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 (56)

Search Parameters:
Keywords = personalized cardiovascular drug therapy

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
36 pages, 928 KiB  
Review
Reprogramming Atherosclerosis: Precision Drug Delivery, Nanomedicine, and Immune-Targeted Therapies for Cardiovascular Risk Reduction
by Paschalis Karakasis, Panagiotis Theofilis, Panayotis K. Vlachakis, Konstantinos Grigoriou, Dimitrios Patoulias, Antonios P. Antoniadis and Nikolaos Fragakis
Pharmaceutics 2025, 17(8), 1028; https://doi.org/10.3390/pharmaceutics17081028 (registering DOI) - 7 Aug 2025
Abstract
Atherosclerosis is a progressive, multifactorial disease driven by the interplay of lipid dysregulation, chronic inflammation, oxidative stress, and maladaptive vascular remodeling. Despite advances in systemic lipid-lowering and anti-inflammatory therapies, residual cardiovascular risk persists, highlighting the need for more precise interventions. Targeted drug delivery [...] Read more.
Atherosclerosis is a progressive, multifactorial disease driven by the interplay of lipid dysregulation, chronic inflammation, oxidative stress, and maladaptive vascular remodeling. Despite advances in systemic lipid-lowering and anti-inflammatory therapies, residual cardiovascular risk persists, highlighting the need for more precise interventions. Targeted drug delivery represents a transformative strategy, offering the potential to modulate key pathogenic processes within atherosclerotic plaques while minimizing systemic exposure and off-target effects. Recent innovations span a diverse array of platforms, including nanoparticles, liposomes, exosomes, polymeric carriers, and metal–organic frameworks (MOFs), engineered to engage distinct pathological features such as inflamed endothelium, dysfunctional macrophages, oxidative microenvironments, and aberrant lipid metabolism. Ligand-based, biomimetic, and stimuli-responsive delivery systems further enhance spatial and temporal precision. In parallel, advances in in-silico modeling and imaging-guided approaches are accelerating the rational design of multifunctional nanotherapeutics with theranostic capabilities. Beyond targeting lipids and inflammation, emerging strategies seek to modulate immune checkpoints, restore endothelial homeostasis, and reprogram plaque-resident macrophages. This review provides an integrated overview of the mechanistic underpinnings of atherogenesis and highlights state-of-the-art targeted delivery systems under preclinical and clinical investigation. By synthesizing recent advances, we aim to elucidate how precision-guided drug delivery is reshaping the therapeutic landscape of atherosclerosis and to chart future directions toward clinical translation and personalized vascular medicine. Full article
Show Figures

Figure 1

29 pages, 510 KiB  
Systematic Review
Impact of Biologic Drugs on Comorbidity Outcomes in Rheumatoid Arthritis: A Systematic Review
by Soumaya Boussaid, Rim Dhahri, Safa Rahmouni, Halil İbrahim Ceylan, Marwa Hassayoun, Maissa Abbes, Khaoula Zouaoui, Ismail Dergaa, Sonia Rekik, Nadia Boussaid, Imen Gharsallah, Raul Ioan Muntean and Hela Sahli
J. Clin. Med. 2025, 14(13), 4547; https://doi.org/10.3390/jcm14134547 - 26 Jun 2025
Viewed by 624
Abstract
Background: The management of rheumatoid arthritis (RA) has advanced significantly with the introduction of biologic disease-modifying antirheumatic drugs (bDMARDs). Despite these therapeutic strides, RA prognosis remains profoundly affected by comorbid conditions, particularly cardiovascular and metabolic complications, which increase both morbidity and mortality. The [...] Read more.
Background: The management of rheumatoid arthritis (RA) has advanced significantly with the introduction of biologic disease-modifying antirheumatic drugs (bDMARDs). Despite these therapeutic strides, RA prognosis remains profoundly affected by comorbid conditions, particularly cardiovascular and metabolic complications, which increase both morbidity and mortality. The role of bDMARDs in modulating comorbidities remains underexplored, with limited evidence on their effects across various non-RA conditions, such as respiratory, diabetic, and hematologic disorders. This systematic review aimed to evaluate the impact of bDMARDs on the progression and outcomes of comorbidities in RA patients, providing insights to guide personalized treatment approaches. Methods: This systematic review was registered in PROSPERO (CRD42022345903) and followed the PRISMA guidelines. Original research articles from PubMed and Scopus, published up to 18 July 2024, were included. Studies assessing the impact of bDMARDs on comorbidities in RA patients met the eligibility criteria. Results: A total of thirteen studies met the inclusion criteria. They were published from inception until July 2024. The studied comorbidities included pulmonary conditions (asthma, chronic obstructive pulmonary disease, and interstitial lung disease) (n = 2); diabetes (n = 3); anemia (n = 3); and malignancies (n = 3). The bDMARDs studied were tumor necrosis factor inhibitors (TNFis) (n = 9); Rituximab (n = 5); Tocilizumab (n = 5); Abatacept (n = 5); and Anakinra (n = 2). The most reported effects of bDMARDs on comorbidities were the following: (i) an exacerbation of pulmonary comorbidities for Abatacept and TNFis; (ii) patients switched to or initiated on Abatacept as their first targeted disease-modifying antirheumatic drug (tDMARD) showed directionally lower rates and costs of T2DM-related complications compared with patients switching to or initiating other tDMARDs; (iii) there was no difference between Abatacept and TNFis or Rituximab/Tocilizumab regarding diabetes treatment switching or intensification; (iv) Anakinra significantly reduced the HbA1c%; (v) decreased serum hepcidin levels and improvement in anemia were observed in patients treated with TNFis or Tocilizumab; and (vi) no decrease in overall survival time or the significant incident malignancy rate was noted in RA patients. Conclusions: Overall, bDMARDs appear safe for use in RA patients with comorbidities and may even provide specific benefits for conditions such as anemia and diabetes. These findings suggest that clinicians could consider tailoring biologic therapy based on each patient’s comorbidity profile, potentially enhancing both RA management and comorbidity outcomes. For instance, selecting biologics such as Anakinra or Tocilizumab might be advantageous for RA patients with concurrent diabetes or anemia, given their observed metabolic and hematologic benefits. This personalized approach could improve the quality of life and reduce healthcare costs by addressing RA and associated comorbidities more effectively. Full article
(This article belongs to the Section Immunology)
Show Figures

