the Children’s Oncology Group (COG), the International Late Effects of Childhood Cancer Guideline Harmonization Group (IGHG), the Pan-European Network for Care of Survivors after Childhood and Adolescent Cancer (PanCare) and the National Comprehensive Cancer Network (NCCN), and synthesized evidence on environmental determinants, exposomics, toxicogenomics, and implementation. Building on two decades of real-world practice, we describe the evolution from the Pediatric Environmental History (PEHis) to the Ambiomic Health Compass (AHC), integrating genomic, exposomic, geospatial, clinical, and biomonitoring layers into routine care. In this framework, survivorship is conceptualized as beginning at the time of cancer diagnosis (“day 0”). Results: PEHis operationalizes guideline-based care with structured environmental and social assessment, personalized plans, and community integration, contributing to improved survival, healthier behaviors, reduced treatment-related mortality and stronger oncology–primary-care coordination. AHC extends PEHis with dynamic risk recalibration, contextual alerts, targeted biomonitoring, and toxicogenomic interpretation, enabling anticipatory decisions from day 0. The manuscript summarizes the paradigm shift (current vs. Ambiomic models), the domain-specific expansion over existing guidelines, the core clinical/system tools, and time-bound metrics (12, 24, 60 months) to support implementation and evaluation. Conclusions: Survivorship should move upstream—from late surveillance to ambiomic, exposure-aware care beginning at diagnosis. Integrating advanced exposomics, mutational epidemiology, and explainable analytics can reduce preventable events and chronicity, enhance equity, and align pediatric oncology with planetary health. The PEHis–AHC continuum offers a scalable blueprint for next-generation survivorship programs in Europe and beyond. Ambiomic medicine does not replace precision medicine—it completes and extends it by integrating exposomics, social context, and anticipatory analytics from day 0. Full article
Show Figures

Figure 1

25 pages, 926 KB  
Review
Extracellular Vesicle-Derived microRNAs: Novel Non-Invasive Biomarkers for Gastrointestinal Malignancies
by Daniela Nardozi, Valeria Lucarini, Valentina Angiolini, Nicole Feverati, Monica Benvenuto, Chiara Focaccetti, Letizia Del Conte, Olga Buccitti, Camilla Palumbo, Loredana Cifaldi, Elisabetta Ferretti, Roberto Bei and Laura Masuelli
Int. J. Mol. Sci. 2026, 27(1), 10; https://doi.org/10.3390/ijms27010010 - 19 Dec 2025
Viewed by 233
Abstract
Gastrointestinal (GI) cancers represent a heterogeneous group of malignant neoplasms arising from the digestive tract, including gastric, colorectal, hepatic, pancreatic, and biliary cancers. These tumors represent a major public health challenge due to their aggressive nature and poor prognosis. Although significant progress has [...] Read more.
Gastrointestinal (GI) cancers represent a heterogeneous group of malignant neoplasms arising from the digestive tract, including gastric, colorectal, hepatic, pancreatic, and biliary cancers. These tumors represent a major public health challenge due to their aggressive nature and poor prognosis. Although significant progress has been made in diagnostic imaging, endoscopy, and multimodal therapies, early detection remains difficult. Conventional serum biomarkers often lack sufficient sensitivity and specificity for reliable diagnosis, prompting a growing interest in identifying novel, minimally invasive biomarkers. In this context, liquid biopsy is emerging as a revolutionary tool in oncology. Among its components, extracellular vesicles (EVs) have gained increasing attention because they carry a wide range of molecular cargoes that reflect the biological state of their tumor of origin. In particular, EV-associated microRNAs (miRNAs) hold great promise as biomarkers for early cancer detection, real-time monitoring of disease progression, and assessment of therapeutic response. This review discusses the diagnostic and prognostic potential of EVs as novel biomarkers in GI cancers, emphasizing EV-contained miRNAs as a key resource for the development of personalized and precision medicine strategies. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

46 pages, 3465 KB  
Review
Cancer and Environmental Xenobiotics: Mechanisms, Controversies, and Innovations
by Alice N. Mafe and Dietrich Büsselberg
J. Xenobiot. 2026, 16(1), 2; https://doi.org/10.3390/jox16010002 - 19 Dec 2025
Viewed by 604
Abstract
Although cancer biology has advanced considerably, the impact of environmental toxins on carcinogenesis remains underrecognized and scattered across disciplines. Evidence increasingly shows that chronic exposure to a broad range of toxins—including persistent organic pollutants, heavy metals, pesticides, phthalates, microplastics, and fine particulate matter [...] Read more.
Although cancer biology has advanced considerably, the impact of environmental toxins on carcinogenesis remains underrecognized and scattered across disciplines. Evidence increasingly shows that chronic exposure to a broad range of toxins—including persistent organic pollutants, heavy metals, pesticides, phthalates, microplastics, and fine particulate matter (PM2.5), which significantly contributes to cancer initiation, progression, and treatment resistance. This review synthesizes mechanistic, molecular, and epidemiological findings from 2015 to 2025, identified through systematic searches of PubMed, Scopus, Web of Science, and MeSH. Key pathways include oxidative stress-mediated DNA damage, epigenetic reprogramming (DNA methylation, histone modifications, miRNA dysregulation), hormone receptor modulation, chronic inflammation, immune evasion, and tumor microenvironment remodeling. Case studies of benzene, arsenic, aflatoxins, pesticides, and microplastics detail exposure routes, molecular targets, and associated cancers, highlighting significant public health risks. Ongoing debates persist regarding safe exposure thresholds, latency periods, and the effects of mixed toxin exposures. The review also highlights recent innovations in environmental oncology, including AI-based predictive models, CRISPR screens for susceptibility genes, organoid/3D models, green chemistry interventions, and real-time exposure monitoring, which provide mechanistic insight and inform early detection and personalized prevention strategies. Additionally, regional data gaps, particularly in low- and middle-income countries, indicate the need for stronger interdisciplinary collaboration. By integrating molecular mechanisms, epidemiology, and technological advances, this review offers a comprehensive framework for understanding toxin-induced carcinogenesis and guiding future research, public health policy, and preventive strategies. Full article
Show Figures

