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

Search Results (2,042)

Search Parameters:
Keywords = therapeutic landscapes

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 439 KB  
Review
Neuroblastoma in Childhood: Biological Insights, Risk Stratification, and Advances in Multimodal Therapy
by Amina De Bona, Martina Barbieri, Nicole Rinaldi and Susanna Esposito
J. Clin. Med. 2026, 15(3), 1101; https://doi.org/10.3390/jcm15031101 - 30 Jan 2026
Abstract
Neuroblastoma is the most common extracranial solid tumor of childhood and remains a leading cause of cancer-related mortality in pediatric patients. Characterized by marked clinical and biological heterogeneity, the disease ranges from spontaneously regressing tumors in infants to highly aggressive, treatment-resistant malignancies in [...] Read more.
Neuroblastoma is the most common extracranial solid tumor of childhood and remains a leading cause of cancer-related mortality in pediatric patients. Characterized by marked clinical and biological heterogeneity, the disease ranges from spontaneously regressing tumors in infants to highly aggressive, treatment-resistant malignancies in older children. Advances in molecular biology and genomics have significantly improved understanding of neuroblastoma pathogenesis, revealing the critical role of genetic and epigenetic alterations—such as MYCN amplification, ALK mutations, and chromosomal aberrations—in disease behavior and prognosis. Contemporary risk stratification systems now integrate clinical, biological, and molecular features to guide therapy more precisely. Management strategies have evolved toward risk-adapted, multimodal approaches. Low- and intermediate-risk patients often achieve excellent outcomes with surgery alone or limited chemotherapy, whereas high-risk neuroblastoma requires intensive multimodal treatment including induction chemotherapy, surgical resection, high-dose chemotherapy with autologous stem cell rescue, radiotherapy, and maintenance therapy. The incorporation of immunotherapeutic approaches, particularly anti-GD2 monoclonal antibodies, has significantly improved survival in high-risk disease. Emerging therapies such as targeted agents, radiopharmaceuticals, and cellular immunotherapies are further expanding the therapeutic landscape. Despite these advances, high-risk and relapsed neuroblastoma remain associated with substantial morbidity and mortality. Ongoing challenges include treatment resistance, long-term toxicity, and disparities in access to advanced therapies. Continued progress will depend on integrating molecular profiling into clinical decision-making, refining risk-adapted treatment strategies, and expanding international collaborative research efforts. This narrative review summarizes current knowledge on neuroblastoma epidemiology, biology, staging, and treatment, highlighting recent advances and future directions aimed at improving outcomes for affected children. Full article
Show Figures

Figure 1

48 pages, 3621 KB  
Review
Mining the Hidden Pharmacopeia: Fungal Endophytes, Natural Products, and the Rise of AI-Driven Drug Discovery
by Ruqaia Al Shami and Walaa K. Mousa
Int. J. Mol. Sci. 2026, 27(3), 1365; https://doi.org/10.3390/ijms27031365 - 29 Jan 2026
Abstract
Emerging from millions of years of evolutionary optimization, Natural products (NPs) remain unique, unparalleled sources of bioactive scaffolds. Unlike synthetic molecules engineered around single therapeutic targets, NPs often exhibit multi-target, system-level bioactivity, aligned with the principles of network pharmacology, which modulates pathways in [...] Read more.
Emerging from millions of years of evolutionary optimization, Natural products (NPs) remain unique, unparalleled sources of bioactive scaffolds. Unlike synthetic molecules engineered around single therapeutic targets, NPs often exhibit multi-target, system-level bioactivity, aligned with the principles of network pharmacology, which modulates pathways in a coordinated, non-disruptive manner. This approach reduces resistance, buffers compensatory feedback loops, and enhances therapeutic resilience. Fungal endophytes represent one of the most chemically diverse and biologically sophisticated NP reservoirs known, producing polyketides, alkaloids, terpenoids, and peptides with intricate three-dimensional architectures and emergent bioactivity patterns that remain exceptionally difficult to design de novo. Advances in artificial intelligence (AI), machine learning, deep learning, and multi-omics integration have redefined the discovery landscape, transforming previously intractable fungal metabolomes and cryptic biosynthetic gene clusters (BGCs) into tractable, predictable, and engineerable systems. AI accelerates genome mining, metabolomic annotation, BGC-metabolite linking, structure prediction, and activation of silent pathways. Generative AI and diffusion models now enable de novo design of NP-inspired scaffolds while preserving biosynthetic feasibility, opening new opportunities for direct evolution, pathway refactoring, and precision biomanufacturing. This review synthesizes the chemical and biosynthetic diversity of major NP classes from fungal endophytes and maps them onto the rapidly expanding ecosystem of AI-driven tools. We outline how AI transforms NP discovery from empirical screening into a predictive, hypothesis-driven discipline with direct industrial implications for drug discovery and synthetic biology. By coupling evolutionarily refined chemistry with modern computational intelligence, the field is poised for a new era in which natural-product leads are not only rediscovered but systematically expanded, engineered, and industrialized to address urgent biomedical and sustainability challenges. Full article
(This article belongs to the Section Bioactives and Nutraceuticals)
Show Figures

