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

Journals

Article Types

Countries / Regions

Search Results (144)

Search Parameters:
Keywords = molecularly targeted drugs

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
29 pages, 1395 KiB  
Review
Advances and Challenges in Targeted Therapy and Its Combination Strategies for Leukemia
by Zhiyuan Zhong, Ran Yao, Yifei Duan, Cheng Ouyang, Zefan Du, Lindi Li, Hailin Zou, Yong Liu, Hongman Xue, Liang Li and Chun Chen
Biomedicines 2025, 13(7), 1652; https://doi.org/10.3390/biomedicines13071652 - 7 Jul 2025
Viewed by 740
Abstract
Leukemia is a group of hematological malignancies with a complex pathogenesis and diverse clinical manifestations. Although traditional treatments such as chemotherapy, radiotherapy, and hematopoietic stem cell transplantation have improved patient outcomes, their efficacy is often limited by non-specificity, drug resistance, and relapse. In [...] Read more.
Leukemia is a group of hematological malignancies with a complex pathogenesis and diverse clinical manifestations. Although traditional treatments such as chemotherapy, radiotherapy, and hematopoietic stem cell transplantation have improved patient outcomes, their efficacy is often limited by non-specificity, drug resistance, and relapse. In recent years, targeted therapy has emerged as a major breakthrough, offering new opportunities for precision medicine in leukemia. The development of molecularly targeted agents has significantly advanced our ability to treat specific leukemia subtypes. However, challenges such as resistance to targeted drugs, adverse effects, and tumor heterogeneity remain significant obstacles. As a result, treatment strategies are shifting from single-agent chemotherapy toward combination therapies that integrate targeted agents, aiming to enhance therapeutic efficacy and reduce the likelihood of resistance. This review summarizes the current research landscape, clinical applications, and limitations of targeted therapies in leukemia, with a focus on recent progress in combination treatment strategies and ongoing clinical trials. Full article
Show Figures

Figure 1

26 pages, 1382 KiB  
Review
Drug-Induced Sarcoid-like Reactions Associated to Targeted Therapies and Biologic Agents
by Federica Andolfi, Luca Caffarri, Matilde Neviani, Silvia Rubini, Dario Andrisani, Filippo Gozzi, Bianca Beghé, Enrico Clini, Roberto Tonelli and Stefania Cerri
Diagnostics 2025, 15(13), 1658; https://doi.org/10.3390/diagnostics15131658 - 29 Jun 2025
Viewed by 883
Abstract
Background: Sarcoidosis is a multisystem inflammatory disease characterized by the immune-mediated formation of non-necrotizing epithelioid granulomas. Several commonly used medications can induce similar granulomatous reactions, known as drug-induced sarcoid-like reactions (DISRs), which closely mimic sarcoidosis. Despite their specificity in targeting molecular pathways, [...] Read more.
Background: Sarcoidosis is a multisystem inflammatory disease characterized by the immune-mediated formation of non-necrotizing epithelioid granulomas. Several commonly used medications can induce similar granulomatous reactions, known as drug-induced sarcoid-like reactions (DISRs), which closely mimic sarcoidosis. Despite their specificity in targeting molecular pathways, certain therapies—particularly targeted treatments—have increasingly been linked to DISRs. Methods: This narrative review was based on a PubMed search using the terms “SARCOID LIKE REACTION” and “DRUG”. A cross-check was performed with “SARCOID” combined with each identified drug to identify misclassified cases. Drugs with limited evidence or weak pathogenetic plausibility were excluded, leaving only molecularly targeted therapies for consideration. Sources included case reports, case series, and reviews selected based on their clinical and scientific relevance, without any restrictions on time or language. Results: In light of the available data, five main pharmacological groups were found to be associated to DISR: immune checkpoint inhibitors, TNF-α antagonists, BRAF inhibitors, monoclonal antibodies, and miscellaneous agents. Each group has distinct mechanisms of action and clinical indications, which likely affect the frequency, presentation, and timing of DISRs. Conclusions: Diagnosing DISRs is challenging, and a structured approach is crucial for differentiating them from other conditions. To support clinicians, we propose a diagnostic algorithm to guide decision-making in suspected cases. Management should be individualized, as most DISRs either resolve spontaneously or improve after the discontinuation of the causative drug. Important factors influencing therapeutic decisions include the severity of the underlying disease, the availability of alternative treatments, and the extent of DISR manifestations. Full article
(This article belongs to the Special Issue Sarcoidosis: From Diagnosis to Management)
Show Figures

