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
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
remove_circle_outline
remove_circle_outline

Search Results (4,433)

Search Parameters:
Keywords = combination chemotherapy

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 787 KB  
Review
Targeting Cancer Through Thymoquinone: From Molecular Mechanisms to Clinical Prospects
by Nosayba Al-Damook, Molham Sakkal, Mostafa Khair, Walaa K. Mousa, Ghalia Khoder and Rose Ghemrawi
Int. J. Mol. Sci. 2025, 26(22), 11029; https://doi.org/10.3390/ijms262211029 - 14 Nov 2025
Abstract
Thymoquinone (TQ), the active compound in Nigella sativa (black seed), has shown promising effects against cancer in many laboratory studies. In this review, we explore how TQ works on different aspects of cancer, from stopping cancer cell growth and spread, to triggering cancer [...] Read more.
Thymoquinone (TQ), the active compound in Nigella sativa (black seed), has shown promising effects against cancer in many laboratory studies. In this review, we explore how TQ works on different aspects of cancer, from stopping cancer cell growth and spread, to triggering cancer cell death, reducing inflammation, and helping the immune system fight back. We also highlight how TQ may overcome one of the biggest problems in cancer treatment—chemoresistance. When used together with common treatments like chemotherapy, radiation, or immunotherapy, TQ has been shown to improve their effects and reduce harmful side effects in preclinical models. Our review further discusses how TQ affects cancer stem cells, the tumor environment, and gene regulation through epigenetics. While these findings are encouraging, the lack of human studies remains a major gap. We also address TQ’s limited absorption and suggest ways to improve its delivery in the body, such as using nanoparticles or other carriers. Through this review, we aim to show the wide-ranging potential of TQ as a natural compound that may help make cancer treatments more effective and better tolerated. We call for clinical studies to take this research further and bring TQ closer to use in real-world cancer care. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Graphical abstract

19 pages, 1167 KB  
Review
mRNA-Based Neoantigen Vaccines in Pancreatic Ductal Adenocarcinoma (PDAC)—A Promising Avenue in Cancer Immunotherapy
by Jacek Kabut, Małgorzata Stopyra, Natalia Nafalska, Grzegorz J. Stępień, Michał Miciak, Marcin Jezierzański, Tomasz Furgoł, Krzysztof Feret and Iwona Gisterek-Grocholska
Int. J. Mol. Sci. 2025, 26(22), 10988; https://doi.org/10.3390/ijms262210988 - 13 Nov 2025
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most aggressive malignancies, with 5-year survival rates consistently below 5% despite advances in surgery, chemotherapy, and targeted therapy. Worldwide, PDAC remains highly lethal, with 458,918 new cases and 432,242 deaths in 2018—about a 94% mortality-to-incidence [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most aggressive malignancies, with 5-year survival rates consistently below 5% despite advances in surgery, chemotherapy, and targeted therapy. Worldwide, PDAC remains highly lethal, with 458,918 new cases and 432,242 deaths in 2018—about a 94% mortality-to-incidence ratio. The limited therapeutic efficacy is largely attributed to the pronounced heterogeneity of the disease, late clinical presentation, and the strongly immunosuppressive tumor microenvironment. In recent years, mRNA-based vaccines encoding patient-specific neoantigens have emerged as a promising immunotherapeutic modality. By delivering tailored antigenic sequences, these vaccines are capable of eliciting potent cytotoxic T-cell responses against tumor-restricted epitopes, thereby enhancing tumor immunogenicity while minimizing off-target effects. This review summarizes the biological rationale underlying mRNA vaccination in PDAC, recent progress in preclinical and early clinical trials, and key obstacles related to antigen selection, delivery platforms, and the immunosuppressive stroma. The potential integration of neoantigen mRNA vaccines into multimodal therapeutic strategies, including immune checkpoint inhibition and chemotherapy, is also discussed, underscoring their prospective role in overcoming resistance mechanisms and improving clinical outcomes in PDAC. However, most current data come from early-phase trials, with long-term benefits yet unproven. Definitive conclusions on efficacy and survival await results from ongoing randomized studies expected by 2028–2029. Further progress in neoantigen identification, delivery systems, and combination strategies is crucial to fully harness mRNA vaccine potential in PDAC. Full article
(This article belongs to the Special Issue Molecular Research for Cancer Immunotherapy)
Show Figures