Figure 1

42 pages, 2266 KiB  
Review
Innovative Approaches in Cancer Treatment: Emphasizing the Role of Nanomaterials in Tyrosine Kinase Inhibition
by Antónia Kurillová, Libor Kvítek and Aleš Panáček
Pharmaceutics 2025, 17(6), 783; https://doi.org/10.3390/pharmaceutics17060783 - 16 Jun 2025
Viewed by 845
Abstract
Medical research is at the forefront of addressing pressing global challenges, including preventing and treating cardiovascular, autoimmune, and oncological diseases, neurodegenerative disorders, and the growing resistance of pathogens to antibiotics. Understanding the molecular mechanisms underlying these diseases, using advanced medical approaches and cutting-edge [...] Read more.
Medical research is at the forefront of addressing pressing global challenges, including preventing and treating cardiovascular, autoimmune, and oncological diseases, neurodegenerative disorders, and the growing resistance of pathogens to antibiotics. Understanding the molecular mechanisms underlying these diseases, using advanced medical approaches and cutting-edge technologies, structure-based drug design, and personalized medicine, is critical for developing effective therapies, specifically anticancer treatments. Background/Objectives: One of the key drivers of cancer at the cellular level is the abnormal activity of protein enzymes, specifically serine, threonine, or tyrosine residues, through a process known as phosphorylation. While tyrosine kinase-mediated phosphorylation constitutes a minor fraction of total cellular phosphorylation, its dysregulation is critically linked to carcinogenesis and tumor progression. Methods: Small-molecule inhibitors, such as imatinib or erlotinib, are designed to halt this process, restoring cellular equilibrium and offering targeted therapeutic approaches. However, challenges persist, including frequent drug resistance and severe side effects associated with these therapies. Nanomedicine offers a transformative potential to overcome these limitations. Results: By leveraging the unique properties of nanomaterials, it is possible to achieve precise drug delivery, enhance accumulation at target sites, and improve therapeutic efficacy. Examples include nanoparticle-based delivery systems for TKIs and the combination of nanomaterials with photothermal or photodynamic therapies to enhance treatment effectiveness. Combining nanomedicine with traditional treatments holds promise and perspective for synergistic and more effective cancer management. Conclusions: This review delves into recent advances in understanding tyrosine kinase activity, the mechanisms of their inhibition, and the innovative integration of nanomedicine to revolutionize cancer treatment strategies. Full article
Show Figures