Figure 1

36 pages, 3205 KiB  
Review
Multidrug Resistance: Are We Still Afraid of the Big Bad Wolf
by Abdulelah Alhazza, Adenike Oyegbesan, Emira Bousoik and Hamidreza Montazeri Aliabadi
Pharmaceuticals 2025, 18(6), 895; https://doi.org/10.3390/ph18060895 - 14 Jun 2025
Viewed by 993
Abstract
After the era of multidrug resistance (MDR) against cytotoxic chemotherapy, the development of resistance against newly developed molecularly targeted drugs also seems inevitable. While the mechanisms involved in resistance against these two categories of anticancer drugs are different, the principles are similar: inherent [...] Read more.
After the era of multidrug resistance (MDR) against cytotoxic chemotherapy, the development of resistance against newly developed molecularly targeted drugs also seems inevitable. While the mechanisms involved in resistance against these two categories of anticancer drugs are different, the principles are similar: inherent resistance (also known as primary resistance) is a result of heterogeneity in cancer cells where a subpopulation of the cells do not show a favorable initial response to the drug, while acquired resistance (or secondary resistance), as the name suggests, is developed after repeated treatments due to the plasticity of cancer cells. Despite the introduction of a variety of molecularly targeted drugs to clinical practice, chemotherapy is still at the forefront of the battle against cancer. In this manuscript, we review the major mechanisms involved in MDR and resistance against different categories of molecularly targeted drugs separately, and review some of the strategies studied to overcome the resistance against cancer therapy. While MDR mechanisms have been reviewed previously, the molecular mechanisms of resistance to the latest generations of anticancer drugs are rarely reviewed as a group, and the connection between the two categories of resistance is often missing in this type of publication. Our aim is to illustrate a comprehensive picture of what the landscape of cancer treatment is today with respect to resistance. While this picture seems bleak, and it is the common belief that resistance is inevitable, understanding the mechanisms involved could potentially lead to more efficient approaches to overcoming this so far unbeatable obstacle. Full article
(This article belongs to the Special Issue Drug Resistance Against Cancer Treatment)
Show Figures

Figure 1

18 pages, 1252 KiB  
Review
Precision Oncology Framework Using Circulating Tumor Cells
by Fumihiko Kakizaki, Kyoichi Oshiro, Yuya Enoki, Kana Kawanishi, Norikazu Masuda, Hisatsugu Maekawa, Jun Matsubayashi, Masahiro Kawashima, Hiroyuki Miyoshi, Yukitoshi Takemura and Kazutaka Obama
Int. J. Mol. Sci. 2025, 26(12), 5539; https://doi.org/10.3390/ijms26125539 - 10 Jun 2025
Viewed by 1213
Abstract
Circulating tumor cells (CTCs) are multifaceted biomarkers with significant potential for precision oncology, offering opportunities to refine diagnoses and personalize treatments across various cancer types, including colorectal and breast cancer. CTC assays serve as reliable prognostic indicators, even during chemotherapy and/or molecularly targeted [...] Read more.
Circulating tumor cells (CTCs) are multifaceted biomarkers with significant potential for precision oncology, offering opportunities to refine diagnoses and personalize treatments across various cancer types, including colorectal and breast cancer. CTC assays serve as reliable prognostic indicators, even during chemotherapy and/or molecularly targeted therapies. Notably, CTCs exhibit heterogeneity that gradually develops during carcinogenesis and becomes more pronounced in advanced disease stages. These intra- and intertumoral heterogeneities pose challenges, particularly when drug-resistant clones emerge following therapy. The dynamic behavior of CTCs provides valuable insights into treatment response and prognosis. Extensive efforts have led to the development of technologies for effective CTC isolation, accelerating their clinical implementation. While both CTC and circulating tumor DNA (ctDNA) tests offer prognostic value, they reflect different aspects of tumor biology: CTC counts indicate tumor progression, while ctDNA levels correlate with tumor burden. The combined analysis is expected to yield complementary insights. CTC tests are feasible in general hospitals and may serve as tumor markers comparable to, or even superior to, conventional markers such as carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9) for colorectal cancer, and CA15-3 for breast cancer. Early incorporation of CTC tests into routine blood panels appears to be a rational and promising approach. Full article
Show Figures

