-
Homologous Recombination Is Associated with Enhanced Anti-Tumor Innate Immunity and Favorable Prognosis in Head and Neck Cancer -
Secondary Genetic Events and Their Relationship to TP53 Mutation in Mantle Cell Lymphoma: A Sub-Study from the FIL_MANTLE-FIRST BIO on Behalf of Fondazione Italiana Linfomi (FIL) -
The Tumor Environment in Peritoneal Carcinomatosis and Malignant Pleural Effusions: Implications for Therapy
Journal Description
Cancers
Cancers
is a peer-reviewed, open access journal of oncology published semimonthly online. The Irish Association for Cancer Research (IACR), Spanish Association for Cancer Research (ASEICA), Biomedical Research Centre (CIBM), British Neuro-Oncology Society (BNOS) and Spanish Group for Cancer Immuno-Biotherapy (GÉTICA) are affiliated with Cancers and their members receive a discount on the article processing charges.
- Open Access— free for readers, with article processing charges (APC) paid by authors or their institutions.
- High Visibility: indexed within Scopus, SCIE (Web of Science), PubMed, PMC, Embase, CAPlus / SciFinder, and other databases.
- Journal Rank: JCR - Q2 (Oncology) / CiteScore - Q1 (Oncology)
- Rapid Publication: manuscripts are peer-reviewed and a first decision is provided to authors approximately 19.1 days after submission; acceptance to publication is undertaken in 2.6 days (median values for papers published in this journal in the second half of 2025).
- Recognition of Reviewers: reviewers who provide timely, thorough peer-review reports receive vouchers entitling them to a discount on the APC of their next publication in any MDPI journal, in appreciation of the work done.
- Sections: published in 18 topical sections.
- Companion journals for Cancers include: Radiation and Onco.
- Journal Clusters of Oncology: Cancers, Current Oncology, Onco and Targets.
Impact Factor:
4.4 (2024);
5-Year Impact Factor:
4.8 (2024)
Latest Articles
Targeting Mesenchymal-Epidermal Transition (MET) Aberrations in Non-Small Cell Lung Cancer: Current Challenges and Therapeutic Advances
Cancers 2026, 18(2), 207; https://doi.org/10.3390/cancers18020207 (registering DOI) - 8 Jan 2026
Abstract
The mesenchymal–epithelial transition (MET) receptor is a tyrosine kinase activated by its sole known ligand, hepatocyte growth factor (HGF). MET signaling regulates key cellular processes, including proliferation, survival, migration, motility, and angiogenesis. Dysregulation and hyperactivation of this pathway are implicated in multiple malignancies,
[...] Read more.
The mesenchymal–epithelial transition (MET) receptor is a tyrosine kinase activated by its sole known ligand, hepatocyte growth factor (HGF). MET signaling regulates key cellular processes, including proliferation, survival, migration, motility, and angiogenesis. Dysregulation and hyperactivation of this pathway are implicated in multiple malignancies, including lung, breast, colorectal, and gastrointestinal cancers. In non–small cell lung cancer (NSCLC), aberrant activation of the MET proto-oncogene contributes to 1% of known oncogenic drivers and is associated with poor clinical outcomes. Several mechanisms can induce MET hyperactivation, including MET gene amplification, transcriptional upregulation of MET or HGF, MET fusion genes, and MET exon 14 skipping mutations. Furthermore, MET pathway activation represents a frequent mechanism of acquired resistance to EGFR- and ALK-targeted tyrosine kinase inhibitors (TKIs) in EGFR- and ALK-driven NSCLCs. Although MET has long been recognized as a promising therapeutic target in NSCLC, the clinical efficacy of MET-targeted therapies has historically lagged behind that of EGFR and ALK inhibitors. Encouragingly, several MET TKIs such as capmatinib, tepotinib, and savolitinib have been approved for the treatment of MET exon 14 skipping mutations. They have also demonstrated potential in overcoming MET-driven resistance to EGFR TKIs or ALK TKIs. On 14 May 2025, the U.S. Food and Drug Administration granted accelerated approval to telisotuzumab vedotin-tllv for adult patients with locally advanced or metastatic non-squamous NSCLC whose tumors exhibit high c-Met protein overexpression and who have already received prior systemic therapy. In this review, we summarize the structure and physiological role of the MET receptor, the molecular mechanisms underlying aberrant MET activation, its contribution to acquired resistance against targeted therapies, and emerging strategies for effectively targeting MET alterations in NSCLC.
Full article
(This article belongs to the Special Issue Advances in Precision Medicine: Targeting Known and Emerging Oncogenic Targets in Lung Cancer (2nd Edition))
►
Show Figures
Open AccessReview
Adipokine Metabolic Drivers, Gut Dysbiosis, and the Prostate Microbiome: Novel Pathway Enrichment Analysis of the Adiposity-Based Chronic Disease—Prostate Cancer Network
by
Zachary Dovey, Elena Tomas Bort and Jeffrey I. Mechanick
Cancers 2026, 18(2), 206; https://doi.org/10.3390/cancers18020206 (registering DOI) - 8 Jan 2026
Abstract
Adiposity-Based Chronic Disease (ABCD) is known to increase the risk of aggressive prostate cancer (PCa), recurrent disease after treatment for localized PCa, and PCa mortality. A key mechanistic link contributing to this enhanced risk is chronic inflammation originating from excess white visceral adipose
[...] Read more.
Adiposity-Based Chronic Disease (ABCD) is known to increase the risk of aggressive prostate cancer (PCa), recurrent disease after treatment for localized PCa, and PCa mortality. A key mechanistic link contributing to this enhanced risk is chronic inflammation originating from excess white visceral adipose tissue (WAT; VAT) and periprostatic adipose tissue (ppWAT). Contributing to systemic inflammation is gut dysbiosis, which itself may be caused by ABCD as well as background local inflammation (prostatitis), which is common in aging men and may be exacerbated by the urinary microbiome. Investigating the molecular biology driving inflammation and its association with increased PCa risk, a recent paper applied a network and gene set enrichment to adipokine drivers in the ABCD-PCa network. It found prominent roles for MCP-1, IL-1β, and CXCL-1 in addition to confirming the importance of exposure to lipopolysaccharides and bacterial components, corroborating the role of gut dysbiosis. To further unravel the mechanistic links between ABCD and PCa risk, this critical review will discuss the current literature on prominent inflammatory signaling pathways activated in ABCD; the influence of gut dysbiosis, the urinary microbiome, and chronic prostatitis; and current hypotheses on how these domains may result in the development of aggressive PCa over a man’s life. Moreover, we performed a novel pathway enrichment analysis to further evaluate the associations between ABCD, PCa risk, gut dysbiosis, and the prostate microbiome, the results of which were partitioned into extracellular and intracellular signaling pathways. In the extracellular space, novel mechanistic links between gut dysbiosis and MCP-1, IL-1β, CXCL1, and leptin via bacterial pathogen signaling and the intestinal immune network (for IgA production), crucial for gut immune homeostasis, were found. Within the intracellular space, there were downstream signals activating chemokine and type 2 interferon pathways, focal adhesion PI3K/Akt/mTOR pathways, as well as the JAK/STAT, NF-κB, and PI3K/Akt pathways. Overall, these findings point to an emerging molecular pathway for PCa oncogenesis influenced by ABCD, gut dysbiosis, and inflammation, and further research, possibly with lifestyle program-based clinical trials, may discover novel biomarker panels and molecular targeted therapies for the prevention and treatment of PCa.