Graphical abstract

18 pages, 3937 KiB  
Article
Preliminary Evaluation of 3D-Printed Alginate/Gelatin Scaffolds for Protein Fast Release as Suitable Devices for Personalized Medicine
by Benedetta Ghezzi, Ruben Foresti, Luisa Pia Scialoia, Maddalena Botti, Arianna Mersanne, Fulvio Ratto, Francesca Rossi, Chiara Martini, Paolo Perini, Elda Favari and Antonio Freyrie
Biomedicines 2025, 13(6), 1365; https://doi.org/10.3390/biomedicines13061365 - 2 Jun 2025
Viewed by 733
Abstract
Background/Objectives: Drug-coated balloons (DCBs) are emerging as a promising treatment for peripheral artery disease; however, current technologies lack flexibility in customizing drug release profiles and composition, limiting their therapeutic potential. This study aims to develop a Gelatin (Gel) and Sodium Alginate (Alg) bioink [...] Read more.
Background/Objectives: Drug-coated balloons (DCBs) are emerging as a promising treatment for peripheral artery disease; however, current technologies lack flexibility in customizing drug release profiles and composition, limiting their therapeutic potential. This study aims to develop a Gelatin (Gel) and Sodium Alginate (Alg) bioink loaded with apolipoprotein A-I (apoA-I) for controlled drug delivery by using additive manufacturing technologies. Methods: We developed and printed via rapid freeze prototyping (RFP) a Gel and Alg bioink loaded with different concentrations of apoA-I. Mechanical properties related to compressional and tensile forces have been studied, as well as the structural stability and active release from the 3D structure of apoA-I (cholesterol efflux assays). The biological behavior of HUVEC cells with and without ApoA-I was assessed by proliferation assay, metabolic activity analysis, and fluorescence imaging. Results: The 3D structures presented breakpoint stress values consistent with the mechanical requirements for integration within a DCB, and the ability to effectively promote cholesterol transport in J774 cells. Moreover, in vitro studies on HUVECs revealed that the scaffolds exhibited no cytotoxic effects, leading to increased ATP levels and enhanced metabolic activity over time, confirming the possibility to obtain RFP-printed Alg/Gel scaffolds able to provide a stable structure capable of controlled apoA-I release. Conclusions: These findings support the potential of Alg/Gel+apoA-I scaffolds as biocompatible drug delivery systems for vascular applications. Their ability to maintain structural integrity while enabling controlled biomolecular release positions them as promising candidates for personalized cardiovascular therapy, facilitating the rapid customization of bioprinted therapeutic platforms. Full article
(This article belongs to the Section Biomedical Engineering and Materials)
Show Figures

Figure 1

8 pages, 945 KiB  
Case Report
Triple Non-Statin Therapy with Ezetimibe, Inclisiran, and Bempedoic Acid in Patients with Genetically Confirmed Statin-Induced Rhabdomyolysis: A Dual Case Report
by Jozef Dodulík, Jiří Plášek, Ivana Kacířová, Romana Uřinovská, Jiří Vrtal and Jan Václavík
Pharmaceuticals 2025, 18(6), 818; https://doi.org/10.3390/ph18060818 - 29 May 2025
Cited by 1 | Viewed by 1042
Abstract
Background: Statin intolerance is a serious therapeutic dilemma in secondary cardiovascular prevention (e.g., ESC/EAS Guidelines 2023). This is especially true when confirmed by genetic predisposition and complicated by rhabdomyolysis. Although several non-statin agents have become available in recent years, evidence regarding their combined [...] Read more.
Background: Statin intolerance is a serious therapeutic dilemma in secondary cardiovascular prevention (e.g., ESC/EAS Guidelines 2023). This is especially true when confirmed by genetic predisposition and complicated by rhabdomyolysis. Although several non-statin agents have become available in recent years, evidence regarding their combined use in high-risk statin-intolerant patients remains limited. Furthermore, the pharmacokinetics of statins in toxic concentrations are poorly characterized in clinical settings. Case Presentation: We present two cases of genetically confirmed statin-induced rhabdomyolysis, both accompanied by severe acute kidney injury requiring renal replacement therapy. In both patients, serial measurements of rosuvastatin plasma concentrations revealed markedly delayed elimination, with detectable levels persisting for several weeks despite ongoing dialysis. Estimated half-lives exceeded 7 days in both cases, far beyond the known therapeutic range. Genetic testing identified SLCO1B1, ABCB1, and CYP2C9 polymorphisms linked to reduced hepatic uptake and impaired drug clearance. Following biochemical recovery, both patients were initiated on a triple non-statin lipid-lowering regimen consisting of ezetimibe, bempedoic acid, and inclisiran. The combination was well tolerated, with no recurrence of muscle-related symptoms or biochemical toxicity. LDL-C levels were reduced from 3.05 to 1.59 mmol/L and from 4.99 to 1.52 mmol/L, respectively, with sustained response over 12 and 40 weeks. Full lipid profiles demonstrated favorable changes across all parameters. Conclusions: These two cases suggest that the combination of ezetimibe, inclisiran, and bempedoic acid may serve as a safe and effective therapeutic option in patients with severe statin intolerance. Pharmacogenetic testing and serial pharmacokinetic assessment may guide personalized lipid-lowering strategies and improve outcomes in this challenging patient population. Full article
(This article belongs to the Topic Research in Pharmacological Therapies, 2nd Edition)
Show Figures