Figure 1

15 pages, 304 KiB  
Review
Improving the Dosing Schedules of Targeted Anticancer Agents
by Dominique Levêque
Pharmaceuticals 2025, 18(6), 848; https://doi.org/10.3390/ph18060848 - 6 Jun 2025
Viewed by 755
Abstract
Beyond developing new agents, cancer treatment can also be optimized by modifying the dosing regimen of approved drugs. Academic teams have experimented with different ways of improving drug regimens, leading to off-label practices for therapeutic and/or economic purposes, and currently, drug regulatory agencies [...] Read more.
Beyond developing new agents, cancer treatment can also be optimized by modifying the dosing regimen of approved drugs. Academic teams have experimented with different ways of improving drug regimens, leading to off-label practices for therapeutic and/or economic purposes, and currently, drug regulatory agencies have begun to reappraise this often-neglected topic. This concept also considers the patient’s perspective in terms of quality of life and convenience, including the concept of time toxicity. Overall, the optimization of drug dosing of anticancer agents may be viewed on three sides: the improvement of the benefits/risks balance (patient), the improvement of the convenience of the treatment (patient, healthcare professionals), and the mitigation of the financial impact (health insurance, patient). Examples of dose reassessments of targeted therapies (approved since 1997) are chosen to illustrate the context. Suboptimal/overdosed regimens are found for certain molecularly targeted agents, mostly based on the ancient concept of maximum tolerated dose in oncology. This underlines the lack of comparative effective dose trials before approval. Fortunately, dosing regimens of newly approved molecularly targeted agents is going to evolve with the hope of more convenient and better tolerated treatments. This optimization will bring greater benefit to patients and to healthcare professionals but without addressing the economic issue. Full article
(This article belongs to the Special Issue Therapeutic Drug Monitoring and Adverse Drug Reactions: 2nd Edition)
20 pages, 3643 KiB  
Article
High-Throughput Screens of Repurposing Hub and DOS Chemical Libraries Reveal Compounds with Novel and Potent Inhibitory Activity Against the Essential Non-Neuronal Acetylcholinesterase of Schistosoma mansoni (SmTAChE)
by Patrick J. Skelly and Akram A. Da’dara
Int. J. Mol. Sci. 2025, 26(11), 5415; https://doi.org/10.3390/ijms26115415 - 5 Jun 2025
Viewed by 473
Abstract
Schistosomiasis is a parasitic disease caused by helminth parasites of the genus Schistosoma, affecting >200 million people worldwide. Current schistosomiasis treatment relies on a single drug, praziquantel, highlighting the urgent need for new therapies. We have identified a non-neuronal tegumental acetylcholinesterase from [...] Read more.
Schistosomiasis is a parasitic disease caused by helminth parasites of the genus Schistosoma, affecting >200 million people worldwide. Current schistosomiasis treatment relies on a single drug, praziquantel, highlighting the urgent need for new therapies. We have identified a non-neuronal tegumental acetylcholinesterase from Schistosoma mansoni (SmTAChE) as a rational and molecularly defined drug target. Molecular modeling reveals significant structural differences between SmTAChE and human AChE, suggesting the potential for identifying parasite-specific inhibitors. Here, we screened recombinant SmTAChE (rSmTAChE) against two chemical libraries: the Broad Institute Drug Repurposing Hub (5440 compounds) and the Diversity-Oriented Synthesis (DOS)-A library (3840 compounds). High-throughput screening identified 116 hits from the Repurposing Hub (2.13% hit rate) and 44 from the DOS-A (1.14% hit rate) library that inhibited rSmTAChE ≥60% at 20 µM. Dose–response assays using both rSmTAChE and recombinant human AChE (rHsAChE) revealed 19 Repurposing Hub compounds (IC50: 0.4–24 µM) and four DOS-A scaffolds (IC50: 13–29 µM), with higher selectivity for rSmTAChE. Selective inhibitors such as cepharanthine, primaquine, mesalazine, and embelin emerged as promising candidates for further evaluation in schistosomiasis treatment. These 23 newly identified selective hits provide a foundation for the further development of novel anti-schistosome therapies. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