Full article
(This article belongs to the Special Issue Advances in the Diagnosis and Treatment of Prostate Cancer: From Biomarkers to Precision Surgery)
►▼
Show Figures

Figure 1
Open AccessReview
Autophagy Modulates Immunogenic Cell Death in Cancer
by
Maiko Matsushita and Miyu Moriwaki
Cancers 2026, 18(2), 205; https://doi.org/10.3390/cancers18020205 - 8 Jan 2026
Abstract
Immunogenic cell death (ICD) is a subtype of regulated cell death characterized by the spatiotemporally coordinated emission of damage-associated molecular patterns (DAMPs), such as calreticulin (CALR), ATP, and high-mobility group box-1 (HMGB1), which collectively prime tumor-specific T-cell responses. Autophagy, a lysosome-dependent catabolic process,
[...] Read more.
Immunogenic cell death (ICD) is a subtype of regulated cell death characterized by the spatiotemporally coordinated emission of damage-associated molecular patterns (DAMPs), such as calreticulin (CALR), ATP, and high-mobility group box-1 (HMGB1), which collectively prime tumor-specific T-cell responses. Autophagy, a lysosome-dependent catabolic process, is increasingly recognized as a key modifier of antitumor immunity and the tumor microenvironment (TME). In preclinical models, autophagy can not only promote ICD by sustaining endoplasmic reticulum (ER) stress, eukaryotic translation initiation factor-2α (eIF2α) phosphorylation, and secretory pathways, but it can also limit ICD by degrading DAMPs, antigenic cargo, and major histocompatibility complex (MHC) molecules. The net outcome is highly context-dependent and determined by the tumor type, the nature and intensity of the stress, and the level at which autophagy is modulated. Herein, we summarize how autophagy affects the three canonical ICD-associated DAMPs, highlight solid-tumor models in which autophagy supports ICD, and contrast them with systems wherein autophagy inhibition is required for immunogenicity. We then focus on hematological malignancies, especially multiple myeloma, where recent reports implicate the autophagy-related protein GABARAP in bortezomib-induced ICD. Finally, we discuss the translational implications, including rational combinations of autophagy modulators with ICD-inducing chemotherapies, targeted drugs, and cellular immunotherapies, and outline the remaining challenges for safely harnessing the autophagy–ICD axis in the clinical setting.
Full article
(This article belongs to the Special Issue Autophagy and Apoptosis in Cancer Progression)
►▼
Show Figures

Figure 1
Open AccessCorrection
Correction: Kingshott et al. Alteration of Metabolic Conditions Impacts the Regulation of IGF-II/H19 Imprinting Status in Prostate Cancer. Cancers 2021, 13, 825
by
Georgina Kingshott, Kalina Biernacka, Alex Sewell, Paida Gwiti, Rachel Barker, Hanna Zielinska, Amanda Gilkes, Kathryn McCarthy, Richard M. Martin, J. Athene Lane, Lucy McGeagh, Anthony Koupparis, Edward Rowe, Jon Oxley, Jeff M. P. Holly and Claire M. Perks
Cancers 2026, 18(2), 204; https://doi.org/10.3390/cancers18020204 - 8 Jan 2026
Abstract
►▼
Show Figures
In the original publication [...]
Full article

Figure 1
Open AccessReview
Dietary Fiber and Melanoma: Exploring Microbiome-Driven Immune Modulation
by
Laci Turner, Connor K. Sisk and Nabiha Yusuf
Cancers 2026, 18(2), 203; https://doi.org/10.3390/cancers18020203 - 8 Jan 2026
Abstract
Background/Objectives: The gut microbiome influences melanoma biology and response to immune checkpoint inhibitors. Dietary fiber is a key modifiable factor that shapes the microbial composition and metabolite production. This review summarizes mechanistic, preclinical, and clinical evidence describing how fiber and fiber-responsive taxa
[...] Read more.
Background/Objectives: The gut microbiome influences melanoma biology and response to immune checkpoint inhibitors. Dietary fiber is a key modifiable factor that shapes the microbial composition and metabolite production. This review summarizes mechanistic, preclinical, and clinical evidence describing how fiber and fiber-responsive taxa may affect melanoma immunity and treatment outcomes. Methods: A literature search of MEDLINE, Embase, and Scopus identified studies published within the past five years examining dietary fiber, gut microbiome interactions, immune modulation, or melanoma outcomes. After screening 491 unique records, 49 peer-reviewed mechanistic, preclinical, observational, and interventional studies were synthesized qualitatively in this narrative review. Results: Fiber fermentation produces short-chain fatty acids that regulate dendritic cell activation, T-cell priming, and cytokine signaling. Preclinical melanoma models show that fibers such as inulin and β-glucan enhance IFN-γ-driven antitumor immunity, increase CD8+ infiltration, and improve checkpoint blockade efficacy in a microbiota-dependent manner. In humans, fiber-rich diets and enrichment of taxa such as Bifidobacterium, Faecalibacterium, and Akkermansia are associated with improved PD-1 inhibitor responses, longer progression-free survival, and possible reductions in ICI-related colitis. Although epidemiologic studies suggest no clear association between fiber intake and melanoma incidence, dietary fiber intake appears to correlate strongly with treatment-related outcomes. Conclusions: Dietary fiber represents a potentially safe and plausible adjunct to melanoma immunotherapy. However, study variability and emerging counterevidence highlight the need for controlled trials to clarify causality and define optimal fiber-based interventions.