Graphical abstract

21 pages, 356 KiB  
Review
Overview of Oncology: Drug-Induced Cardiac Toxicity
by Nilima Rajpal Kundnani, Vincenzo Passini, Iulia Stefania Carlogea, Patrick Dumitrescu, Vlad Meche, Roxana Buzas and Daniel Marius Duda-Seiman
Medicina 2025, 61(4), 709; https://doi.org/10.3390/medicina61040709 - 12 Apr 2025
Cited by 1 | Viewed by 1366
Abstract
Cancer medications can cause cardiac issues, which are difficult to treat in oncologic patients because of the risk of complications. In some cases, this may significantly impact their well-being and treatment outcomes. Overall, these complications fall under the term “drug induced cardiotoxicity”, mainly [...] Read more.
Cancer medications can cause cardiac issues, which are difficult to treat in oncologic patients because of the risk of complications. In some cases, this may significantly impact their well-being and treatment outcomes. Overall, these complications fall under the term “drug induced cardiotoxicity”, mainly due to chemotherapy drugs being specifically toxic to the heart, causing a decrease in the heart’s capacity to pump blood efficiently and leading to a reduction in the left ventricular ejection fraction (LVEF), and subsequently possibly leading to heart failure. Anthracyclines, alkylating agents, and targeted therapies for cancer hold the potential of causing harmful effects on the heart. The incidence of heart-related issues varies from patient to patient and depends on multiple factors, including the type of medication, dosage, duration of the treatment, and pre-existing heart conditions. The underlying mechanism leading to oncologic-drug-induced cardiovascular harmful effects is quite complex. One particular group of drugs, called anthracyclines, have garnered attention due to their impact on oxidative stress and their ability to cause direct harm to heart muscle cells. Reactive oxygen species (ROS) cause harm by inducing damage and programmed cell death in heart cells. Conventional biomarkers alone can only indicate some degree of damage that has already occurred and, therefore, early detection is key. Novel methods like genetic profiling are being developed to detect individuals at risk, prior to the onset of clinical symptoms. Key management strategies—including early detection, personalized medicine approaches, and the use of novel biomarkers—play a crucial role in mitigating cardiotoxicity and improving patient outcomes. Identification of generated genetic alterations and the association to an increased likelihood of cardiotoxicity will allow treatment in a more personalized approach, aiming at decreasing rates of cardiac events while maintaining high oncological efficacy. Oncology drug-induced cardiotoxicity is managed through a combination of preventive strategies and therapeutic interventions from the union of cardiac and oncological knowledge. Full article
(This article belongs to the Special Issue New Insights into Heart Failure)
14 pages, 1433 KiB  
Review
Antiplatelet and Anticoagulation Therapy in Athletes: A Cautious Compromise… If Possible!
by Flavio D’Ascenzi, Guglielmo Leonardo Manfredi, Vincenzo Minasi, Gian Luca Ragazzoni, Luna Cavigli, Alessandro Zorzi, Giulia Elena Mandoli, Maria Concetta Pastore, Marta Focardi, Matteo Cameli, Massimo Fineschi and Serafina Valente
J. Cardiovasc. Dev. Dis. 2025, 12(4), 151; https://doi.org/10.3390/jcdd12040151 - 10 Apr 2025
Viewed by 887
Abstract
Antiplatelet and anticoagulation therapy are commonly used in the general population and sometimes in athletes experiencing cardiovascular disorders. In these cases, the treatment has to be tailored according to the individual bleeding and thrombotic risk profile, also considering the intrinsic risk of sports [...] Read more.
Antiplatelet and anticoagulation therapy are commonly used in the general population and sometimes in athletes experiencing cardiovascular disorders. In these cases, the treatment has to be tailored according to the individual bleeding and thrombotic risk profile, also considering the intrinsic risk of sports activities when advising athletes for eligibility for competitive sports. In athletes, it is necessary to pre-assess the individual bleeding risk, considering not only the personal bleeding risk (usually low in athletes) but also the type of sport the athlete would like to practice, with careful consideration in sports where traumatic collisions are highly likely. Additionally, non-steroidal anti-inflammatory drugs are commonly used among athletes, and antiplatelet therapy may further increase the bleeding risk. Therefore, in selected competitive athletes, the default approach for antithrombotic therapy could be personalized. This review discusses the clinical management challenges of competitive athletes under antithrombotic or antiplatelet therapy, focusing on the intrinsic risks of sports practice and the indications for sports eligibility and disqualification. Full article
Show Figures