19 pages, 314 KiB  
Review
Current Status of Precision Medicine in Colorectal Cancer in Japan
by Yoshiki Kojitani and Masayuki Takeda
Int. J. Mol. Sci. 2025, 26(11), 5029; https://doi.org/10.3390/ijms26115029 - 23 May 2025
Viewed by 1203
Abstract
Colorectal cancer (CRC) remains a major health burden in Japan, with precision medicine playing an increasingly critical role in treatment optimization. Key biomarkers, including RAS, BRAF, microsatellite instability/mismatch repair, and human epidermal growth factor receptor 2, can be used as a [...] Read more.
Colorectal cancer (CRC) remains a major health burden in Japan, with precision medicine playing an increasingly critical role in treatment optimization. Key biomarkers, including RAS, BRAF, microsatellite instability/mismatch repair, and human epidermal growth factor receptor 2, can be used as a guide for molecularly targeted therapies and immunotherapy. Advances in molecular diagnostics, including comprehensive genomic profiling, have enabled more precise treatment selection such as RET and NTRK fusions. Nationwide initiatives, such as c-CAT and SCRUM-Japan, can leverage real-world data to refine clinical strategies. Recent developments in circulating tumor DNA analysis have led to novel approaches for minimal residual disease monitoring, as demonstrated by the CIRCULATE-Japan GALAXY study. However, certain challenges persist, including the time required for genetic testing, the limited availability of targeted therapies, and disparities in access to molecular tumor boards. This review summarizes the current landscape of precision medicine in CRC in Japan, emphasizing key biomarkers, genetic testing strategies, targeted therapies, and emerging technologies. Future research should focus on expanding clinical trial access, accelerating drug approvals, and integrating real-world data into clinical practice to further advance precision medicine. Full article
14 pages, 1625 KiB  
Communication
Last Resort? Rationale for Comprehensive Molecular Analysis in Treatment-Refractory R/M HNSCC: A Case Report of Remarkable Response to Sacituzumab Govitecan Following Molecular and Functional Characterization
by Henrike Barbara Zech, Philippe Schafhausen, Leonie Ramke, Janna-Lisa Velthaus, Simon Kreutzfeldt, Daniel Hübschmann, Kai Rothkamm, Carsten Bokemeyer, Anna Sophie Hoffmann, Stefan Fröhling, Hanno Glimm, Christian Stephan Betz, Malte Kriegs and Maximilian Christopeit
Biomedicines 2025, 13(5), 1266; https://doi.org/10.3390/biomedicines13051266 - 21 May 2025
Viewed by 741
Abstract
Background/Objectives: In recurrent/metastatic head and neck squamous cell carcinoma (R/M HNSCC), the overall prognosis is poor, and systemic treatment options remain limited. While precision therapy approaches have revolutionized treatment strategies in several tumor types, molecularly informed therapies in R/M HNSCC are rare, [...] Read more.
Background/Objectives: In recurrent/metastatic head and neck squamous cell carcinoma (R/M HNSCC), the overall prognosis is poor, and systemic treatment options remain limited. While precision therapy approaches have revolutionized treatment strategies in several tumor types, molecularly informed therapies in R/M HNSCC are rare, primarily due to the low number of actionable genetic alterations identified through next-generation sequencing (NGS) panels. There is an urgent need to establish precision therapy approaches in R/M HNSCC using innovative predictive testing. Methods: We report the case of a 43-year-old patient with recurrent oral cancer who was extensively pretreated and comprehensively characterized using both descriptive and functional testing. Results: NGS revealed no targetable alterations. A tumor tissue slice radiosensitivity assay suggested radioresistance, arguing against re-irradiation. Kinome profiling identified upregulated Src-family kinases (SFK), and SFK inhibition reduced kinase activity in vitro. Most notably, mRNA analysis demonstrated high Trop-2 overexpression, confirmed by immunohistochemistry (3+ in 100% of tumor cells). Following six cycles of the Trop-2-directed antibody–drug conjugate Sacituzumab govitecan (SG), the patient had an impressive clinical response. Conclusions: Tumor characterization beyond genetic profiling can identify novel treatment options in therapy-refractory HNSCC. This is the first report of “real-world” data on promising antitumor efficacy of SG in a heavily pretreated oral cancer patient with Trop-2 overexpression. Consistent with the findings of the Basket TROPiCS-03 study, SG appears to be a promising novel therapy option for R/M HNSCC after failure of immunotherapy and chemotherapy, particularly in patients with Trop-2 overexpression. Full article
(This article belongs to the Special Issue Novel Approaches towards Targeted Head and Neck Cancer Therapies)
Show Figures