Full article
(This article belongs to the Special Issue Gut Microbiome, Diet and Cancer Risk)
►▼
Show Figures

Figure 1
Open AccessCorrection
Correction: Rizzo et al. Endoscopic Ultrasound-Guided Anastomoses of the Gastrointestinal Tract: A Multicentric Experience. Cancers 2025, 17, 910
by
Giacomo Emanuele Maria Rizzo, Chiara Coluccio, Edoardo Forti, Alessandro Fugazza, Cecilia Binda, Giuseppe Vanella, Francesco Maria Di Matteo, Stefano Francesco Crinò, Andrea Lisotti, Marcello Fabio Maida, Giovanni Aragona, Aurelio Mauro, Alessandro Repici, Andrea Anderloni, Carlo Fabbri, Ilaria Tarantino and on behalf of the I-EUS Group
Cancers 2026, 18(2), 202; https://doi.org/10.3390/cancers18020202 - 8 Jan 2026
Abstract
In the published publication [...]
Full article
(This article belongs to the Special Issue Gastrointestinal and Its Associated Malignancies: Diagnosis, Targets and Therapies)
Open AccessReview
Rewiring Dendritic Cell Immunity: The β-Catenin–TIM-3 Axis as a Target to Improve DC Cancer Vaccines
by
Chunmei Fu, Tianle Ma, Li Zhou, Qing-Sheng Mi and Aimin Jiang
Cancers 2026, 18(2), 201; https://doi.org/10.3390/cancers18020201 - 8 Jan 2026
Abstract
The success of cancer vaccines relies on the ability of dendritic cells (DCs) to efficiently prime cytotoxic CD8 T cell responses against tumors. However, in solid tumors this process is often undermined by tumor-driven immunosuppression and intrinsic defects in DC activation. Among the
[...] Read more.
The success of cancer vaccines relies on the ability of dendritic cells (DCs) to efficiently prime cytotoxic CD8 T cell responses against tumors. However, in solid tumors this process is often undermined by tumor-driven immunosuppression and intrinsic defects in DC activation. Among the signaling pathways implicated in DC dysfunction, β-catenin signaling has emerged as a key regulator of immune tolerance in DCs. In parallel, inhibitory receptors such as PD-L1 and TIM-3 on DCs have been recognized as critical DC-intrinsic brakes on CD8 T cell priming and on responses to immune checkpoint blockade (ICB). Recent work has identified a DC-intrinsic immunoregulatory circuit in which β-catenin activation in DCs—particularly in cross-presenting cDC1s—induces expression of TIM-3, thereby suppressing CD8 T cell cross-priming and limiting anti-tumor CD8 T cell immunity. This β-catenin–TIM-3 axis represents a previously underappreciated layer of negative regulation that may help explain, at least in part, the limited efficacy of many current DC-based cancer vaccines. In this review, we examine how β-catenin activation in DCs, particularly in cDC1s, induces TIM-3 and related inhibitory programs that suppress cross-priming of tumor antigen-specific CD8 T cells and constrain the efficacy of DC-based vaccines. We further discuss how selectively targeting this β-catenin–TIM-3 checkpoint axis—alone or together with PD-L1 and other β-catenin–linked receptors—could restore DC function and inform rational combinations of DC-based vaccination with ICB and other T cell-based immunotherapies.
Full article
(This article belongs to the Special Issue The Cellular Ecosystem of Cancer: New Insights into Cell Biology)
►▼
Show Figures

Figure 1
Open AccessArticle
Prognosis from Pixels: A Vendor-Protocol-Specific CT-Radiomics Model for Predicting Recurrence in Resected Lung Adenocarcinoma
by
Abdalla Ibrahim, Eduardo J. Ortiz, Stella T. Tsui, Cameron N. Fick, Kay See Tan, Binsheng Zhao, Michelle Ginsberg, Lawrence H. Schwartz and David R. Jones
Cancers 2026, 18(2), 200; https://doi.org/10.3390/cancers18020200 - 8 Jan 2026
Abstract
Background: Radiomics can provide quantitative descriptors of tumor phenotype, but translation is often limited by feature instability across scanners and protocols. We aimed to develop and internally validate a protocol-specific CT-radiomics model using preoperative imaging to predict 5-year recurrence in patients with stage
[...] Read more.
Background: Radiomics can provide quantitative descriptors of tumor phenotype, but translation is often limited by feature instability across scanners and protocols. We aimed to develop and internally validate a protocol-specific CT-radiomics model using preoperative imaging to predict 5-year recurrence in patients with stage I lung adenocarcinoma after complete surgical resection. Methods: The retrospective study included 270 patients with completely resected stage I lung adenocarcinoma from January 2010–December 2021, among whom 23 (8.5%) experienced recurrence within five years. Radiomic features were extracted from routine preoperative CT scans. After preprocessing to remove highly constant and highly correlated features, the Synthetic Minority Over-sampling Technique addressed class imbalance in the training set. Recursive Feature Elimination identified the most predictive radiomic features. An XGBoost classifier was trained using optimized hyperparameters identified through RandomizedSearchCV with cross-validation. Model performance was evaluated using the ROC curve and predictive metrics. Results: Five radiomic features differed significantly between recurrence groups (p = 0.007 to <0.001): Shape Sphericity, first-order 90Percentile, GLCM Autocorrelation, GLCM Cluster Shade, and GLDM Large Dependence Low Gray Level Emphasis. The radiomics model showed excellent discriminatory ability with AUC values of 0.99 (95% CI: 0.98–1.00), 0.97 (95% CI: 0.91–1.00), and 0.96 (95% CI: 0.85–1.00) on the training, validation, and test sets, respectively. On the test set, the model achieved sensitivity of 100% (95% CI: 51–100%), specificity of 94% (95% CI: 81–98%), PPV of 67% (95% CI: 30–90%), NPV of 100% (95% CI: 90–100%), and overall accuracy of 95% (95% CI: 83–99%). Conclusions: Under protocol-homogeneous imaging conditions, CT radiomics accurately predicted recurrence in patients with completely resected stage I lung adenocarcinoma. External multi-vendor validation is needed before broader deployment.
Full article
(This article belongs to the Section Methods and Technologies Development)
►▼
Show Figures

Figure 1
Open AccessEditorial
Synthesizing Molecular Insights to Redefine the Battle Against Metastatic Cancer
by
Babak Behnam
Cancers 2026, 18(2), 199; https://doi.org/10.3390/cancers18020199 - 8 Jan 2026
Abstract
Metastasis represents the most formidable challenge in clinical oncology, accounting for over 90% of cancer-related deaths [...]