Figure 1

36 pages, 5976 KB  
Review
The Unfolded Protein Response—Novel Mechanisms, Challenges, and Key Considerations for Therapeutic Intervention
by P. M. Quan Mai, Tam-Anh Truong, Sai Kumar Samala, Bhoomika Muruvekere Lakshmisha, Prapannajeet Biswal, Khadijeh Koushki, Prudhvi Chand Mallepaddi, Geraldine Vijay and Sunil Krishnan
Cancers 2025, 17(22), 3639; https://doi.org/10.3390/cancers17223639 - 13 Nov 2025
Viewed by 20
Abstract
Background: The unfolded protein response (UPR) is an evolutionarily conserved, synchronized, and orchestrated process triggered by eukaryotic cells in response to endoplasmic reticulum (ER) stress. UPR restores the ER’s capacity to handle large protein loads within it, and still fold and process these [...] Read more.
Background: The unfolded protein response (UPR) is an evolutionarily conserved, synchronized, and orchestrated process triggered by eukaryotic cells in response to endoplasmic reticulum (ER) stress. UPR restores the ER’s capacity to handle large protein loads within it, and still fold and process these proteins accurately. Many recent studies have documented the non-canonical roles of the UPR, outside of protein quality control, in the context of lipid metabolism and the immune system in cancer. Cancer cells have been known to hijack the UPR to promote survival and evade immune surveillance. However, the underlying mechanisms remain poorly understood. Objectives: Here, we critically summarize canonical and non-canonical UPR mechanisms in the contexts of tumor immune microenvironment and lipid metabolism, dissect their crosstalk with other cell fate signaling pathways within cancer, and propose therapeutic strategies to exploit this relationship. We also discuss the fundamental challenges of solely targeting UPR and emphasize the importance of patient stratification, biomarker development, and rational combination therapies to maximize the potential for therapeutic gain. We provide a deconvoluted mechanistic understanding of the UPR process in an attempt to spark prospective clinically relevant therapeutics research. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

27 pages, 1211 KB  
Review
Locally Advanced Cervical Cancer: Multiparametric MRI in Gynecologic Oncology and Precision Medicine
by Sara Boemi, Matilde Pavan, Roberta Siena, Carla Lo Giudice, Alessia Pagana, Marco Marzio Panella and Maria Teresa Bruno
Diagnostics 2025, 15(22), 2858; https://doi.org/10.3390/diagnostics15222858 - 12 Nov 2025
Viewed by 120
Abstract
Background: Locally advanced cervical cancer (LACC) represents a significant challenge in oncology, requiring accurate assessment of local extent and metastatic spread. Multiparametric magnetic resonance imaging (mpMRI) has assumed a central role in the loco-regional characterization of the tumor due to its high soft-tissue [...] Read more.
Background: Locally advanced cervical cancer (LACC) represents a significant challenge in oncology, requiring accurate assessment of local extent and metastatic spread. Multiparametric magnetic resonance imaging (mpMRI) has assumed a central role in the loco-regional characterization of the tumor due to its high soft-tissue resolution and the ability to integrate functional information. Objectives: In this narrative review, we explore the use of mpMRI in the diagnosis, staging, and treatment response of LACC, comparing its performance with that of PET/CT, which remains complementary for remote staging. The potential of whole-body magnetic resonance imaging (WB-MRI) and hybrid PET/MRI techniques is also analyzed, as well as the emerging applications of radiomics and artificial intelligence. The paper also discusses technical limitations, interpretative variability, and the importance of protocol standardization. The goal is to provide an updated and translational summary of imaging in LACC, with implications for clinical practice and future research. Methods: Prospective and retrospective studies, systematic reviews, and meta-analyses on adult patients with cervical cancer were included. Results: Fifty-two studies were included. MRI demonstrated a sensitivity and specificity greater than 80% for parametrial and bladder invasion, but limited sensitivity (45–60%) for lymph node disease, lower than PET/CT. Multiparametric MRI was useful in early prediction of response to chemotherapy and radiotherapy and in distinguishing residual disease from fibrosis. The integration of MRI into Image-Guided Adaptive Brachytherapy (IGABT) resulted in improved oncological outcomes and reduced toxicity. The applications of radiomics and AI demonstrated enormous potential in predicting therapeutic response and lymph node status in the MRI study, but multicenter validation is still needed. Conclusions: MRI is the cornerstone of the local–regional staging of advanced cervical cancer; it has become an essential and crucial tool in treatment planning. Its use, combined with PET/CT for lymph node assessment and metastatic disease staging, is now the standard of care. Future prospects include the use of whole-body MRI and the development of predictive models based on radiomics and artificial intelligence. Full article
(This article belongs to the Section Medical Imaging and Theranostics)
Show Figures