Figure 1

48 pages, 7068 KiB  
Review
Colorimetric Molecularly Imprinted Polymer-Based Sensors for Rapid Detection of Organic Compounds: A Review
by Juan Carlos Bravo-Yagüe, Gema Paniagua-González, Rosa María Garcinuño, Asunción García-Mayor and Pilar Fernández-Hernando
Chemosensors 2025, 13(5), 163; https://doi.org/10.3390/chemosensors13050163 - 4 May 2025
Cited by 6 | Viewed by 2812
Abstract
This review offers a comprehensive examination of the development and current state of the art in the field of molecularly imprinted polymer (MIP)-based colorimetric sensors, focusing on their potential for the rapid detection of organic compounds. These MIP-sensors are gaining considerable attention due [...] Read more.
This review offers a comprehensive examination of the development and current state of the art in the field of molecularly imprinted polymer (MIP)-based colorimetric sensors, focusing on their potential for the rapid detection of organic compounds. These MIP-sensors are gaining considerable attention due to their distinctive capacity to modify sensor surfaces by creating recognition cavities within the polymer matrix, providing a versatile and highly selective platform for detecting a broad spectrum of analytes. This review systematically examines different types of MIP-based colorimetric sensors, attending to the target analyte, highlighting their applications in on-site sample detection, drug monitoring, environmental analysis, and food safety detection. The integration of novel technologies, such as nanozymes and smartphone-based detection, which enhance the capabilities of colorimetric MIP sensors, is also addressed. The sensors are particularly valuable due to their low cost, rapid response times, portability, and ease of use. Finally, the review outlines the future challenges for the development of MIP-based colorimetric sensors, focusing on overcoming existing limitations, improving sensor performance, and expanding their applications across various fields. Full article
Show Figures