Full article
(This article belongs to the Special Issue Molecular Mechanisms of Cancer Development and Metastasis)
Open AccessArticle
L1CAM Promotes Human Endometrial Cancer Via NF-κB Activation
by
Hiroyuki Kurosu, Hiroshi Asano, Alaa-eldin Salah-eldin, Kazuya Hamada, Shugo Tanaka, Asuka Ishii, Issei Kawakita, Kentaro Kumagai, Kensuke Nakazono, Yuko Katayama, Rino Saito, Chihiro Terasaka, Sari Iwasaki, Satoshi Tanaka, Atsushi Niida, Hidemichi Watari and Koji Taniguchi
Cancers 2026, 18(2), 198; https://doi.org/10.3390/cancers18020198 - 8 Jan 2026
Abstract
Background/Objectives: Endometrial cancer is one of the most common gynecological malignancies, with increasing incidence and mortality rates, particularly in developed countries. L1 cell adhesion molecule (L1CAM) has been identified as a poor prognostic factor for human endometrial cancer; however, the molecular mechanisms
[...] Read more.
Background/Objectives: Endometrial cancer is one of the most common gynecological malignancies, with increasing incidence and mortality rates, particularly in developed countries. L1 cell adhesion molecule (L1CAM) has been identified as a poor prognostic factor for human endometrial cancer; however, the molecular mechanisms underlying its role in tumor progression remain unclear. Methods: We investigated the biological significance of L1CAM in human endometrial cancer using multiple cell lines. Functional analyses, including cell proliferation, cell cycle, and apoptosis assays, were performed after L1CAM knockdown or overexpression. Results: L1CAM promoted the transition of endometrial cancer cells from the G0/G1 phase and enhanced cell proliferation. L1CAM knockdown inhibited NF-κB signaling by reducing NF-κB (p65) phosphorylation and downregulating the expression of downstream targets such as TNF. Overexpression of constitutively active IKKβ restored the proliferation defect caused by L1CAM knockdown, supporting the role of NF-κB as a key downstream effector of L1CAM. Immunohistochemical analysis revealed a significant correlation between L1CAM expression and nuclear NF-κB (p65) positivity rates in human patient samples. Furthermore, combination therapy with cisplatin and an IKK inhibitor enhanced the anti-proliferative effect. Conclusions: Our study demonstrated that L1CAM promotes proliferation and chemotherapy resistance in human endometrial cancer through activation of the NF-κB signaling pathway. Therapeutic strategies targeting the L1CAM-NF-κB pathway may represent a promising treatment option for improving prognosis in L1CAM-positive human endometrial cancer.
Full article
(This article belongs to the Section Molecular Cancer Biology)
►▼
Show Figures

Figure 1
Open AccessReview
Minimally Invasive Pancreatoduodenectomy for Pancreatic Cancer: Current Perspectives and Future Directions
by
Munseok Choi and Chang Moo Kang
Cancers 2026, 18(2), 197; https://doi.org/10.3390/cancers18020197 - 7 Jan 2026
Abstract
Background: Minimally invasive pancreatoduodenectomy (MIPD) has evolved from an experimental technique to a feasible surgical option for pancreatic cancer in selected settings. However, its oncologic adequacy, safety, and generalizability remain debated, particularly given the biological aggressiveness of pancreatic ductal adenocarcinoma (PDAC) and the
[...] Read more.
Background: Minimally invasive pancreatoduodenectomy (MIPD) has evolved from an experimental technique to a feasible surgical option for pancreatic cancer in selected settings. However, its oncologic adequacy, safety, and generalizability remain debated, particularly given the biological aggressiveness of pancreatic ductal adenocarcinoma (PDAC) and the technical complexity of the procedure. Methods: This narrative review critically summarizes contemporary evidence regarding MIPD for pancreatic cancer, with particular attention to randomized controlled trials (RCTs), meta-analyses, and large observational studies. We distinguish findings derived from mixed periampullary tumor cohorts from those specific to PDAC and evaluate methodological limitations, learning-curve effects, and sources of heterogeneity across studies. Results: Recent RCTs and meta-analyses demonstrate that, when performed by experienced surgeons in high-volume centers, MIPD achieves perioperative outcomes comparable to open pancreatoduodenectomy, with advantages including reduced blood loss, shorter hospital stay, and faster functional recovery. Importantly, oncologic parameters such as R0 resection rates and lymph node yield appear equivalent between approaches, although robust long-term survival data from PDAC-specific RCTs remain lacking. Emerging evidence supports the feasibility of MIPD in complex clinical scenarios, including after neoadjuvant therapy, in frail or elderly patients, and in selected cases requiring vascular resection. Nonetheless, outcomes are strongly influenced by surgeon experience, institutional volume, and patient selection. Cost-effectiveness analyses and data from lower-volume centers remain limited. Conclusions: Current evidence supports MIPD as a viable alternative to open surgery for pancreatic cancer in carefully selected patients treated at specialized centers. However, claims of oncologic superiority are premature. Future research should focus on PDAC-specific randomized trials, standardized quality metrics, and strategies to mitigate learning-curve and resource-related barriers to broader implementation.
Full article
(This article belongs to the Special Issue Advances in Pancreatoduodenectomy)
Open AccessArticle
Crosstalk of Tumor-Derived Extracellular Vesicles with Immune Recipient Cells and Cancer Metastasis
by
Han Jie, Alicja C Gluszko and Theresa L. Whiteside
Cancers 2026, 18(2), 196; https://doi.org/10.3390/cancers18020196 - 7 Jan 2026
Abstract
Background. Contributions of tumor-derived extracellular vesicles, TEX, to tumor progression and metastasis involve their crosstalk with immune cells in the tumor microenvironment. This crosstalk results in metabolic reprogramming of immune cells from anti-tumor to pro-tumor activity. Mechanistic underpinnings of the TEX entry
[...] Read more.