Figure 1

28 pages, 1940 KB  
Review
Practical Pharmacokinetic–Pharmacodynamic Models in Oncology
by Su Guan, Mei-Juan Tu and Ai-Ming Yu
Pharmaceutics 2025, 17(11), 1452; https://doi.org/10.3390/pharmaceutics17111452 - 11 Nov 2025
Viewed by 158
Abstract
Integrated pharmacokinetic (PK) and pharmacodynamic (PD) models are essential for the understanding of quantitative relationship between drug exposure and response towards the identification of optimal dosing regimens in drug development and clinical therapy. This article summarizes the common PK–PD models being established in [...] Read more.
Integrated pharmacokinetic (PK) and pharmacodynamic (PD) models are essential for the understanding of quantitative relationship between drug exposure and response towards the identification of optimal dosing regimens in drug development and clinical therapy. This article summarizes the common PK–PD models being established in oncology, with a focus on combination therapies. Among them, the PK models include those used for practical non-compartmental and compartmental analyses, as well as those for physiologically based modeling that describe and predict exposure to various chemotherapy, targeted therapy, and immunotherapy drugs. Built on proper natural disease progression models, such as the empirical logistic growth curve, the Gompertzian growth model, and their modifications, the integrated PK–PD models recapitulate and predict antitumor drug efficacy, in which the PD models include practical indirect response model and various tumor growth inhibition models, as driven by the mechanistic actions of the drugs administered. Since anticancer drugs are usually co-administered, PK–PD modeling has been extended from monotherapy to combination therapy. However, relying on a single interaction factor or parameter to capitulate complex drug interactions, predict outcomes of different combinations, and determine possible synergism is problematic. Considering the apparent contributions from individual drugs following mutual interactions, a new PK–PD model has been developed for combination therapy, which may be integrated with proper algorism (e.g., the Combination Index method) to critically define combination effects, synergism, additivity, or antagonism. As drug combinations become more complex and individual drug actions are variable, these models should be optimized further to advance the understanding of PK–PD relationships and facilitate the development of improved therapies. Full article
(This article belongs to the Special Issue Mechanism-Based Pharmacokinetic and Pharmacodynamic Modeling)
Show Figures

Graphical abstract

13 pages, 1681 KB  
Article
Na+/H+ Exchanger 1 Inhibition Overcomes Venetoclax Resistance in Acute Myeloid Leukemia
by Shin Young Hyun, Eun Jung Na, Yu Ri Kim, Yoo Hong Min and June-Won Cheong
Cells 2025, 14(22), 1759; https://doi.org/10.3390/cells14221759 - 10 Nov 2025
Viewed by 201
Abstract
Despite advances with novel targeted agents (e.g., BCL-2 or IDH inhibitors) combined with chemotherapy for acute myeloid leukemia (AML), drug resistance persists. We investigated whether blocking Na+/H+ exchanger 1 (NHE1) could enhance AML cell sensitivity to the BCL-2 inhibitor venetoclax [...] Read more.
Despite advances with novel targeted agents (e.g., BCL-2 or IDH inhibitors) combined with chemotherapy for acute myeloid leukemia (AML), drug resistance persists. We investigated whether blocking Na+/H+ exchanger 1 (NHE1) could enhance AML cell sensitivity to the BCL-2 inhibitor venetoclax and sought to determine the molecular mechanisms. Our results demonstrated that co-treatment with venetoclax and the NHE1 inhibitor 5-(N,N-hexamethylene) amiloride (HMA) synergistically induced apoptosis in both venetoclax-sensitive and -resistant leukemic cell lines. Specifically, the combination significantly increased apoptosis in venetoclax-resistant THP-1 cells to 72.28% (17.79% with 100 nM venetoclax and 10.15% with 10 μM HMA alone; p < 0.001). Conversely, another venetoclax-resistant line, U-937, showed no significant apoptotic response to the combination. In THP-1 cells, this synergy was mediated via a caspase-dependent programmed cell death pathway, evidenced by an increased BAX/BCL-2 ratio, mitochondrial cytochrome c release, and subsequent caspase-9 and caspase-3 activation. Furthermore, co-treatment downregulated the anti-apoptotic protein MCL-1 and reduced PI3K and Akt phosphorylation, suggesting that inhibition of these survival pathways also contributed to the synergistic effect. Inhibition of NHE1 may substantially enhance venetoclax sensitivity in certain AML models, particularly in venetoclax-resistant THP-1 cells but not in U-937, highlighting biological diversity and the probable involvement of alternative survival pathways. Full article
Show Figures