Figure 1

20 pages, 2258 KiB  
Review
A Comprehensive Review of Nanoparticle-Based Drug Delivery for Modulating PI3K/AKT/mTOR-Mediated Autophagy in Cancer
by Md Ataur Rahman, Maroua Jalouli, Sujay Kumar Bhajan, Mohammed Al-Zharani and Abdel Halim Harrath
Int. J. Mol. Sci. 2025, 26(5), 1868; https://doi.org/10.3390/ijms26051868 - 21 Feb 2025
Cited by 2 | Viewed by 2300
Abstract
The phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of the rapamycin (mTOR) pathway plays a crucial role in the regulation of autophagy, a cellular mechanism vital for homeostasis through the degradation of damaged organelles and proteins. The dysregulation of this pathway is significantly associated with cancer [...] Read more.
The phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of the rapamycin (mTOR) pathway plays a crucial role in the regulation of autophagy, a cellular mechanism vital for homeostasis through the degradation of damaged organelles and proteins. The dysregulation of this pathway is significantly associated with cancer progression, metastasis, and resistance to therapy. Targeting the PI3K/AKT/mTOR signaling pathway presents a promising strategy for cancer treatment; however, traditional therapeutics frequently encounter issues related to nonspecific distribution and systemic toxicity. Nanoparticle-based drug delivery systems represent a significant advancement in addressing these limitations. Nanoparticles enhance the bioavailability, stability, and targeted delivery of therapeutic agents, facilitating the precise modulation of autophagy in cancer cells. Functionalized nanoparticles, such as liposomes, polymeric nanoparticles, and metal-based nanocarriers, facilitate targeted drug delivery to tumor tissues, minimizing off-target effects and improving therapeutic efficacy. These systems can deliver multiple agents concurrently, enhancing the modulation of PI3K/AKT/mTOR-mediated autophagy and related oncogenic pathways. This review examines advancements in nanoparticle-mediated drug delivery that target the PI3K/AKT/mTOR pathway, emphasizing their contribution to improving precision and minimizing side effects in cancer therapy. The integration of nanotechnology with molecularly targeted therapies presents substantial potential for addressing drug resistance. Future initiatives must prioritize the optimization of these systems to enhance clinical translation and patient outcomes. Full article
Show Figures

Figure 1

35 pages, 1042 KiB  
Review
Optimizing Osimertinib for NSCLC: Targeting Resistance and Exploring Combination Therapeutics
by Yan-You Liao, Chia-Luen Tsai and Hsiang-Po Huang
Cancers 2025, 17(3), 459; https://doi.org/10.3390/cancers17030459 - 29 Jan 2025
Cited by 2 | Viewed by 4902
Abstract
Non-small-cell lung cancer (NSCLC) is a leading cause of cancer-related deaths worldwide, with epidermal growth factor receptor (EGFR) mutations present in a substantial proportion of patients. Third-generation EGFR tyrosine kinase inhibitors (EGFR TKI), exemplified by osimertinib, have dramatically improved outcomes by effectively targeting [...] Read more.
Non-small-cell lung cancer (NSCLC) is a leading cause of cancer-related deaths worldwide, with epidermal growth factor receptor (EGFR) mutations present in a substantial proportion of patients. Third-generation EGFR tyrosine kinase inhibitors (EGFR TKI), exemplified by osimertinib, have dramatically improved outcomes by effectively targeting the T790M mutation—a primary driver of acquired resistance to earlier-generation EGFR TKI. Despite these successes, resistance to third-generation EGFR TKIs inevitably emerges. Mechanisms include on-target mutations such as C797S, activation of alternative pathways like MET amplification, histologic transformations, and intricate tumor microenvironment (TME) alterations. These resistance pathways are compounded by challenges in tolerability, adverse events, and tumor heterogeneity. In light of these hurdles, this review examines the evolving landscape of combination therapies designed to enhance or prolong the effectiveness of third-generation EGFR TKIs. We explore key strategies that pair osimertinib with radiotherapy, anti-angiogenic agents, immune checkpoint inhibitors, and other molecularly targeted drugs, and we discuss the biological rationale, preclinical evidence, and clinical trial data supporting these approaches. Emphasis is placed on how these combinations may circumvent diverse resistance mechanisms, improve survival, and maintain a favorable safety profile. By integrating the latest findings, this review aims to guide clinicians and researchers toward more individualized and durable treatment options, ultimately enhancing both survival and quality of life for patients with EGFR-mutated NSCLC. Full article
Show Figures