Figure 1

11 pages, 687 KB  
Review
Challenges in Balancing Hemostasis and Thrombosis in Therapy Tailoring for Hemophilia: A Narrative Review
by Gili Kenet, Sarina Levy-Mendelovich, Tami Livnat and Benjamin Brenner
Int. J. Mol. Sci. 2026, 27(3), 1373; https://doi.org/10.3390/ijms27031373 - 29 Jan 2026
Abstract
Hemostasis and thrombosis reflect a delicate balance, regulated by the interplay between procoagulant and anticoagulant mechanisms. Hemophilia is traditionally viewed as a bleeding disorder, but emerging evidence highlights the paradoxical risks of thrombosis in hemophilia patients. We explore the landscape of hemophilia management, [...] Read more.
Hemostasis and thrombosis reflect a delicate balance, regulated by the interplay between procoagulant and anticoagulant mechanisms. Hemophilia is traditionally viewed as a bleeding disorder, but emerging evidence highlights the paradoxical risks of thrombosis in hemophilia patients. We explore the landscape of hemophilia management, emphasizing challenges of balancing hemostasis in the context of aging, novel non-factor replacement therapies (NRTs), and comorbidity-driven thrombotic complications. Therapeutic approaches, including innovative NRTs, such as emicizumab, or rebalancing agents (e.g., concizumab, marstacimab, fitusiran), offer promising advancements in bleeding prophylaxis but may increase thrombotic risks. Conversely, novel anticoagulants, such as FXI inhibitors, offer potential thrombosis protection with minimal bleeding risk. Our review examines the impact of aging-related comorbidities, including cardiovascular disease, atrial fibrillation, HIV-associated complications, and acute coronary syndromes, on thrombotic risk in hemophilia patients. Evidence-based strategies for balancing hemostasis and thrombosis are outlined alongside experimental models, thrombin generation assays, and advancements in rebalancing coagulation through natural anticoagulant modulation. FXI inhibition emerges as a paradigm shift in thrombosis management, offering reduced bleeding risks while preserving vascular health. Finally, this review highlights the need for global laboratory assays to personalize treatments, emphasizing strategies to optimize safety and efficacy, particularly as hemophilia patients live longer with complex comorbidity profiles. Full article
(This article belongs to the Special Issue Hemophilia: From Pathophysiology to Novel Therapies)
Show Figures