Background. Contributions of tumor-derived extracellular vesicles, TEX, to tumor progression and metastasis involve their crosstalk with immune cells in the tumor microenvironment. This crosstalk results in metabolic reprogramming of immune cells from anti-tumor to pro-tumor activity. Mechanistic underpinnings of the TEX entry and delivery of molecular signals responsible for metabolic reprogramming may be unique for different types of immune cells. Methods. An in vitro model of THP-1 myeloid cells co-incubated with TEX illustrates the role TEX play in polarization of macrophages to TAMs. Results. In THP-1 cells, the dominant signaling pathway of melanoma cell-derived TEX involves HSP-90/TLR2. This leads to activation of the NF-κB and MAP kinase pathways and initiates THP-1 cell polarization from M0 to M2 with strong expression of immunosuppressive PD-L1. TEX may be seen as “danger” by the myeloid cells, which utilize the pattern recognition receptors (PRR), such as PAMPs or DAMPs, for engaging the complementary ligands carried by TEX. The same melanoma TEX signaling to T cells via DAMPs induced mitochondrial stress, resulting in T-cell apoptosis. Conclusions. As the signaling receptors/ligands in TEX are determined by the tumor, it appears that the tumor equips TEX with an address recognizing specific PRRs expressed on different recipient immune cells. Thus, TEX, acting like pathogens, are equipped by the tumor to alter the context of intercellular crosstalk and impose a distinct autophagy-not-apoptosis signature in recipient THP-1 cells. The tumor might endorse TEX to promote tumor progression and metastasis by enabling them to engage the signaling system normally used by immune cells for defense against pathogens.
Full article
(This article belongs to the Special Issue Exosomes in Cancer Metastasis (2nd Edition))
►▼
Show Figures

Figure 1
Open AccessReview
Nuclear Imaging in Renal Cell Carcinoma: Current Evidence and Clinical Applications
by
Abdullah Al-Khanaty, Shane Qin, Carlos Delgado, David Hennes, Eoin Dinneen, David Chen, Lewis Au, Renu S. Eapen, Damien Bolton, Declan G. Murphy, Nathan Lawrentschuk, Gregory Jack, Daniel Moon, Michael S. Hofman and Marlon L. Perera
Cancers 2026, 18(2), 195; https://doi.org/10.3390/cancers18020195 - 7 Jan 2026
Abstract
Introduction: Radiotracer-based nuclear imaging, including positron emission tomography (PET) and single-photon emission computed tomography (SPECT), can complement conventional cross-sectional imaging in renal cell carcinoma (RCC) by providing biological characterisation of tumour metabolism, angiogenesis, hypoxia, and the tumour microenvironment. While computed tomography (CT) and
[...] Read more.
Introduction: Radiotracer-based nuclear imaging, including positron emission tomography (PET) and single-photon emission computed tomography (SPECT), can complement conventional cross-sectional imaging in renal cell carcinoma (RCC) by providing biological characterisation of tumour metabolism, angiogenesis, hypoxia, and the tumour microenvironment. While computed tomography (CT) and magnetic resonance imaging (MRI) remain the diagnostic standard, accumulating evidence suggests that selected nuclear imaging techniques may offer incremental value in specific clinical scenarios. Methods: A narrative literature review was performed using PubMed, Embase, and Web of Science to identify preclinical, retrospective, and prospective studies evaluating PET and SPECT radiotracers in localised and metastatic RCC. Priority was given to meta-analyses, multicentre prospective trials, and studies with histopathological correlation. Results: [18F]fluorodeoxyglucose (FDG) PET/CT demonstrates limited sensitivity for primary renal tumours (pooled sensitivity of approximately 60%) but performs substantially better in metastatic and recurrent disease (pooled sensitivity and specificity of approximately 85–90%), where uptake correlates with tumour grade, progression-free survival, and overall survival. [99mTc]sestamibi SPECT/CT differentiates oncocytoma and hybrid oncocytic/chromophobe tumours from malignant RCC with pooled sensitivity and specificity of around 85–90%, supporting its role in evaluating indeterminate renal masses rather than staging. Prostate-specific membrane antigen (PSMA) PET/CT shows high detection rates in clear-cell RCC, particularly in metastatic disease, with reported sensitivities of approximately 85–90% and management changes in up to 40–50% of selected cohorts. Carbonic anhydrase IX (CAIX)-targeted PET/CT enables the biologically specific visualisation of clear-cell RCC, achieving sensitivities and specificities in the range of 85–90% in prospective phase II and III trials for primary tumour characterisation. Fibroblast activation protein inhibitor (FAPI) PET/CT demonstrates high tumour-to-background uptake in early RCC studies, but evidence remains preliminary, with small cohorts and recognised non-specific uptake in benign inflammatory and fibrotic conditions. Conclusions: Radiotracer-based nuclear imaging provides complementary, biology-driven insights in RCC that extend beyond anatomical assessment. While most modalities remain adjunctive or investigational and are not recommended for routine use, selective application in carefully chosen clinical scenarios may enhance tumour characterisation, prognostication, and personalised treatment planning.
Full article
(This article belongs to the Section Methods and Technologies Development)
Open AccessArticle
Prognostic Impact of Unplanned Hospitalization During First-Line Gemcitabine Plus Nab-Paclitaxel Therapy for Unresectable Pancreatic Cancer: A Single-Center Retrospective Observational Study
by
Kazuki Watabe, Motoyasu Kan, Izumi Ohno, Sodai Uchida, Taiga Sudo, Koki Yokozuka, Akinori Abe, Yoshiki Nakaya, Yoshiki Ogane, Hiroki Kurosaki, Miho Sakai, Yu Sekine, Tomoya Takahashi, Mayu Ouchi, Hiroshi Ohyama, Nozomu Sakai, Shigetsugu Takano, Tsukasa Takayashiki, Masayuki Ohtsuka and Jun Kato
Cancers 2026, 18(2), 194; https://doi.org/10.3390/cancers18020194 - 7 Jan 2026
Abstract
Background: Pancreatic cancer (PC) is a refractory malignancy with a dismal prognosis. For unresectable PC, gemcitabine plus nab-paclitaxel (GnP) is widely used as first-line chemotherapy. During treatment, patients may require unplanned hospitalization (UPH) due to tumor progression, biliary obstruction, or chemotherapy-related adverse events.
[...] Read more.