Figure 1

30 pages, 1346 KiB  
Review
Preclinical Models for Functional Precision Lung Cancer Research
by Jie-Zeng Yu, Zsofia Kiss, Weijie Ma, Ruqiang Liang and Tianhong Li
Cancers 2025, 17(1), 22; https://doi.org/10.3390/cancers17010022 - 25 Dec 2024
Cited by 1 | Viewed by 3435
Abstract
Patient-centered precision oncology strives to deliver individualized cancer care. In lung cancer, preclinical models and technological innovations have become critical in advancing this approach. Preclinical models enable deeper insights into tumor biology and enhance the selection of appropriate systemic therapies across chemotherapy, targeted [...] Read more.
Patient-centered precision oncology strives to deliver individualized cancer care. In lung cancer, preclinical models and technological innovations have become critical in advancing this approach. Preclinical models enable deeper insights into tumor biology and enhance the selection of appropriate systemic therapies across chemotherapy, targeted therapies, immunotherapies, antibody–drug conjugates, and emerging investigational treatments. While traditional human lung cancer cell lines offer a basic framework for cancer research, they often lack the tumor heterogeneity and intricate tumor–stromal interactions necessary to accurately predict patient-specific clinical outcomes. Patient-derived xenografts (PDXs), however, retain the original tumor’s histopathology and genetic features, providing a more reliable model for predicting responses to systemic therapeutics, especially molecularly targeted therapies. For studying immunotherapies and antibody–drug conjugates, humanized PDX mouse models, syngeneic mouse models, and genetically engineered mouse models (GEMMs) are increasingly utilized. Despite their value, these in vivo models are costly, labor-intensive, and time-consuming. Recently, patient-derived lung cancer organoids (LCOs) have emerged as a promising in vitro tool for functional precision oncology studies. These LCOs demonstrate high success rates in growth and maintenance, accurately represent the histology and genomics of the original tumors and exhibit strong correlations with clinical treatment responses. Further supported by advancements in imaging, spatial and single-cell transcriptomics, proteomics, and artificial intelligence, these preclinical models are reshaping the landscape of drug development and functional precision lung cancer research. This integrated approach holds the potential to deliver increasingly accurate, personalized treatment strategies, ultimately enhancing patient outcomes in lung cancer. Full article
Show Figures

Figure 1

13 pages, 992 KiB  
Review
The Application of Molecularly Imprinted Polymers in Forensic Toxicology: Issues and Perspectives
by Susan Mohamed, Simone Santelli, Arianna Giorgetti, Guido Pelletti, Filippo Pirani, Paolo Fais and Jennifer P. Pascali
Chemosensors 2024, 12(12), 279; https://doi.org/10.3390/chemosensors12120279 - 23 Dec 2024
Cited by 5 | Viewed by 1608
Abstract
Molecularly imprinted polymers (MIPs) are synthetic receptors designed to selectively bind specific molecules, mimicking natural antibody–antigen interactions. Produced through polymerization around a target molecule (template), MIPs create imprints that confer high specificity and binding affinity upon template removal. Initially developed in the 1970s [...] Read more.
Molecularly imprinted polymers (MIPs) are synthetic receptors designed to selectively bind specific molecules, mimicking natural antibody–antigen interactions. Produced through polymerization around a target molecule (template), MIPs create imprints that confer high specificity and binding affinity upon template removal. Initially developed in the 1970s with organic polymers, MIPs now play critical roles in separation sciences, catalysis, drug delivery, and sensor technology. In forensic science, MIPs offer potential for sample preparation, pre-concentration, and analyte detection, especially with complex biological and non-biological matrices. They exhibit superior stability under extreme conditions, enabling their use in challenging forensic contexts such as detecting new psychoactive substances or trace explosives. Despite advantages like reusability and high selectivity, MIPs face limitations in forensic analysis due to their complex synthesis, potential template leakage, and non-specific binding. Moreover, the lack of standardized protocols limits their mainstream adoption, as forensic applications require validated, reproducible methods. This review systematically assesses MIPs in forensic toxicology, focusing on their current capabilities, limitations, and potential for broader integration into forensic workflows. Future research should address standardization and evaluate MIPs’ effectiveness in diverse forensic applications to realize their full potential. Full article
(This article belongs to the Special Issue Chemical Sensing and Analytical Methods for Forensic Applications)
Show Figures