Figure 1

17 pages, 642 KB  
Review
FGFR Testing in Metastatic Urothelial Carcinoma—Who, When, and How to Test
by André Mansinho, José Carlos Machado, Cátia Faustino, Arnaldo Figueiredo, João Moreira Pinto, Nuno Vau, João Ramalho-Carvalho and Manuel R. Teixeira
Cancers 2026, 18(3), 444; https://doi.org/10.3390/cancers18030444 - 29 Jan 2026
Abstract
Metastatic urothelial carcinoma (mUC) is a lethal cancer with limited therapeutic options. Advances in genomic and transcriptomic research have deepened the understanding of mUC biology, leading to the identification of clinically relevant molecular alterations that represent potential actionable targets. This has broadened the [...] Read more.
Metastatic urothelial carcinoma (mUC) is a lethal cancer with limited therapeutic options. Advances in genomic and transcriptomic research have deepened the understanding of mUC biology, leading to the identification of clinically relevant molecular alterations that represent potential actionable targets. This has broadened the treatment landscape of the disease to include novel agents, such as antibody–drug conjugates (e.g., enfortumab vedotin) and targeted therapies, including the pan-fibroblast growth factor receptor (FGFR) inhibitor erdafitinib. Genomic alterations in FGFR3 are well-established oncogenic drivers in bladder cancer and represent predictive biomarkers of response to FGFR-targeted therapies. The phase III THOR trial demonstrated the clinical benefit of erdafitinib in previously treated mUC patients harboring FGFR3 alterations and supported its subsequent approval by the European Medicines Agency. In this context, accurate molecular profiling is essential to guide patient selection for FGFR inhibitor therapy. Equally important is the standardization and timely implementation of FGFR3 testing in clinical practice to optimize treatment planning. This review addresses key considerations in FGFR3 testing in mUC and discusses how it can be routinely incorporated into clinical practice. Full article
(This article belongs to the Section Cancer Biomarkers)
15 pages, 706 KB  
Review
Multi-Omics Approaches to Understanding Therapy Resistance in Acute Lymphoblastic Leukemia
by Xiuyun Wu and Jingxin Zhang
Lymphatics 2026, 4(1), 8; https://doi.org/10.3390/lymphatics4010008 - 29 Jan 2026
Abstract
Therapy resistance remains a major cause of relapse and poor outcomes in acute lymphoblastic leukemia (ALL). Recent multi-omics studies in ALL have revealed that resistance arises from a combination of leukemia-specific genetic lesions, treatment-driven clonal evolution, and adaptive non-genetic programs. Genomic analyses have [...] Read more.
Therapy resistance remains a major cause of relapse and poor outcomes in acute lymphoblastic leukemia (ALL). Recent multi-omics studies in ALL have revealed that resistance arises from a combination of leukemia-specific genetic lesions, treatment-driven clonal evolution, and adaptive non-genetic programs. Genomic analyses have identified recurrent alterations associated with resistance to chemotherapy, tyrosine kinase inhibitors, and immunotherapies, while single-cell profiling has uncovered heterogeneous cell states that persist during treatment and contribute to minimal residual disease. Emerging epigenetic, proteomic, and metabolic data further indicate that reversible regulatory and signaling changes play a central role in leukemic persistence. Integrative analyses are beginning to define convergent resistance pathways and clinically relevant biomarkers, although longitudinal sampling and clinical translation remain limited. This review summarizes the current multi-omics landscape of therapy resistance in ALL and discusses opportunities to improve risk stratification and therapeutic strategies. Full article
(This article belongs to the Collection Acute Lymphoblastic Leukemia (ALL))
Show Figures