Background: Pancreatic cancer (PC) is a refractory malignancy with a dismal prognosis. For unresectable PC, gemcitabine plus nab-paclitaxel (GnP) is widely used as first-line chemotherapy. During treatment, patients may require unplanned hospitalization (UPH) due to tumor progression, biliary obstruction, or chemotherapy-related adverse events. Although UPH during chemotherapy may be linked to poorer survival, its prognostic impact as a time-dependent clinical event during active treatment has not been empirically evaluated in unresectable PC. We investigated the prognostic impact of UPH occurring during first-line GnP therapy. Objective: To clarify the association between UPH during first-line GnP and overall survival (OS). Methods: We retrospectively analyzed 189 patients with histologically confirmed unresectable PC who received first-line GnP at our institution between February 2016 and February 2023. The occurrence of UPH during GnP and the reason for the first UPH were categorized. Associations with OS were assessed using the Kaplan–Meier method and Cox proportional hazards models, including a time-varying covariate (TVC) analysis. Risk factors for UPH were examined with logistic regression. Results: UPH occurred in 76 patients (40.2%) during GnP. Pancreatic head tumors and pre-treatment biliary drainage were significantly more frequent in the UPH group. Median OS was 10.88 months in the UPH group versus 19.23 months in the non-UPH group; UPH was a significant adverse prognostic factor (hazard ratio [HR] 1.97, p < 0.01). In multivariable analysis incorporating a TVC, UPH remained an independent predictor of worse prognosis (HR 3.02, p < 0.01). Reasons for first UPH were progression (n = 28), recurrent biliary obstruction (RBO; n = 26), GnP-related adverse event (AE; n = 16), and other (n = 6). Hospitalization due to progression or RBO was associated with poorer survival. Pancreatic head location was identified as a risk factor for UPH. Conclusions: UPH during first-line GnP is an independent adverse prognostic factor in patients with unresectable PC, even after accounting for TVC. In pancreatic head cancer, closer monitoring for biliary and obstructive complications may be particularly important during treatment.
Full article
(This article belongs to the Section Clinical Research of Cancer)
►▼
Show Figures

Figure 1
Open AccessReview
“Radiological Grading” for Preoperative Assessment of Central Cartilaginous Tumors
by
Shinji Miwa, Katsuhiro Hayashi, Takashi Higuchi, Hirotaka Yonezawa, Sei Morinaga, Yohei Asano and Satoru Demura
Cancers 2026, 18(2), 193; https://doi.org/10.3390/cancers18020193 - 7 Jan 2026
Abstract
In the treatment of musculoskeletal tumors, surgeons sometimes experience discrepancies in the histological grade between the preoperative biopsy and resected tumor specimen, and the frequency of the histological discrepancies in cartilaginous tumors is higher than that in other bone tumors. For cartilaginous tumors,
[...] Read more.
In the treatment of musculoskeletal tumors, surgeons sometimes experience discrepancies in the histological grade between the preoperative biopsy and resected tumor specimen, and the frequency of the histological discrepancies in cartilaginous tumors is higher than that in other bone tumors. For cartilaginous tumors, new diagnostic tools or methods for the prediction of histological grades are required to determine appropriate surgical procedures for each patient. Several radiological findings have been reported to be useful in differentiating benign cartilaginous tumors, atypical cartilaginous tumors/grade 1 chondrosarcomas, and high-grade chondrosarcomas. Furthermore, recent studies have shown the high accuracy of radiological scoring systems that integrate several radiological findings to predict the histological grades of cartilaginous tumors. Radiomics, which converts features in radiological images into quantitative data, enables the comprehensive analysis of cartilaginous tumors. Recent reports suggest that radiological diagnoses are highly reliable compared with preoperative histological diagnoses for predicting the final histological diagnosis. Based on previous reports, “radiological grading,” i.e., the prediction of histological aggressiveness using radiological modalities, can be important for determining the surgical procedure, in addition to “histological grading.” This review article discusses radiological findings, integrated radiological scoring systems, and radiomics-based predictions of histological grades in cartilaginous tumors.
Full article
(This article belongs to the Special Issue Challenges and New Developments in the Diagnosis and Treatment of Sarcoma)
Open AccessArticle
Combinatorial Analysis of CD4+Tregs, CD8+Teffs, and Inflammatory Indices Predict Response to ICI in ES-SCLC Patients
by
Anastasia Xagara, Konstantinos Tsapakidis, Vassileios Papadopoulos, Alexandros Kokkalis, Evangelia Chantzara, Chryssovalantis Aidarinis, Alexandros Lazarou, George Christodoulopoulos, Matina Perifanou-Sotiri, Dimitris Verveniotis, Vasiliki Rammou, Maria Smaragdi Vlachou, Galatea Kallergi, Alexandra Markou, Ioannis Samaras, Filippos Koinis, Emmanouil Saloustros and Athanasios Kotsakis
Cancers 2026, 18(2), 192; https://doi.org/10.3390/cancers18020192 - 7 Jan 2026
Abstract
Background: Small-cell lung cancer (SCLC) is an aggressive type of lung cancer, and several factors are currently used to predict poor outcomes, including performance status (PS), extensive-stage disease, male sex, advanced age, and elevated lactate dehydrogenase (LDH) levels. In this study, we
[...] Read more.
Background: Small-cell lung cancer (SCLC) is an aggressive type of lung cancer, and several factors are currently used to predict poor outcomes, including performance status (PS), extensive-stage disease, male sex, advanced age, and elevated lactate dehydrogenase (LDH) levels. In this study, we aimed to explore the role of Tegs and inflammatory indices, such as CRP and NLR, in predicting response to immunotherapy. Methods: Fifty-one therapy-naïve ES-SCLC patients and ten healthy donors were enrolled. Peripheral blood mononuclear cells (PBMCs) were isolated and stained with fluorochrome-conjugated monoclonal antibodies. Multicolor flow cytometry was performed to determine the levels of CD8+ T cells and CD4+ Tregs, as well as their correlation with inflammatory indices and clinical outcomes. Results: ES-SCLC patients harbored higher percentages of CD8+ Teffs (p = 0.005) and FOXP3+ Tregs (p < 0.0001) in circulation before therapy compared with healthy donors. In addition, high levels of CD3+CD8+ T effectors were associated with longer PFS (p = 0.018) and longer OS (p = 0.012) compared with patients bearing low levels, while Tregs were not found to be predictive. More importantly, a survival benefit was observed in ES-SCLC patients with a low Treg/Teff ratio, as longer OS was observed in those with high percentages of CD8+ Teffs and low FOXP3+CTLA-4+ Tregs (p = 0.014) compared with those bearing low CD8+ Teffs and high FOXP3+CTLA-4+ Tregs. A low Treg/Teff ratio was further associated with low eosinophil levels and a low NLR before treatment initiation. Conclusions: These findings suggest a novel, easily obtainable blood-based signature that may help predict response to ICIs in ES-SCLC patients.