Figure 1

23 pages, 371 KB  
Review
Microbiota and Pancreatic Cancer: New Therapeutic Frontiers Between Engineered Microbes, Metabolites and Non-Bacterial Components
by Sara Sofia De Lucia, Enrico Celestino Nista, Marcello Candelli, Sebastiano Archilei, Franziska Deutschbein, Enrico Capuano, Antonio Gasbarrini, Francesco Franceschi and Giulia Pignataro
Cancers 2025, 17(22), 3618; https://doi.org/10.3390/cancers17223618 - 10 Nov 2025
Viewed by 373
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most aggressive and lethal human malignancies, with five-year survival rates showing only marginal improvement despite decades of intensive research. Its dismal prognosis reflects a combination of intrinsic biological aggressiveness, late clinical presentation, and marked resistance [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most aggressive and lethal human malignancies, with five-year survival rates showing only marginal improvement despite decades of intensive research. Its dismal prognosis reflects a combination of intrinsic biological aggressiveness, late clinical presentation, and marked resistance to standard therapies, underscoring the urgent need for innovative diagnostic and therapeutic approaches. Growing evidence indicates that the microbiome is a modifiable factor influencing the onset, progression, and treatment response of PDAC. Microbial communities originating from the gut, oral cavity, and even the tumor microenvironment can shape carcinogenic pathways, modulate immune activity, and alter the efficacy of chemotherapy and immunotherapy. In addition to bacteria, fungal and viral populations are emerging as relevant contributors within this complex ecosystem. This review provides a comprehensive overview of the current mechanistic and translational evidence linking the microbiome to PDAC biology and therapy. It further explores microbiota-targeted interventions—such as probiotics, postbiotics, engineered bacterial strains, bacteriophages, oncolytic viruses, and fecal microbiota transplantation—as promising adjuncts to conventional treatments. A deeper understanding of host–microbiome interactions could yield novel biomarkers and open innovative avenues for precision medicine in PDAC, ultimately improving patient outcomes and reshaping therapeutic paradigms. Integrating microbiome-based strategies into PDAC management may thus represent a crucial step toward more effective and personalized oncologic care. Full article
(This article belongs to the Special Issue New Insights into Microbial-Based Cancer Therapy)
24 pages, 7943 KB  
Article
Clofazimine Treatment Modulates Key Non-Coding RNAs Associated with Tumor Progression and Drug Resistance in Lethal Prostate Cancer
by Sarah Batten, Harish Kumar, Jeremiah Pfitzer, Daniel Chinedu Nweze, Suman Mazumder, Robert D. Arnold, Panagiotis Mistriotis, Taraswi Mitra Ghosh and Amit Kumar Mitra
Int. J. Mol. Sci. 2025, 26(22), 10892; https://doi.org/10.3390/ijms262210892 - 10 Nov 2025
Viewed by 305
Abstract
Metastatic castration-resistant prostate cancer/PCa (mCRPC) is a clinically advanced form of PCa that is associated with increased aggressiveness, cancer stemness, morbidity, and the risk of developing resistance to taxanes, currently the first-line chemotherapy for mCRPC. Clofazimine (CLF) is a potential immunomodulator drug that [...] Read more.
Metastatic castration-resistant prostate cancer/PCa (mCRPC) is a clinically advanced form of PCa that is associated with increased aggressiveness, cancer stemness, morbidity, and the risk of developing resistance to taxanes, currently the first-line chemotherapy for mCRPC. Clofazimine (CLF) is a potential immunomodulator drug that is FDA-approved for the treatment of leprosy. Recently, using in vitro, in vivo, and ex vivo models, we established the efficacy of CLF in chronic myeloid leukemia and multiple myeloma. Here, we demonstrate that CLF is effective as a single agent and in combination with taxanes in a panel of cell lines representing the diversity of CRPC patients. Using a microfluidic assay, we showed the impact of CLF on cancer cell migration and metastatic potential. Further, we also found that CLF reduces ALDH activity—a marker for cancer ‘stem-like’ cells (CSCs), a subtype of cancer cells with self-renewal and differentiation capacities (epithelial-to-mesenchymal transdifferentiation/EMT). Bulk and single-cell RNAseq followed by functional validation and in silico analysis showed that CLF treatment is associated with apoptosis, ER stress, oxidative phosphorylation, and mitochondrial dysfunction. Most importantly, CLF modulates the expression of several non-coding RNAs, including MALAT1 and NEAT1, that are linked to tumor cell proliferation, cell migration, and drug resistance. Full article
(This article belongs to the Special Issue Roles of Non-Coding RNAs in Cancer)
Show Figures