Figure 1

6 pages, 813 KiB  
Case Report
Precision Oncology Approach for Urachal Carcinoma: A Clinical Case Report
by Dániel Juhász, Anita Csizmarik, János Szalontai, Attila Keszthelyi, Bálint Dér, András Kubik, Miklós Szűcs, István Kenessey, Iris E. Ertl, Walter Berger, Bernhard Englinger, Shahrokh F. Shariat, Péter Nyirády and Tibor Szarvas
Int. J. Mol. Sci. 2024, 25(24), 13315; https://doi.org/10.3390/ijms252413315 - 12 Dec 2024
Cited by 2 | Viewed by 1496
Abstract
Urachal cancer (UrC) is a rare disease which is mostly diagnosed late due to symptoms caused by its local invasion to the urinary bladder. Given the lack of clinical trials and guideline recommendations for systemic treatment, a molecularly informed precision oncology approach is [...] Read more.
Urachal cancer (UrC) is a rare disease which is mostly diagnosed late due to symptoms caused by its local invasion to the urinary bladder. Given the lack of clinical trials and guideline recommendations for systemic treatment, a molecularly informed precision oncology approach is a viable option for UrC already in the early lines of systemic treatment. While single case experiences may provide valuable reference for later decision-making, well-documented clinical experience with off-label targeted treatments is limited to a few patients. Here, we report a case of a 31-year-old female UrC patient who underwent intensive therapy with three surgeries and five lines of systemic treatments, including chemo-, checkpoint inhibitor and tyrosine kinase inhibitor therapies. In addition, next-generation sequencing (NGS) analysis and an ex vivo drug-screening analysis were performed on patient-derived tumor cells and the results were implemented into the therapeutic decision-making. Finally, serum carcinoembryonic antigen (CEA) levels proved to be helpful for therapy monitoring during the whole follow-up period. Full article
Show Figures

Figure 1

16 pages, 588 KiB  
Review
Lung Cancer with Brain Metastasis—Treatment Strategies and Molecular Characteristics
by Shuai Wang, Matan Uriel and Haiying Cheng
J. Clin. Med. 2024, 13(23), 7371; https://doi.org/10.3390/jcm13237371 - 3 Dec 2024
Cited by 4 | Viewed by 3361
Abstract
Lung cancer is a leading cause of brain metastases (BMs), with 10–20% of patients with non-small cell lung cancer (NSCLC) presenting with BMs at diagnosis and 25–50% developing them over the course of their disease. Historically, BMs have posed significant therapeutic challenges, partly [...] Read more.
Lung cancer is a leading cause of brain metastases (BMs), with 10–20% of patients with non-small cell lung cancer (NSCLC) presenting with BMs at diagnosis and 25–50% developing them over the course of their disease. Historically, BMs have posed significant therapeutic challenges, partly due to the blood brain barrier (BBB), which restricts drug penetration to the central nervous system. Consequently, BMs were initially managed with local treatments, including surgical resection, stereotactic radiosurgery, and whole brain radiation therapy. In recent years, however, systemic treatments for BMs have advanced significantly, particularly with the development of molecularly-targeted therapies and immunotherapies. The discovery of driver mutations and the development of novel tyrosine kinase inhibitors (TKIs) have yielded encouraging intracranial responses in NSCLC patients with actionable genetic alterations (e.g., EGFR, ALK, ROS1). Genomic profiling has also suggested genetic heterogeneity between BMs and primary sites. Immunotherapies, alone or in combination with other treatments, have demonstrated promising results in NSCLC with BMs, although most clinical trials have included only selected patients with asymptomatic or previously treated BMs. In this review, we discuss the molecular and immune characteristics of NSCLC with BMs, analyze intracranial efficacy findings from clinical trials, and explore treatment strategies for lung cancer patients with BMs. Full article
(This article belongs to the Section Respiratory Medicine)
Show Figures

Figure 1

Back to TopTop