Full article
(This article belongs to the Special Issue Immune Biomarkers in Solid Tumors: From Peripheral Blood Parameters to Tissue-Based Indicators for Prognosis and Therapy Response)
►▼
Show Figures

Figure 1
Open AccessArticle
Effect of Preoperative Sarcopenic Obesity on Outcomes in Patients with Gastric Cancer After Surgery
by
Itaru Hashimoto, Keisuke Komori, Norihiro Akimoto, Yuta Nakayama, Shinsuke Nagasawa, Yukio Maezawa, Kyohei Kanematsu, Takanobu Yamada, Norio Yukawa, Aya Saito, Takashi Ogata and Takashi Oshima
Cancers 2026, 18(2), 191; https://doi.org/10.3390/cancers18020191 - 7 Jan 2026
Abstract
Background/Objectives: Preoperative body composition has been implicated as a factor affecting clinical outcomes in several types of cancer. However, there is limited evidence regarding whether preoperative body composition can predict the prognosis following gastrectomy for gastric cancer (GC). We aimed to investigate the
[...] Read more.
Background/Objectives: Preoperative body composition has been implicated as a factor affecting clinical outcomes in several types of cancer. However, there is limited evidence regarding whether preoperative body composition can predict the prognosis following gastrectomy for gastric cancer (GC). We aimed to investigate the role of preoperative body composition as a prognostic factor for overall survival (OS) and relapse-free survival (RFS) after gastrectomy for GC. Methods: This prospective study included 540 patients who underwent gastrectomy for GC at the Kanagawa Cancer Center, Japan, between December 2013 and November 2017. Preoperative body composition was assessed using the skeletal muscle index and visceral adipose tissue area derived from computed tomography scans. Patients were classified into four groups: non-sarcopenic non-obesity (NN), sarcopenic non-obesity (SN), non-sarcopenic obesity (NO), and sarcopenic obesity (SO). Results: A total of 448 patients (NN, 184; SN, 52; NO, 186; SO, 26) were included in the final analysis. In terms of OS, the SO group showed significantly worse survival than the NN group (72.1% vs. 87.6%, p = 0.01). Similarly, regarding RFS, the SO group had significantly worse outcomes than the NN group (68.4% vs. 86.2%, p = 0.007). Multivariate analysis identified SO as an independent risk factor for both OS (hazard ratio [HR], 3.18; 95% confidence interval [CI], 1.33–7.64; p = 0.01) and RFS (HR, 3.08; 95% CI, 1.36–6.95; p = 0.01). Conclusions: Preoperative SO was associated with poorer outcomes in patients undergoing gastrectomy for GC.
Full article
(This article belongs to the Special Issue Clinical Outcomes in Upper GI Cancers)
►▼
Show Figures

Figure 1
Open AccessSystematic Review
Application of Transthoracic and Endobronchial Elastography—A Systematic Review
by
Christian Kildegaard, Rune W. Nielsen, Christian B. Laursen, Ariella Denize Nielsen, Amanda D. Juul, Tai Joon An, Dinesh Addala and Casper Falster
Cancers 2026, 18(2), 190; https://doi.org/10.3390/cancers18020190 - 7 Jan 2026
Abstract
Introduction: Ultrasound elastography is increasingly used across medical imaging, yet its role in thoracic disease remains poorly defined. While both transthoracic ultrasonography (TUS) and endobronchial ultrasound (EBUS) offer real-time assessment of pleural and pulmonary structures, the diagnostic and clinical value of elastography in
[...] Read more.
Introduction: Ultrasound elastography is increasingly used across medical imaging, yet its role in thoracic disease remains poorly defined. While both transthoracic ultrasonography (TUS) and endobronchial ultrasound (EBUS) offer real-time assessment of pleural and pulmonary structures, the diagnostic and clinical value of elastography in this context remains uncertain. Materials and Method: A systematic search of MEDLINE, EMBASE, and the Cochrane Library was conducted according to PRISMA guidelines (April 2023; updated January 2025). Original studies evaluating transthoracic or endobronchial elastography for pleural or pulmonary conditions were included. Data extraction and quality assessment were performed independently by three reviewers, with QUADAS-2 used to evaluate risk of bias. Results: Thirty studies met inclusion criteria. Twenty-eight evaluated TUS elastography and two examined EBUS. Shear wave elastography was most frequently applied, particularly for differentiating malignant from benign pleural effusion or subpleural lesions. Surface wave elastography demonstrated consistently higher stiffness values in patients with interstitial lung disease compared with healthy controls, correlating with radiological and functional disease severity. Elastography-guided pleural biopsy improved diagnostic yield compared with conventional ultrasound-guided biopsy. Overall, substantial methodological variation existed among scanning techniques, elastography modalities, reporting methods, and diagnostic thresholds, limiting cross-study comparison. Conclusions: Ultrasound elastography shows promise for evaluating pleural effusion and pulmonary lesions, procedural guidance, and interstitial lung disease possibly improving diagnostic possibilities with bedside evaluation and reducing patient exposure to radiation. However, methodological variation and limited high-quality evidence preclude clinical implementation. Standardized acquisition protocols and multicentre validation studies are necessary to define its diagnostic utility in thoracic imaging.
Full article
(This article belongs to the Special Issue Application of Ultrasound in Cancer Diagnosis and Treatment)
►▼
Show Figures

Figure 1
Open AccessReview
Oligometastatic Bladder Cancer: Current Definitions, Diagnostic Challenges, and Evolving Therapeutic Strategies
by
Kieran Sandhu, David T. Hopkins, Matilda Newton, Niranjan Sathianathen, Sachin Perera, Nathan Lawrentschuk, Declan Murphy and Marlon Perera
Cancers 2026, 18(2), 189; https://doi.org/10.3390/cancers18020189 - 7 Jan 2026
Abstract
Background: Oligometastatic bladder cancer (OMBC) is increasingly recognised as an intermediate state between localised and widespread metastatic disease, although its definition and optimal management remain uncertain. Patients with OMBC have a generally more favourable prognosis compared to patients with metastatic disease. However,
[...] Read more.
Background: Oligometastatic bladder cancer (OMBC) is increasingly recognised as an intermediate state between localised and widespread metastatic disease, although its definition and optimal management remain uncertain. Patients with OMBC have a generally more favourable prognosis compared to patients with metastatic disease. However, its definition, diagnostic criteria, and optimal management remain poorly standardised. Methods: This narrative review summarises current evidence on the definitions, diagnostic approaches, and treatment strategies for OMBC, with an emphasis on emerging biological and molecular insights that may refine disease classification and guide therapy. Results: Existing definitions of OMBC rely on lesion count and anatomical distribution, overlooking molecular and clinicopathological heterogeneity that influences prognosis and treatment response. Advances in Positron Emission Tomography (PET)/Computed Tomography (CT) and magnetic resonance imaging (MRI) have improved detection of small-volume disease, while liquid biopsy and circulating tumour DNA show promise for assessing micrometastatic burden. Therapeutic approaches, including metastasis-directed and consolidative therapies, are under investigation. Nonetheless, most data are derived from small, retrospective series, and evidence from prospective studies remains limited. Conclusions: Prospective, biomarker-integrated, and randomised trials are essential to refine definitions, optimise patient selection for therapy, and define the role of precision-based multimodal therapy in OMBC management.