Graphical abstract

14 pages, 8426 KB  
Article
Genetic Landscape of Solid Malignant Tumors in a Russian Cohort of Patients
by Iurii K. Slepov, Evgeniy D. Kopylov, Anton A. Turchin, Darya N. Khmelkova, Vladimir S. Kaimonov, Artur A. Isaev and Roman V. Deev
Diagnostics 2026, 16(1), 1; https://doi.org/10.3390/diagnostics16010001 - 19 Dec 2025
Viewed by 175
Abstract
Background/Objectives: Comprehensive genomic profiling (CGP) is a cornerstone of personalized oncology. However, large-scale, systematic data on the somatic mutation spectrum in Russian cancer patients are scarce. This study aimed to characterize the genomic landscape and assess the potential for matched therapy in [...] Read more.
Background/Objectives: Comprehensive genomic profiling (CGP) is a cornerstone of personalized oncology. However, large-scale, systematic data on the somatic mutation spectrum in Russian cancer patients are scarce. This study aimed to characterize the genomic landscape and assess the potential for matched therapy in a Russian cohort of patients with solid tumors. Methods: This retrospective study included 204 patients with various solid tumors. CGP was performed using the FoundationOne®CDx (FFPE tissue) and FoundationOne®Liquid CDx (cfDNA) platforms. The analysis assessed single-nucleotide variants, indels, copy number alterations, gene fusions, tumor mutational burden (TMB), microsatellite instability (MSI), and PD-L1 expression. Results: The most frequently mutated genes were TP53 (61.5%) and KRAS. The median TMB was 4.0 mut/Mb and was significantly lower in stage IV tumors. Significant co-occurrence was observed between KRAS and TP53 mutations, as well as between APC and KRAS mutations, which were particularly characteristic of colorectal cancer. KRAS mutations were associated with higher combined positive score (CPS) values in cases with lung cancer. Based on the CGP results, 44% of patients had findings that supported the use of an approved matched targeted therapy or immunotherapy for their tumor type. An additional 36% of patients had alterations indicating potential benefit from off-label targeted therapy. Conclusions: This study reveals the distinct genomic characteristics of solid tumors in a Russian cohort and confirms the high clinical utility of CGP for identifying actionable targets. Implementing CGP early in the diagnostic process is a necessary step towards realizing personalized treatment strategies for cancer patients. Full article
(This article belongs to the Section Pathology and Molecular Diagnostics)
Show Figures

Figure 1

16 pages, 534 KB  
Review
The Management of Muscle Invasive Bladder Cancer: State of the Art and Future Perspectives
by Antonio Cigliola, Brigida Anna Maiorano, Doga Dengur, Valentina Tateo, Chiara Mercinelli, Michela Piacentini, Sara Inguglia, Carlo Messina and Andrea Necchi
Cancers 2025, 17(24), 4017; https://doi.org/10.3390/cancers17244017 - 17 Dec 2025
Viewed by 630
Abstract
Background: Muscle-invasive bladder cancer (MIBC) represents a highly aggressive malignancy associated with significant morbidity and mortality. The current standard treatment, which includes radical cystectomy and platinum-based chemotherapy, is burdened by high toxicity and a substantial risk of relapse. For this reason, over the [...] Read more.
Background: Muscle-invasive bladder cancer (MIBC) represents a highly aggressive malignancy associated with significant morbidity and mortality. The current standard treatment, which includes radical cystectomy and platinum-based chemotherapy, is burdened by high toxicity and a substantial risk of relapse. For this reason, over the past decade, novel therapeutic strategies involving immune checkpoint inhibitors (ICIs), antibody–drug conjugates (ADCs), and targeted therapies have been investigated. This review aims to summarize current clinical evidence and ongoing trials evaluating these approaches in the perioperative setting. Methods: A systematic search was conducted using PubMed, EMBASE, and Cochrane databases, along with abstracts from major oncology conferences (ASCO, ESMO, SGO). Clinical trials assessing ICIs, ADCs, and targeted therapies, either alone or in combination with each other or with chemotherapy, in MIBC, were included. Results: Several early-phase and phase III trials have investigated the perioperative management of MIBC. Various studies evaluated the addition of ICIs to standard chemotherapy, demonstrating promising results in terms of pathological complete response. In parallel, the encouraging outcomes with ICIs and ADCs alone in the neoadjuvant or adjuvant setting paved the way for their combination in integrated strategies. Biomarker-driven approaches, based on circulating tumor DNA and specific genomic alterations, are being actively explored to improve patient selection and personalize treatment. Conclusions: ICIs, ADCs, and targeted therapies are reshaping the therapeutic landscape of MIBC. While early results are promising, further data and biomarker validation are essential to establish their definitive role and guide clinical decision-making in the perioperative setting. Full article
(This article belongs to the Special Issue Advances in Neoadjuvant Therapy for Urologic Cancer)
Show Figures

Figure 1

Back to TopTop