Figure 1

32 pages, 378 KB  
Review
Neuroprotective Herbs Associated with Parkinson’s and Alzheimer’s Disease
by Georgiana Smaranda Marțiș, Rodica Ana Ungur, Anamaria Pop, Evelina Maria Bordean, Claudia Pașca and Ileana Monica Borda
Nutrients 2026, 18(3), 439; https://doi.org/10.3390/nu18030439 - 29 Jan 2026
Abstract
There is currently no treatment for Parkinson’s (PD) and Alzheimer’s (AD) diseases, and medications that target the blockage of amyloid plaque cascades appear to be the most promising for preventing these diseases. However, it is believed that consuming natural antioxidants, particularly phytochemicals such [...] Read more.
There is currently no treatment for Parkinson’s (PD) and Alzheimer’s (AD) diseases, and medications that target the blockage of amyloid plaque cascades appear to be the most promising for preventing these diseases. However, it is believed that consuming natural antioxidants, particularly phytochemicals such as phenolic compounds, may help the treatment process for neurodegenerative illnesses. Phenolic substances such as phenolic acids, polyphenols, and flavonoids have been shown to have antioxidant properties in plants and are thought to have a similar impact in humans. This review provides an analysis of the current landscape of PD and AD pathophysiology, paying particular attention to phytochemical-based therapeutic, preventive, and management strategies using disclosed herb candidates in in vivo/vitro studies. We also highlight the herb-derived components that have recently been identified for their effects in the treatment of PD/AD to provide a review and perspectives for the development of the next generation of drugs and preparations for the treatment of PD/AD. Full article
18 pages, 6113 KB  
Article
Whole-Transcriptome Analysis of Gene Expression in Canine Splenic Lymphoid Hyperplasia, Complex Hyperplasia, Histiocytic Sarcoma, and Stromal Sarcoma
by Cleide Spröhnle-Barrera, Rachel Allavena and Chiara Palmieri
Animals 2026, 16(3), 422; https://doi.org/10.3390/ani16030422 - 29 Jan 2026
Abstract
Splenic nodules in dogs that were historically classified under the broad term “fibrohistiocytic nodules” are now recognised as distinct entities within likely a biological continuum. These include lymphoid hyperplasia extending to indolent lymphoma and complex hyperplasia to stromal sarcoma. However, the molecular mechanisms [...] Read more.
Splenic nodules in dogs that were historically classified under the broad term “fibrohistiocytic nodules” are now recognised as distinct entities within likely a biological continuum. These include lymphoid hyperplasia extending to indolent lymphoma and complex hyperplasia to stromal sarcoma. However, the molecular mechanisms underpinning these proposed progressions remain largely unexplored, particularly at the genomic and transcriptomic levels. This study aimed to delineate and compare the transcriptomic landscapes of four distinct canine splenic nodules through differential gene expression profiling. RNA sequencing was performed on twelve formalin-fixed, paraffin-embedded (FFPE) splenic tissue samples obtained from dogs diagnosed with lymphoid hyperplasia, complex hyperplasia, histiocytic sarcoma, and stromal sarcoma, with normal canine spleen serving as a control tissue. Comparative transcriptomic analysis identified 47 differentially expressed genes (DEGs) between splenic nodules and normal spleen, including CSRP1, SLC40A1, C1QA, C1QC, DLA-12, FTL, FXYD6, MPEG1, OAS3, CSF1, and JMJD6. Furthermore, 39 DEGs were significantly altered among the four splenic lesion types, such as MLC1, ERAS, MOV10L1, LOC102152143, COL4A1, COL4A2, COL12A1, NOTCH3, PLOD2, CPXM2, MRC1, GALNT5, TIMP1, and TFPI2. Many of these genes have previously been implicated in tumorigenesis and metastasis in other malignancies. These findings suggest that dysregulated gene expression may contribute to the activation of stromal cells and macrophages within the spleen, facilitating malignant transformation. Overall, these findings deliver novel transcriptomic insights into canine splenic tumorigenesis that may improve diagnostic precision, inform prognostic assessment, and support the development of targeted therapeutic strategies in veterinary oncology. Full article
(This article belongs to the Collection Genetic Diversity in Livestock and Companion Animals)
Show Figures