Figure 1

34 pages, 771 KiB  
Review
Polygenic Risk Scores for Personalized Cardiovascular Pharmacogenomics―A Scoping Review
by Aaryan Dwivedi, Jobanjit S. Phulka, Peyman Namdarimoghaddam and Zachary Laksman
Sci. Pharm. 2025, 93(2), 18; https://doi.org/10.3390/scipharm93020018 - 8 Apr 2025
Viewed by 2325
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality worldwide, often involving a strong genetic background. Polygenic risk scores (PRSs) combine the cumulative effects of multiple genetic variants to quantify an individual’s susceptibility to CVD. Pharmacogenomics (PGx) can further personalize treatment by tailoring [...] Read more.
Cardiovascular disease (CVD) is the leading cause of mortality worldwide, often involving a strong genetic background. Polygenic risk scores (PRSs) combine the cumulative effects of multiple genetic variants to quantify an individual’s susceptibility to CVD. Pharmacogenomics (PGx) can further personalize treatment by tailoring medication choices to an individual’s genetic profile. Even with these potential benefits, the extent to which PRS can be integrated into the PGx of CVD remains unclear. Our review provides an overview of current evidence on the application of PRS in the PGx of CVD, examining clinical utility and limitations and providing directions for future research. Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews protocol, we conducted a comprehensive literature search in PubMed, EMBASE, and the Web of Science. Studies investigating the relationship between PRS in predicting the efficacy, adverse effects, or cost-effectiveness of cardiovascular medications were selected. Of the 1894 articles identified, 32 met the inclusion criteria. These studies predominantly examined lipid-lowering therapies, antihypertensives, and antiplatelets, although other medication classes (e.g., rate-control drugs, ibuprofen/acetaminophen, diuretics, and antiarrhythmics) were also included. Our findings showed that PRS is most robustly validated in lipid-lowering therapies, especially statins, where studies reported that individuals with higher PRSs derived the greatest reduction in lipids while on statins. Studies analyzing antihypertensives, antiplatelets, and antiarrhythmic medications demonstrated more variable outcomes, though certain PRSs did identify subgroups with significantly improved response rates or a higher risk of adverse events. Though PRS was a strong tool in many cases, we found some key limitations in its applicability in research, such as the under-representation of non-European-ancestry cohorts in the examined studies and a lack of standardized outcome reporting. In conclusion, though PRS offers promise in improving the efficacy of PGx of CVD by enhancing the personalization of medication on an individual level, several obstacles, such as the need for including a broader ancestral diversity and more robust cost-effectiveness data remain. Future research must (i) prioritize validating PRS in ethnically diverse populations, (ii) refine PRS derivation methods to tailor them for drug response phenotypes, and (iii) establish clear and attainable guidelines for standardizing the reporting of outcomes. Full article
Show Figures

Figure 1

29 pages, 1998 KiB  
Review
Pentraxin-3 as a Biomarker in Diabetes Mellitus: Insights into Inflammation, Vascular Complications, and Modulation by Antidiabetic Medications
by Roxana-Cristina Dobriceanu, Andreea Daniela Meca, Ianis Kevyn Stefan Boboc, Liliana Mititelu-Tartau, Mihaela Simona Naidin, Adina Turcu-Stiolica and Maria Bogdan
Biomedicines 2025, 13(4), 891; https://doi.org/10.3390/biomedicines13040891 - 7 Apr 2025
Viewed by 919
Abstract
Diabetes mellitus (DM) is a multifactorial metabolic disorder associated with systemic inflammation and vascular complications. Pentraxin-3 (PTX3) has emerged as a key biomarker of inflammation and endothelial dysfunction in DM. We aimed to examine the role of PTX3 in DM and assesses the [...] Read more.
Diabetes mellitus (DM) is a multifactorial metabolic disorder associated with systemic inflammation and vascular complications. Pentraxin-3 (PTX3) has emerged as a key biomarker of inflammation and endothelial dysfunction in DM. We aimed to examine the role of PTX3 in DM and assesses the impact of pharmacological interventions on its expression. The review included studies analyzing PTX3 modulation by antidiabetic therapies, such as sodium-glucose cotransporter-2 inhibitors (SGLT-2i), glucagon-like peptide-1 agonists (GLP-1a), and dipeptidyl peptidase-4 inhibitors (DPP-4i), as well as the effects of lifestyle interventions. Clinical and experimental studies demonstrated a strong correlation between PTX3 levels and DM progression. Elevated PTX3 levels were associated with diabetic complications, including nephropathy, retinopathy, and cardiovascular diseases. Antidiabetic drugs showed differential effects on PTX3 expression, with GLP-1a and DPP-4i significantly reducing PTX3 levels, while SGLT-2i displayed a paradoxical increase. Lifestyle interventions, including dietary modifications and weight loss, yielded inconsistent effects, suggesting genetic and metabolic factors influence PTX3 regulation. While pharmacological therapies, particularly GLP-1a and DPP-4i, demonstrate anti-inflammatory effects, further research is needed to standardize PTX3 measurement and explore its potential as a therapeutic target. Personalized treatment strategies incorporating genetic profiling may optimize inflammation control and disease management in DM patients. Full article
Show Figures