Figure 1

23 pages, 10690 KB  
Article
SMYD3–CDCP1 Axis Drives EMT and CAF Activation in Colorectal Cancer and Is Targetable for Oxaliplatin Sensitization
by Liming Zhao, Zhexue Wang, Pu Cheng, Guoli Sheng, Mingyu Han and Zhaoxu Zheng
Biomedicines 2025, 13(11), 2737; https://doi.org/10.3390/biomedicines13112737 - 9 Nov 2025
Viewed by 292
Abstract
Background: Colorectal cancer (CRC) mortality is predominantly driven by liver metastasis and poor responsiveness to chemotherapy. The histone methyltransferase SMYD3 has been implicated in oncogenic transcriptional programs; however, its downstream effectors and microenvironmental roles in CRC remain unclear. Methods: We investigated [...] Read more.
Background: Colorectal cancer (CRC) mortality is predominantly driven by liver metastasis and poor responsiveness to chemotherapy. The histone methyltransferase SMYD3 has been implicated in oncogenic transcriptional programs; however, its downstream effectors and microenvironmental roles in CRC remain unclear. Methods: We investigated whether SMYD3 regulates the transcription and function of the membrane receptor CDCP1, which mediates Src/PKCδ signaling and promotes invasion and stromal remodeling. A combination of molecular assays, including ChIP-qPCR, Western blotting, and co-culture experiments, was employed to examine the SMYD3–CDCP1 axis and its impact on epithelial–mesenchymal transition (EMT), cancer-associated fibroblast (CAF) activation, and oxaliplatin (OXA) sensitivity. Results: SMYD3 directly bound to the CDCP1 promoter and catalyzed H3K4me3 enrichment, thereby enhancing CDCP1 transcription. Upregulated CDCP1 activated Src/PKCδ signaling, facilitating EMT and CAF activation within the tumor microenvironment. Genetic suppression of SMYD3 reduced metastatic potential and improved oxaliplatin response in vivo, while genetic or pharmacologic perturbation attenuated tumor–stroma crosstalk and enhanced oxaliplatin sensitivity in vitro. Conclusions: The SMYD3–CDCP1 axis drives CRC progression by epigenetically promoting CDCP1 transcription and remodeling the tumor microenvironment. Targeting this pathway may provide a promising therapeutic strategy to restrain metastasis and enhance chemotherapy efficacy in CRC. Full article
(This article belongs to the Section Cancer Biology and Oncology)
Show Figures