Figure 1

40 pages, 2561 KB  
Review
LncRNAs at the Crossroads of Precision Nutrition and Cancer Chemoprevention
by Camelia Munteanu, Revathy Nadhan, Sabina Turti, Eftimia Prifti, Larisa Achim, Sneha Basu, Alessandra Ferraresi, Ji Hee Ha, Ciro Isidoro and Danny N. Dhanasekaran
Cancers 2026, 18(3), 430; https://doi.org/10.3390/cancers18030430 - 29 Jan 2026
Abstract
Cancer remains a leading cause of morbidity and mortality worldwide, and effective strategies for cancer prevention are urgently needed to complement therapeutic advances. While dietary factors are known to influence cancer risk, the molecular mechanisms that mediate inter-individual responses to nutritional exposures remain [...] Read more.
Cancer remains a leading cause of morbidity and mortality worldwide, and effective strategies for cancer prevention are urgently needed to complement therapeutic advances. While dietary factors are known to influence cancer risk, the molecular mechanisms that mediate inter-individual responses to nutritional exposures remain poorly defined. Emerging evidence identifies long non-coding RNAs (lncRNAs) as pivotal regulators of gene expression, chromatin organization, metabolic homeostasis, immune signaling, and cellular stress responses, the core processes that drive cancer initiation and progression and are highly sensitive to nutritional status. In parallel, advances in precision nutrition have highlighted how variability in genetics, metabolism, microbiome composition, and epigenetic landscapes shape dietary influences on cancer susceptibility. This review integrates these rapidly evolving fields by positioning lncRNAs as molecular conduits that translate dietary exposures into transcriptional and epigenetic programs governing cancer development, progression, and therapeutic vulnerability. We provide mechanistic evidence demonstrating how dietary bioactive compounds and micronutrients, including polyphenols [such as curcumin, resveratrol, epigallocatechin gallate (EGCG)], flavonoids, alkaloids such as berberine, omega-3 (ω-3) fatty acids, folate, vitamin D, probiotic metabolites (such as butyrate and propionate), and trace elements (such as selenium and zinc), modulate oncogenic and tumor-suppressive lncRNAs. These nutrient–lncRNA interactions influence cancer-relevant pathways controlling proliferation, epithelial–mesenchymal transition (EMT), inflammation, oxidative stress, and metabolic rewiring. We further discuss emerging lncRNA signatures that reflect nutritional and metabolic states, their potential utility as biomarkers for individualized dietary interventions, and their integration into liquid biopsy platforms. Leveraging multi-omics datasets and systems biology, we outline AI-driven frameworks to map nutrient–lncRNA regulatory networks and identify targetable nodes for cancer chemoprevention. Finally, we address translational challenges, including compound bioavailability, inter-individual variability, and limited clinical validation, and propose future directions for incorporating lncRNA profiling into precision nutrition-guided cancer prevention trials. Together, these insights position lncRNAs at the nexus of diet and cancer biology and establish a foundation for mechanistically informed precision nutrition strategies in cancer chemoprevention. Full article
(This article belongs to the Special Issue Cancer Causes and Control)
Show Figures

Graphical abstract

14 pages, 585 KB  
Review
Uveal Melanoma: Biology, Prognostication, and Emerging Therapies to Outsmart an Immune-Cold Melanoma
by Danielle Brazel and Elizabeth Buchbinder
Cancers 2026, 18(3), 432; https://doi.org/10.3390/cancers18030432 - 29 Jan 2026
Abstract
Uveal melanoma (UM) is a rare but highly aggressive malignancy arising from melanocytes of the uveal tract. Despite high local control rates for primary disease, half of patients ultimately develop metastatic disease with historically dismal outcomes. Unlike cutaneous melanoma, UM is characterized by [...] Read more.
Uveal melanoma (UM) is a rare but highly aggressive malignancy arising from melanocytes of the uveal tract. Despite high local control rates for primary disease, half of patients ultimately develop metastatic disease with historically dismal outcomes. Unlike cutaneous melanoma, UM is characterized by a low tumor mutational burden, distinct driver mutations, and an immunosuppressive tumor microenvironment which together limit the efficacy of immune checkpoint inhibitors. Over the past decade, major advancements in molecular classification, prognostication, and therapeutic development have reshaped the clinical landscape for some patients with UM. This review synthesizes the current understanding of UM epidemiology, characteristics, prognostic biomarkers, immune biology, and contemporary management for both localized and metastatic disease. While survival gains remain modest, the rapid expansion of biologically informed and immune-based strategies offers cautious optimism for improving outcomes in this historically treatment-refractory disease. Full article
(This article belongs to the Special Issue Precision Oncology for Rare Skin Cancers)
Show Figures