Figure 1

34 pages, 4932 KiB  
Review
Advances and Challenges in 3D Bioprinted Cancer Models: Opportunities for Personalized Medicine and Tissue Engineering
by Sai Liu and Pan Jin
Polymers 2025, 17(7), 948; https://doi.org/10.3390/polym17070948 - 31 Mar 2025
Viewed by 1672
Abstract
Cancer is the second leading cause of death worldwide, after cardiovascular disease, claiming not only a staggering number of lives but also causing considerable health and economic devastation, particularly in less-developed countries. Therapeutic interventions are impeded by differences in patient-to-patient responses to anti-cancer [...] Read more.
Cancer is the second leading cause of death worldwide, after cardiovascular disease, claiming not only a staggering number of lives but also causing considerable health and economic devastation, particularly in less-developed countries. Therapeutic interventions are impeded by differences in patient-to-patient responses to anti-cancer drugs. A personalized medicine approach is crucial for treating specific patient groups and includes using molecular and genetic screens to find appropriate stratifications of patients who will respond (and those who will not) to treatment regimens. However, information on which risk stratification method can be used to hone in on cancer types and patients who will be likely responders to a specific anti-cancer agent remains elusive for most cancers. Novel developments in 3D bioprinting technology have been widely applied to recreate relevant bioengineered tumor organotypic structures capable of mimicking the human tissue and microenvironment or adequate drug responses in high-throughput screening settings. Parts are autogenously printed in the form of 3D bioengineered tissues using a computer-aided design concept where multiple layers include different cell types and compatible biomaterials to build specific configurations. Patient-derived cancer and stromal cells, together with genetic material, extracellular matrix proteins, and growth factors, are used to create bioprinted cancer models that provide a possible platform for the screening of new personalized therapies in advance. Both natural and synthetic biopolymers have been used to encourage the growth of cells and biological materials in personalized tumor models/implants. These models may facilitate physiologically relevant cell–cell and cell–matrix interactions with 3D heterogeneity resembling real tumors. Full article
(This article belongs to the Section Polymer Applications)
Show Figures

Graphical abstract

22 pages, 1598 KiB  
Article
Bevacizumab—Insights from EudraVigilance Database on the Assessments of the Safety Profile of Monoclonal Antibodies Used as Targeted Cancer Treatment
by Razvan Constantin Vonica, Anca Butuca, Claudiu Morgovan, Manuela Pumnea, Remus Calin Cipaian, Adina Frum, Carmen Maximiliana Dobrea, Andreea Loredana Vonica-Tincu, Aliteia-Maria Pacnejer, Steliana Ghibu, Florina Batar and Felicia Gabriela Gligor
Pharmaceuticals 2025, 18(4), 501; https://doi.org/10.3390/ph18040501 - 30 Mar 2025
Cited by 1 | Viewed by 891
Abstract
Background/Objectives: Worldwide, colon cancer is a major cause of cancer-related mortality, with an increasing incidence influenced by genetic, environmental, and lifestyle factors. Despite advances in diagnosis and personalized treatments, challenges remain in improving patient prognosis, particularly in metastatic colorectal cancer (mCRC). Bevacizumab (BEV), [...] Read more.
Background/Objectives: Worldwide, colon cancer is a major cause of cancer-related mortality, with an increasing incidence influenced by genetic, environmental, and lifestyle factors. Despite advances in diagnosis and personalized treatments, challenges remain in improving patient prognosis, particularly in metastatic colorectal cancer (mCRC). Bevacizumab (BEV), a monoclonal antibody, is widely used in colorectal cancer treatment. This study aimed to analyze adverse events associated with BEV compared with other therapies based on data from the EudraVigilance (EV) database. Methods: A descriptive and disproportionality analysis was conducted on signals reported in the EV database related to BEV. The study included comparisons with other antineoplastic treatments, such as chemotherapy, targeted therapy, and immunotherapy. Patient demographics, severity of adverse drug reactions (ADRs), and distribution patterns were analyzed to assess the safety profile of BEV in colorectal cancer treatment. Results: The majority of the signals for BEV were from patients aged 18–64 years (39.42%) and 65–85 years (34.08%). Hypertension, thromboembolism, proteinuria, and gastrointestinal disorders have been the most frequently reported. Serious ADRs, including gastrointestinal perforations, hemorrhage, and arterial thromboembolism, were observed in 93.74% of Individual Case Safety Reports. BEV was associated with a higher likelihood of vascular and endocrine disorders compared with chemotherapy and other targeted therapies. Immunotherapy was linked to increased immunological ADRs, while BEV demonstrated fewer immune-related toxicities. Conclusions: Continuous monitoring is necessary to optimize patient management, particularly in elderly patients or those with cardiovascular comorbidities. Understanding BEV’s safety profile allows for better personalization of treatment strategies, minimizing risks while enhancing therapeutic outcomes. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