Full article
(This article belongs to the Special Issue Molecular and Clinical Challenges in Metastatic and Oligometastatic Genitourinary Cancers)
►▼
Show Figures

Figure 1
Open AccessArticle
Romiplostim for Prevention of Severe Chemotherapy-Induced Thrombocytopenia in Lymphoma Patients—Phase I Study
by
Erel Joffe, Zachary Epstein-Peterson, Lorenzo Falchi, Ariela Noy, Andrew D. Zelenetz, Collette Owens, Leah Gilbert, Gilles Salles and Gerald A. Soff
Cancers 2026, 18(2), 188; https://doi.org/10.3390/cancers18020188 - 6 Jan 2026
Abstract
Background/Objectives: Intensive chemotherapy is the cornerstone of lymphoma treatment but often leads to severe chemotherapy-induced thrombocytopenia (sCIT), resulting in treatment delays, reduced dose intensity, and the need for transfusions. While granulocyte colony-stimulating factors (G-CSFs) are commonly used to manage neutropenia, the use of
[...] Read more.
Background/Objectives: Intensive chemotherapy is the cornerstone of lymphoma treatment but often leads to severe chemotherapy-induced thrombocytopenia (sCIT), resulting in treatment delays, reduced dose intensity, and the need for transfusions. While granulocyte colony-stimulating factors (G-CSFs) are commonly used to manage neutropenia, the use of thrombopoietic growth factors has not been adequately studied. Methods: This phase I dose-finding study evaluated the use of weekly romiplostim as prophylaxis for recurrent sCIT in patients undergoing chemotherapy for lymphoma. Eligible patients were those treated with a 21-day chemotherapy cycle who previously experienced sCIT, thus serving as their own “controls”. sCIT was defined as one of the following: (A) a platelet count (PLT) <50 × 109/L on day 1 of the subsequent cycle, leading to delay or dose reduction in chemotherapy, or (B) grade 4 thrombocytopenia (<25 × 109/L) and/or (C) platelet transfusion for bleeding. The primary endpoints were the incidence of sCIT and the rate of romiplostim-associated-adverse-events, with thromboembolic complications being an event of special interest. Results: Nine patients with sCIT requiring a PLT transfusion on the prior treatment cycle were treated across three dose schedules. The phase 2 recommended schedule was defined as a starting dose of 3–5 mcg/kg based on the baseline PLT count, with weekly adjustments for counts <150 × 109/L and >450 × 109/L. Romiplostim prevented recurrent grade 4 thrombocytopenia in 47% of the chemotherapy cycles and averted recurrent transfusion in 65% of cycles. Notably, low starting doses, as used in solid malignancies, were insufficient, leading to recurrent thrombocytopenia. Conclusions: Romiplostim was well-tolerated, with no thromboembolic events, and allowed most patients to complete their chemotherapy on schedule at full dose intensity.
Full article
(This article belongs to the Section Clinical Research of Cancer)
►▼
Show Figures

Figure 1
Journal Menu
► ▼ Journal Menu-
- Cancers Home
- Aims & Scope
- Editorial Board
- Reviewer Board
- Topical Advisory Panel
- Instructions for Authors
- Special Issues
- Topics
- Sections & Collections
- Article Processing Charge
- Indexing & Archiving
- Editor’s Choice Articles
- Most Cited & Viewed
- Journal Statistics
- Journal History
- Journal Awards
- Society Collaborations
- Conferences
- Editorial Office
Journal Browser
► ▼ Journal BrowserHighly Accessed Articles
Latest Books
E-Mail Alert
News
Topics
Topic in
Biomolecules, Cancers, Cells, JMP, Livers
Signaling Pathways in Liver Disease 2nd Edition
Topic Editors: Ralf Weiskirchen, Amedeo AmedeiDeadline: 20 March 2026
Topic in
Cancers, IJMS, Pharmaceuticals, Pharmaceutics, Sci. Pharm., Current Oncology, Molecules
Recent Advances in Anticancer Strategies, 2nd Edition
Topic Editors: Hassan Bousbaa, Zhiwei HuDeadline: 31 March 2026
Topic in
Biomedicines, Cancers, JCM, Nanomaterials, Pharmaceutics, JNT
Application of Nanomaterials and Nanobiotechnology in Cancer
Topic Editors: Ayan Kumar Barui, Susheel Kumar NethiDeadline: 31 May 2026
Topic in
Biomedicines, Cancers, Diagnostics, JCM, Metabolites, Targets
Biomarkers of Disease: Discovery and Clinical Applications
Topic Editors: Ioannis Kanakis, Andreas TsakalofDeadline: 30 June 2026
Conferences
Special Issues
Special Issue in
Cancers
Tumor Microenvironment of Gynecological Tumors
Guest Editors: Zahid Pranjol, Dmitry Aleksandrovich ZinovkinDeadline: 10 January 2026
Special Issue in
Cancers
The Survival of Colon and Rectal Cancer (2nd Edition)
Guest Editor: Susanne MerkelDeadline: 15 January 2026
Special Issue in
Cancers
Cell Biology of Cancer Invasion: 2nd Edition
Guest Editor: Hideki YamaguchiDeadline: 15 January 2026
Special Issue in
Cancers
Cervical Cancer: Study on Molecular Landscape and Protein Biomarkers. Current Diagnostic and Therapeutic Capabilities
Guest Editors: Christopher Kobierzycki, Piotr DzięgielDeadline: 15 January 2026
Topical Collections
Topical Collection in
Cancers
Regulatory and Non-Coding RNAs in Cancer Epigenetic Mechanisms
Topical Collection in
Cancers
Innovations in Cancer Drug Development Research
Collection Editor: Anupam Bishayee
Topical Collection in
Cancers
Artificial Intelligence and Machine Learning in Cancer Research
Collection Editors: Jean-Emmanuel Bibault, Lei Xing
Topical Collection in
Cancers
The Molecular Predictive and Prognostic Biomarkers in Breast Cancer
Collection Editors: Daniele Generali, Ida Paris