Figure 1

45 pages, 4368 KB  
Review
Interactions Between Nutraceuticals and α-Synuclein Conformational States: Molecular Mechanisms and Neuroprotective Implications in Parkinson’s Disease
by Bruna Amenta, Rosalba Minervini, Maria Laura Matrella and Tiziana Cocco
Int. J. Mol. Sci. 2026, 27(3), 1324; https://doi.org/10.3390/ijms27031324 - 28 Jan 2026
Abstract
Synucleinopathies, including Parkinson’s disease (PD), are neurodegenerative disorders characterized by aberrant aggregation of α-synuclein (α-syn), a presynaptic protein with an intrinsic disorder nature. The transition of soluble monomers into oligomeric and fibrillar species represents a key molecular event driving neuronal dysfunction and neurodegeneration. [...] Read more.
Synucleinopathies, including Parkinson’s disease (PD), are neurodegenerative disorders characterized by aberrant aggregation of α-synuclein (α-syn), a presynaptic protein with an intrinsic disorder nature. The transition of soluble monomers into oligomeric and fibrillar species represents a key molecular event driving neuronal dysfunction and neurodegeneration. Emerging evidence suggests that nutraceuticals, bioactive compounds derived from dietary sources, can modulate α-syn aggregation at multiple conformational stages. Polyphenols, alkaloids, ginsenosides, and food-derived peptides interfere with α-syn structure and assembly, suppressing the formation of toxic oligomer species and promoting the clearance of misfolded assemblies. Despite this potential, clinical translational of nutraceuticals is currently limited by poor systemic bioavailability and restricted central nervous system penetration due to blood–brain barrier constraints, which have largely confined research to preclinical studies. In this context, this review summarizes current knowledge of nutraceutical interventions targeting the conformational landscape of α-syn and highlighting both direct and indirect molecular mechanisms with involved in aggregation-prone species. Furthermore, we critically examine key challenges related to bioavailability and clinical translation, focusing on advanced delivery systems and precision-based approaches to enhance neuroprotective efficacy and support the potential of nutraceuticals as novel or adjunctive therapeutic strategies for PD. Full article
(This article belongs to the Special Issue The Role of Natural Products in Drug Discovery: 2nd Edition)
Show Figures

Figure 1

16 pages, 1523 KB  
Review
ABCB5: A Key Regulator Linking Stem Cell Plasticity, Tumor Microenvironment, and Therapy Resistance in Cutaneous Melanoma
by Andreea Cătălina Tinca, Adrian Horațiu Sabău, Andreea Raluca Cozac-Szoke, Diana Maria Chiorean, Bianca Andreea Lazar, Raluca-Diana Hagău, Iuliu Gabriel Cocuz, Raluca Niculescu, Irina Bianca Kosovski, Sofia Teodora Muntean, Sabin Gligore Turdean and Ovidiu Simion Cotoi
Cancers 2026, 18(3), 424; https://doi.org/10.3390/cancers18030424 - 28 Jan 2026
Viewed by 1
Abstract
Cutaneous melanoma is one of the most aggressive skin cancers. Over the years, multiple studies have focused on identifying novel treatment strategies, with increasing attention directed toward immune-modulating mechanisms within the tumor microenvironment. Among these, ATP-binding cassette transporters and stem-associated pathways have been [...] Read more.
Cutaneous melanoma is one of the most aggressive skin cancers. Over the years, multiple studies have focused on identifying novel treatment strategies, with increasing attention directed toward immune-modulating mechanisms within the tumor microenvironment. Among these, ATP-binding cassette transporters and stem-associated pathways have been shown to influence drug response and immune escape. ABCB5 is a gene with multiple isoforms that significantly influences the immune response. In melanoma, the ABCB5α isoform is predominantly expressed, particularly in tumor stem-like cells where it promotes chemoresistance through active drug efflux. ABCB5 has also been linked to the regulation of PI3K/Akt, BCL-2, and miR-145-associated pathways. Moreover, ABCB5-positive cells contribute to the formation of an immunosuppressive microenvironment by secreting cytokines (IL-6, IL-8, TGF-β) and expressing immune checkpoint ligands, such as PD-L1, thereby favoring tumor progression and a poor prognosis. This review integrates current data on the molecular and microenvironmental mechanisms underlying melanoma progression and therapy resistance, and positions ABCB5 within the broader landscape of melanoma resistance mechanisms, emphasizing both its potential and its current limitations as a biomarker and therapeutic target. Full article
(This article belongs to the Section Cancer Pathophysiology)
Show Figures