37 pages, 2063 KiB  
Review
Analysis of Personalized Cardiovascular Drug Therapy: From Monitoring Technologies to Data Integration and Future Perspectives
by Runxing Lin, Ziyu Huang, Yu Liu and Yinning Zhou
Biosensors 2025, 15(3), 191; https://doi.org/10.3390/bios15030191 - 17 Mar 2025
Viewed by 3393
Abstract
Cardiovascular diseases have long been a major challenge to human health, and the treatment differences caused by individual variability remain unresolved. In recent years, personalized cardiovascular drug therapy has attracted widespread attention. This paper reviews the strategies for achieving personalized cardiovascular drug therapy [...] Read more.
Cardiovascular diseases have long been a major challenge to human health, and the treatment differences caused by individual variability remain unresolved. In recent years, personalized cardiovascular drug therapy has attracted widespread attention. This paper reviews the strategies for achieving personalized cardiovascular drug therapy through traditional dynamic monitoring and multidimensional data integration and analysis. It focuses on key technologies for dynamic monitoring, dynamic monitoring based on individual differences, and multidimensional data integration and analysis. By systematically reviewing the relevant literature, the main challenges in current research and the proposed potential directions for future studies were summarized. Full article
(This article belongs to the Section Biosensors and Healthcare)
Show Figures

Figure 1

31 pages, 2052 KiB  
Systematic Review
Innovations in Snake Venom-Derived Therapeutics: A Systematic Review of Global Patents and Their Pharmacological Applications
by Diana Carolina Zona Rubio, Diana Marcela Aragón and Izabel Almeida Alves
Toxins 2025, 17(3), 136; https://doi.org/10.3390/toxins17030136 - 14 Mar 2025
Cited by 3 | Viewed by 3502
Abstract
Active compounds from natural sources, particularly snake venoms, are crucial for pharmaceutical development despite challenges in drug discovery. Snake venoms, historically used for medicinal purposes, contain bioactive peptides and enzymes that show therapeutic potential for conditions such as arthritis, asthma, cancer, chronic pain, [...] Read more.
Active compounds from natural sources, particularly snake venoms, are crucial for pharmaceutical development despite challenges in drug discovery. Snake venoms, historically used for medicinal purposes, contain bioactive peptides and enzymes that show therapeutic potential for conditions such as arthritis, asthma, cancer, chronic pain, infections and cardiovascular diseases. The objective of this study was to examine pharmacological and biomedical innovations by identifying the key research trends, the most studied snake species, and their therapeutic applications. A systematic review of patents related to snake venoms was conducted using the European Patent Office database, Espacenet, covering 2014 to mid-2024. The search employed the keyword “venom,” applying IPC classification A61K38/00, resulting in 31 patents after screening. A PubMed survey on “snake venom derivatives innovations” was conducted to compare the scientific literature volume with the identified patents. This review highlights the therapeutic potential of snake venom-derived products for coagulation disorders, cancer, inflammation, and pain management. Despite challenges in pharmacokinetics and venom variability, advancements in biotechnology offer promise for personalized therapies. The future of snake venom-based treatments appears promising for addressing complex medical conditions. Full article
(This article belongs to the Special Issue Animals Venom in Drug Discovery: A Valuable Therapeutic Tool)
Show Figures