Graphical abstract

12 pages, 802 KB  
Communication
RBM39 Contributes to MGMT Maintenance in Response to Temozolomide-Induced DNA Damage
by Vahid Khalaj, Jack T. Adams, Solmaz AghaAmiri, Servando Hernandez Vargas, Tyler M. Bateman, Sukhen C. Ghosh, Majid Momeny and Ali Azhdarinia
Cancers 2025, 17(22), 3604; https://doi.org/10.3390/cancers17223604 - 8 Nov 2025
Viewed by 232
Abstract
Resistance to alkylating chemotherapeutic agents such as temozolomide (TMZ) is a significant challenge in treating tumors with high MGMT expression, including MGMT-positive glioblastoma and neuroendocrine neoplasms. In this study, we investigated the effect of RNA-binding motif protein 39 (RBM39) downregulation on MGMT protein [...] Read more.
Resistance to alkylating chemotherapeutic agents such as temozolomide (TMZ) is a significant challenge in treating tumors with high MGMT expression, including MGMT-positive glioblastoma and neuroendocrine neoplasms. In this study, we investigated the effect of RNA-binding motif protein 39 (RBM39) downregulation on MGMT protein levels, based on prior observations suggesting an association between these two proteins. Pharmacological depletion or siRNA-mediated knockdown of RBM39 led to a marked reduction in MGMT protein levels in MGMT-expressing cancer cells. We further showed that dual targeting of RBM39 (using indisulam) and MGMT (with O6-benzylguanine) synergistically enhanced MGMT depletion. Functionally, combined indisulam and TMZ treatment significantly increased apoptosis and decreased clonogenic growth in neuroendocrine tumor cells. These findings identify MGMT as a downstream target of RBM39 in MGMT-expressing cancer cells and highlight the therapeutic potential of co-targeting RBM39 and MGMT to overcome resistance to alkylating chemotherapy. Full article
Show Figures

Figure 1

18 pages, 324 KB  
Review
Drug Repurposing in Veterinary Oncology: Myth or Reality?
by Stefano Ciccarelli, Chiara Perrone, Maria Alfonsa Cavalera and Antonio Giuliano
Vet. Sci. 2025, 12(11), 1067; https://doi.org/10.3390/vetsci12111067 - 6 Nov 2025
Viewed by 636
Abstract
Drug repurposing, that is, the identification of new therapeutic indications for existing medications, has been shown to be a cost-effective and time-efficient alternative to de novo drug development. This review provides a comprehensive overview of repurposed drugs in veterinary oncology, describing their mechanisms [...] Read more.
Drug repurposing, that is, the identification of new therapeutic indications for existing medications, has been shown to be a cost-effective and time-efficient alternative to de novo drug development. This review provides a comprehensive overview of repurposed drugs in veterinary oncology, describing their mechanisms of action, current evidence of clinical benefit, and translational relevance. The therapeutic agents discussed include non-steroidal anti-inflammatory drugs (e.g., piroxicam), metabolic modulators (e.g., metformin), anti-parasitic drugs (e.g., fenbendazole), immunomodulators (e.g., thalidomide, oclacitinib), cardiovascular agents (e.g., propranolol, statins, losartan), and other compounds such as auranofin and disulfiram. A critical evaluation of the extant evidence-based data from preclinical research, naturally occurring tumor models, and clinical studies is provided, with particular emphasis on both the therapeutic potential and the current limitations. The present review also focused on combination strategies and multimodal protocols, where repurposed drugs may enhance the efficacy of chemotherapy, targeted therapies, or immunotherapy. Challenges to clinical implementation, including limited funding, regulatory and ethical considerations, and the need for well-designed, multi-institutional clinical trials, are discussed. Ultimately, drug repurposing represents a practical and translationally valuable approach to broaden therapeutic options, improve quality of life in companion animals, and advance comparative oncology by promoting progress that benefits both veterinary and human patients. Full article
(This article belongs to the Special Issue Focus on Tumours in Pet Animals: 2nd Edition)
34 pages, 3741 KB  
Review
Enhancing Cancer Therapy with TLR7/8 Agonists: Applications in Vaccines and Combination Treatments
by Jagannath Mondal, Swayam Prabha, Thomas S. Griffith, David Ferguson and Jayanth Panyam
Cancers 2025, 17(21), 3582; https://doi.org/10.3390/cancers17213582 - 6 Nov 2025
Viewed by 535
Abstract
Targeting Toll-like receptors 7 and 8 (TLR7/8) has emerged as a promising strategy in cancer immunotherapy. TLR7/8 agonists activate robust Th1-type immune responses and bridge innate and adaptive immunity. Further, TLR7/8 agonists can serve as valuable adjuncts to conventional therapies, such as chemotherapy [...] Read more.
Targeting Toll-like receptors 7 and 8 (TLR7/8) has emerged as a promising strategy in cancer immunotherapy. TLR7/8 agonists activate robust Th1-type immune responses and bridge innate and adaptive immunity. Further, TLR7/8 agonists can serve as valuable adjuncts to conventional therapies, such as chemotherapy and radiotherapy, enhancing efficacy while reducing adverse effects. Their integration into combination regimens for cancer offers a dual advantage: amplifying antitumor immunity and reducing tumor burden. Notably, the incorporation of TLR7/8 agonists into cancer vaccine platforms has yielded encouraging results in preclinical models and is advancing toward clinical application. This review highlights the mechanisms of action, therapeutic potential, and recent progress in the development of TLR7/8 agonist-based strategies for cancer treatment. We also discuss ongoing clinical evaluations and the rationale for combining these agents with existing modalities to enable more effective, personalized, and accessible cancer therapies. Full article
(This article belongs to the Special Issue Insights from the Editorial Board Member)
Show Figures