Figure 1

40 pages, 9917 KB  
Review
A Novel Paradigm for Targeting Challenging Targets: Advancing Technologies and Future Directions of Molecular Glue Degraders
by Yifan Zhang, Linlin Li, Jiajia Xu, Chunchen Che, Jiaqing Jia, Haohao Lu, Qidong You and Xiaoli Xu
Molecules 2026, 31(3), 459; https://doi.org/10.3390/molecules31030459 - 28 Jan 2026
Viewed by 19
Abstract
Molecular glue degraders (MGDs) constitute a class of innovative therapeutic agents within the field of targeted protein degradation (TPD). In contrast to proteolysis-targeting chimeras (PROTACs), MGDs induce protein degradation by stabilizing the interaction between an E3 ubiquitin ligase and a target protein. They [...] Read more.
Molecular glue degraders (MGDs) constitute a class of innovative therapeutic agents within the field of targeted protein degradation (TPD). In contrast to proteolysis-targeting chimeras (PROTACs), MGDs induce protein degradation by stabilizing the interaction between an E3 ubiquitin ligase and a target protein. They typically exhibit favorable drug-like characteristics, including lower molecular weight and enhanced bioavailability. Although their discovery was historically serendipitous, recent advances in high-throughput screening, bioinformatics, and artificial intelligence are enabling more systematic identification and optimization. To date, three MGD-based drugs have been approved for clinical use, with numerous candidates under active investigation. This review comprehensively traces the technological progression of MGDs from serendipitous discovery to the current era of rational design. We systematically introduce and critically evaluate strategies for discovering MGDs, accompanied by illustrative examples. Concurrently, we discuss the major challenges hindering the broader application of MGDs and propose potential approaches to address these issues. Finally, we outline prospective research directions in the field. This review aims to provide a holistic framework for understanding the past, present, and future of molecular glue degraders, underscoring their significant potential to reshape the landscape of drug discovery. Full article
(This article belongs to the Special Issue Innovations and Breakthroughs in Bifunctional Molecules)
Show Figures

Graphical abstract

38 pages, 839 KB  
Review
Ex Vivo Treatment Response Prediction in Multiple Myeloma: Assay Formats, Clinical Correlation, and Future Directions
by Gavin R. Oliver, Carlton C. Barnett, Kendra E. Hightower, Yubin Kang and Muhamed Baljevic
Cancers 2026, 18(3), 411; https://doi.org/10.3390/cancers18030411 - 28 Jan 2026
Viewed by 40
Abstract
Ex vivo functional testing for multiple myeloma is rapidly evolving, yet no single assay has reached the level of reliability and clinical utility needed for routine decision-making. Existing approaches generally fall into three categories: 2D cultures, 3D models, and dynamic systems. Each contributes [...] Read more.
Ex vivo functional testing for multiple myeloma is rapidly evolving, yet no single assay has reached the level of reliability and clinical utility needed for routine decision-making. Existing approaches generally fall into three categories: 2D cultures, 3D models, and dynamic systems. Each contributes valuable but incomplete insight into therapeutic response. Among these, 2D assays remain the most mature, with the most extensive clinical correlations to date, though their simplified architecture limits their ability to reflect the full complexity of the bone marrow microenvironment. However, 3D systems, including spheroids and matrix-based organoids, offer improved preservation of tumor heterogeneity and microenvironmental cues. These platforms show emerging clinical relevance and may hold advantages over traditional 2D formats, and validation efforts are developing. Dynamic systems, including microfluidic models and perfused bone-marrow mimetics, represent the most physiologically ambitious category, yet their technical intricacy and early stage of development have so far limited broad clinical correlation. Altogether, the current landscape highlights substantial progress but lacks an optimal assay. In this review, we take the unique approach of examining published ex vivo tests that have demonstrated a level of clinical correlation. We evaluate their respective formats, strengths and limitations, and discuss considerations for what an ideal future assay may encompass. Full article
(This article belongs to the Special Issue Clinical Trials and Translational Research in Multiple Myeloma)
Show Figures