Figure 1

26 pages, 583 KiB  
Review
The “Silent Enemy” Called Renal Artery Stenosis: A Mini-Review
by José Silva, Juan Tonheiro and Fernanda Rodrigues
J. Vasc. Dis. 2025, 4(1), 10; https://doi.org/10.3390/jvd4010010 - 11 Mar 2025
Viewed by 2971
Abstract
Renal artery stenosis (RAS) is a vascular condition characterized by narrowing of one or both renal arteries, leading to reduced blood flow to the kidneys, activation of the renin–angiotensin–aldosterone system (RAAS), and subsequent renovascular hypertension. Overactivation of the same cascade potentiates the production [...] Read more.
Renal artery stenosis (RAS) is a vascular condition characterized by narrowing of one or both renal arteries, leading to reduced blood flow to the kidneys, activation of the renin–angiotensin–aldosterone system (RAAS), and subsequent renovascular hypertension. Overactivation of the same cascade potentiates the production of angiotensin II, which induces systemic vasoconstriction, increases sodium and water retention via aldosterone, and activates the sympathetic nervous system. Angiotensin II is also implicated in endothelial dysfunction, oxidative stress, and chronic inflammation, thus impairing vascular remodeling and arterial stiffness, all of which serve to accelerate cardiovascular complications, such as left ventricular hypertrophy, heart failure, and myocardial infarction. RAS is usually due in at least 90% of cases to atherosclerosis, which typically affects older people with diabetes and smoking as risk factors. There are two types of RAS: unilateral and bilateral. Bilateral RAS is commonly associated with flash pulmonary edema, a life-threatening emergency condition in which alveolar space flooding can occur within minutes. RAS typically remains asymptomatic until the late stage with complications of hypertension, ischemic nephropathy, or chronic kidney disease. FMD tends to create structural abnormalities of the artery, whereas atherosclerosis causes plaque formation and endothelial dysfunction of the artery. Epidemiological surveys have revealed that the prevalence of RAS ranges from 4% to 53% and is especially high among patients with hypertension, cardiovascular disease, or CKD. Diagnosis is based on clinical suspicion and supported by imaging studies, including Doppler ultrasound, computed tomography angiography, and magnetic resonance angiography. Early detection also relies on certain laboratory biomarkers, especially in identifying high-risk patients. These markers would include increased plasma renin activity, elevated aldosterone-renin ratio, and inflammatory markers, including C-reactive protein and endothelin-1. Treatment would also involve pharmacological approaches, including RAAS inhibitors, beta-blockers, and statins, and interventional treatments, including angioplasty and stenting in patients with severe forms of the disease. However, the Cardiovascular Outcomes in Renal Atherosclerotic Lesions (CORAL) Trial showed that most patients would likely require medical therapy, and that intervention should be reserved for those with uncontrolled hypertension, progressive renal dysfunction, or recurrent episodes of pulmonary edema. Other emerging therapies include drug-eluting balloons, bioresorbable stents, and gene-editing techniques, all of which have shown great promise in the few studies that have been conducted, although further evaluation is needed. Despite these advances, there are still gaps in knowledge regarding patient stratification, biomarker validation, and the development of personalized treatment strategies. This article reviews the complexities of RAAS and its systemic impact on cardiovascular and renal health. Future research can therefore focus on improving early diagnosis, optimizing patient selection for intervention, and developing new therapies to slow disease progression and mitigate complications. Full article
(This article belongs to the Section Peripheral Vascular Diseases)
Show Figures

Figure 1

36 pages, 1654 KiB  
Review
Cyclodextrins: Enhancing Drug Delivery, Solubility and Bioavailability for Modern Therapeutics
by Oana Elena Nicolaescu, Ionela Belu, Andreea Gabriela Mocanu, Valentin Costel Manda, Gabriela Rău, Andreea Silvia Pîrvu, Cătălina Ionescu, Felicia Ciulu-Costinescu, Mariana Popescu and Maria Viorica Ciocîlteu
Pharmaceutics 2025, 17(3), 288; https://doi.org/10.3390/pharmaceutics17030288 - 22 Feb 2025
Cited by 20 | Viewed by 6115
Abstract
Cyclodextrins (CDs) have revolutionized the pharmaceutical industry with their ability to enhance the stability, solubility, and bioavailability of a wide range of active substances. These cyclic oligosaccharides, with a unique hydrophilic exterior and hydrophobic cavity, form inclusion complexes with poorly soluble drugs, improving [...] Read more.
Cyclodextrins (CDs) have revolutionized the pharmaceutical industry with their ability to enhance the stability, solubility, and bioavailability of a wide range of active substances. These cyclic oligosaccharides, with a unique hydrophilic exterior and hydrophobic cavity, form inclusion complexes with poorly soluble drugs, improving their pharmacokinetic profiles and therapeutic efficacy. This review explores the multifaceted roles of cyclodextrins in pharmaceutical formulations, ranging from oral, ophthalmic, parenteral, and topical applications to their emerging use in targeted therapies, gene delivery, and treatment of neurodegenerative, cardiovascular, and infectious diseases. Cyclodextrins not only improve drug solubility and controlled release but also reduce toxicity and side effects, leading to safer and more effective treatments. Recent advancements, such as cyclodextrin-based nanoparticles, offer promising pathways for cancer therapy, chronic disease management, and personalized medicine. As research continues, cyclodextrins remain at the forefront of innovation in drug delivery systems, ensuring better patient outcomes and expanding the possibilities of modern therapeutics. Full article
(This article belongs to the Special Issue Cyclodextrins and Their Pharmaceutical Applications)
Show Figures

Graphical abstract

Back to TopTop