Figure 1

23 pages, 1663 KB  
Review
Management of Musculoskeletal Oligometastatic Disease in Breast Cancer
by Kelly Kon-Liao, Josue Layme, Andrea Otero López-Lavalle, Marcos R. Gonzalez and Juan Pretell-Mazzini
Cancers 2025, 17(21), 3578; https://doi.org/10.3390/cancers17213578 - 6 Nov 2025
Viewed by 467
Abstract
Oligometastatic breast cancer represents an intermediate state between localized and disseminated disease with reasonable potential for clinical cure. Advancements in surgery, radiotherapy, and systemic therapy have improved prognosis. Due to the high prevalence of bone metastases, an increasing number of studies are evaluating [...] Read more.
Oligometastatic breast cancer represents an intermediate state between localized and disseminated disease with reasonable potential for clinical cure. Advancements in surgery, radiotherapy, and systemic therapy have improved prognosis. Due to the high prevalence of bone metastases, an increasing number of studies are evaluating new treatment strategies for oligometastatic bone disease. The decision to perform skeletal surgery is complex and depends on optimal patient selection. Major criteria include impending or pathologic long bone fractures, severe neurologic compromise, and an expected survival of over 3 months. Factors associated with improved survival include solitary bone metastases, preserved performance status, adequate surgical margins, absence of pathologic fracture, metachronous metastases, and ER-positivity status. Radiotherapy, especially SBRT, offers effective local control and palliation. Interventional radiology techniques such as percutaneous thermal ablation have also been described as potential treatment alternatives, particularly for fragile patients. Systemic treatment varies according to the tumor subtype. For HR+ and HER2 subtypes, a combination of endocrine therapy with CDK4/6 inhibitors may be considered. HER2+ patients are often treated with HER2-targeted therapies combined with chemotherapy. For triple-negative breast cancer, chemotherapy is the primary treatment. Bone-modifying agents are also recommended to maintain bone strength, prevent skeletal-related events, and reduce the need for additional interventions. Skeletal muscle metastases in breast cancer patients are rare and typically indicate advanced disease with poor prognosis. Treatment options include chemotherapy, radiotherapy, and surgical excision, but should be tailored to the patient’s clinical condition and prognosis. Full article
(This article belongs to the Section Cancer Metastasis)
Show Figures