Figure 1

16 pages, 1334 KB  
Review
Targeting Macrophages in Immunotherapy: The Ascent of CAR-Macrophages
by Vinod Nadella and Anu Sharma
Int. J. Mol. Sci. 2026, 27(3), 1292; https://doi.org/10.3390/ijms27031292 - 28 Jan 2026
Viewed by 54
Abstract
Chimeric antigen receptor (CAR)-engineered immune cell therapies have revolutionized cancer treatment, with CAR-T cells demonstrating remarkable efficacy against hematological malignancies. However, the effectiveness of CAR-T and other lymphocyte-based therapies against solid tumors remains limited, primarily due to the immunosuppressive tumor microenvironment and poor [...] Read more.
Chimeric antigen receptor (CAR)-engineered immune cell therapies have revolutionized cancer treatment, with CAR-T cells demonstrating remarkable efficacy against hematological malignancies. However, the effectiveness of CAR-T and other lymphocyte-based therapies against solid tumors remains limited, primarily due to the immunosuppressive tumor microenvironment and poor infiltration of effector cells. Recently, CAR-macrophage (CAR-M) immunotherapy has emerged as a promising strategy to overcome these barriers. Leveraging the innate tumor-homing ability, phagocytic function, and antigen-presenting capacity of macrophages, CAR-M therapies offer unique advantages for targeting solid tumors. This review provides a comprehensive overview of the development and current state of CAR-Macrophage immunotherapy, including advances in CAR design and macrophage engineering, preclinical and clinical progress, and mechanistic insights into their anti-tumor activity. The review critically examined both the benefits and limitations of CAR-M approaches, addressing persistent challenges such as cell sourcing, durability, and safety, while also exploring innovative strategies to enhance therapeutic efficacy. Finally, future perspectives and the potential clinical impact of CAR-macrophage therapies were outlined, underscoring their emerging role in the evolving landscape of cancer immunotherapy. Full article
(This article belongs to the Special Issue Advances in Targeting Macrophages in Immunotherapy)
Show Figures

Figure 1

26 pages, 857 KB  
Review
Metabolic Messengers: Extracellular Vesicles as Central Mediators of Metabolic Reprogramming in Renal Cell Cancer
by Qingshu Meng, Liqun Huang, Zhiguo Chen, Rui Lin, Xiaohui Zhou and Guoshen Yang
Biomedicines 2026, 14(2), 282; https://doi.org/10.3390/biomedicines14020282 - 27 Jan 2026
Viewed by 95
Abstract
Renal cell carcinoma (RCC) has been described as a metabolic disease as metabolic alterations are common in disparate RCC etiologies. Extracellular vesicles (EVs), the lipid bilayer-enclosed nanoparticles secreted by all living cells, have emerged as crucial mediators of intercellular and inter-organ communication, capable [...] Read more.
Renal cell carcinoma (RCC) has been described as a metabolic disease as metabolic alterations are common in disparate RCC etiologies. Extracellular vesicles (EVs), the lipid bilayer-enclosed nanoparticles secreted by all living cells, have emerged as crucial mediators of intercellular and inter-organ communication, capable of shuttling functional proteins, lipids, and nucleic acids between cells. This review summarizes the essential events in tumor-associated metabolic reprogramming with a particular focus on renal cancers. We further explore how EVs released by metabolically deranged cells in cancer with altered cargos reprogram the renal cellular landscape, fostering tumor initiation, proliferation, angiogenesis, immune evasion, and therapy resistance. Understanding this EV-mediated axis not only elucidates the pathophysiological link between these conditions but also helps to unveil novel potential therapeutic targets for RCC patients. Full article
Back to TopTop