Figure 1

17 pages, 1906 KB  
Article
Targeted Sequencing Identifies SNPs Associated with Antimalarial Drug Resistance and the CSP Vaccine Antigen in Plasmodium falciparum from Southwest Cameroon
by Mary T. Efeti, Sandra N. Fankem, Mariama T. Diallo, Methodius S. Lahngong, Nelson L. Acha, Robert A. Shey, Kristiaan Demeyer, Jacob Souopgui, Stephen M. Ghogomu and Rose Njemini
Int. J. Mol. Sci. 2025, 26(21), 10764; https://doi.org/10.3390/ijms262110764 - 5 Nov 2025
Viewed by 287
Abstract
Malaria is a major public health challenge in low- and middle-income countries with significant socio-economic impacts. While chemotherapy has greatly contributed to malaria control, the widespread emergence of resistance to antimalarial drugs threatens progress towards elimination goals. In parallel, the recent rollout of [...] Read more.
Malaria is a major public health challenge in low- and middle-income countries with significant socio-economic impacts. While chemotherapy has greatly contributed to malaria control, the widespread emergence of resistance to antimalarial drugs threatens progress towards elimination goals. In parallel, the recent rollout of the RTS,S/AS01 and R21/Matrix-M malaria vaccine—targeting the Plasmodium falciparum circumsporozoite protein (CSP)—offers a new prevention tool but may be influenced by parasite genetic diversity. This study investigated the genetic architecture of Plasmodium falciparum circulating in a community in the Southwest Region of Cameroon. Seventy-two blood samples were analyzed using targeted Oxford Nanopore sequencing of pfcrt, pfmdr1, pfdhfr, pfdhps, pfkelch13 and pfcsp genes. We observed a high prevalence of pfdhfr mutations (98.6% N51I, 98.6% C59R, 97.7% S108N) and pfmdr1 Y184F (76.1%) mutation. Mutations in pfdhps (54.2% S436A, 2.8% A437G, 38.9% A581G) were also observed. No WHO-validated pfkelch13 artemisinin resistance markers were found; however, K189T (63.4%) and R255K (4.2%) variants were detected. Nineteen non-synonymous SNPs were identified in pfcsp, reflecting natural background variations as vaccination status was not known. These findings support the continued use of artemisinin-based combination therapies and underscores the need for sustained molecular surveillance of both antimalarial drug resistance and vaccine-related polymorphisms, to inform malaria control strategies. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

23 pages, 628 KB  
Review
Expanding Horizons in Cholangiocarcinoma: Emerging Targets Beyond FGFR2 and IDH1
by Lily Darman, Quinn Kaurich, Md Sazzad Hassan, Urs von Holzen and Niranjan Awasthi
Int. J. Mol. Sci. 2025, 26(21), 10755; https://doi.org/10.3390/ijms262110755 - 5 Nov 2025
Viewed by 521
Abstract
Cholangiocarcinoma (CCA) is a biliary tract cancer that accounts for approximately 3% of all gastrointestinal cancers. CCA is a “silent” disease that remains undetected for a long period of time, often presenting at an advanced stage with minimal treatment options and a poor [...] Read more.
Cholangiocarcinoma (CCA) is a biliary tract cancer that accounts for approximately 3% of all gastrointestinal cancers. CCA is a “silent” disease that remains undetected for a long period of time, often presenting at an advanced stage with minimal treatment options and a poor prognosis. Advanced CCA remains largely inoperable, and combination gemcitabine plus cisplatin (GemCis) chemotherapy remains the standard treatment for patients affected by this disease. There is a desperate need for new therapeutic alternatives, and extensive research is ongoing to address this gap. Targeted therapies represent a rapidly expanding area of cancer treatment and are currently under active investigation in CCA. The FDA has approved the targeted therapies ivosidenib, pemigatinib, infigratinib, and futibatinib, as well as the immunotherapy durvalumab, for patients with CCA in recent years. Several other therapeutic strategies are still under investigation, targeting molecular pathways including p53/MDM2, JAK/STAT, KRAS, HER2, VEGFR, PDGFR, MET, ALK, MAPK, PI3K/AKT, BRAF, and DNA damage repair signaling. While several promising advancements have been made, further research is required to improve outcomes for patients with CCA. This review provides an up-to-date, comprehensive overview of currently approved targeted therapies in CCA, as well as those under investigation. Full article
(This article belongs to the Collection Latest Review Papers in Molecular Oncology)
Show Figures

Figure 1

Back to TopTop