Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 18.512
Filter
1.
Gene ; 932: 148904, 2025 Jan 10.
Article in English | MEDLINE | ID: mdl-39218415

ABSTRACT

BACKGROUND: Cervical cancer, primarily caused by HPV infection, remains a global health concern. Current treatments face challenges including drug resistance and toxicity. This study investigates combining E5-siRNA with chemotherapy drugs, Oxaliplatin and Ifosfamide, to enhance treatment efficacy in HPV-16 positive cervical cancer cells, targeting E5 oncoprotein to overcome limitations of existing therapies. METHODS: The CaSki cervical cancer cell line was transfected with E5-siRNA, and subsequently treated with Oxaliplatin/Ifosfamide. Quantitative real-time PCR was employed to assess the expression of related genes including p53, MMP2, Nanog, and Caspases. Cell apoptosis, cell cycle progression, and cell viability were evaluated using Annexin V/PI staining, DAPI staining, and MTT test, respectively. Furthermore, stemness ability was determined through a colony formation assay, and cell motility was assessed by wound healing assay. RESULTS: E5-siRNA transfection significantly reduced E5 mRNA expression in CaSki cells compared to the control group. The MTT assay revealed that monotherapy with E5-siRNA, Oxaliplatin, or Ifosfamide had moderate effects on cell viability. However, combination therapy showed synergistic effects, reducing the IC50 of Oxaliplatin from 11.42 × 10-8 M (45.36 µg/ml) to 6.71 × 10-8 M (26.66 µg/ml) and Ifosfamide from 12.52 × 10-5 M (32.7 µg/ml) to 8.206 × 10-5 M (21.43 µg/ml). Flow cytometry analysis demonstrated a significant increase in apoptosis for combination treatments, with apoptosis rates rising from 11.02 % (Oxaliplatin alone) and 16.98 % (Ifosfamide alone) to 24.8 % (Oxaliplatin + E5-siRNA) and 34.9 % (Ifosfamide + E5-siRNA). The sub-G1 cell population increased from 15.7 % (Oxaliplatin alone) and 18 % (Ifosfamide alone) to 21.9 % (Oxaliplatin + E5-siRNA) and 27.1 % (Ifosfamide + E5-siRNA), indicating cell cycle arrest. The colony formation assay revealed a substantial decrease in the number of colonies following combination treatment. qRT-PCR analysis showed decreased expression of stemness-related genes CD44 and Nanog, and migration-related genes MMP2 and CXCL8 in the combination groups. Apoptosis-related genes Casp-3, Casp-9, and pP53 showed increased expression following combination therapy, while BAX expression increased and BCL2 expression decreased relative to the control. CONCLUSION: The study demonstrates that combining E5-siRNA with Oxaliplatin or Ifosfamide enhances the efficacy of chemotherapy in HPV-16 positive cervical cancer cells. This synergistic approach effectively targets multiple aspects of cancer cell behavior, including proliferation, apoptosis, migration, and stemness. The findings suggest that this combination strategy could potentially allow for lower chemotherapy doses, thereby reducing toxicity while maintaining therapeutic efficacy. This research provides valuable insights into targeting HPV E5 as a complementary approach to existing therapies focused on E6 and E7 oncoproteins, opening new avenues for combination therapies in cervical cancer treatment.


Subject(s)
Apoptosis , Human papillomavirus 16 , Ifosfamide , Oxaliplatin , RNA, Small Interfering , Uterine Cervical Neoplasms , Humans , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/virology , Oxaliplatin/pharmacology , Female , RNA, Small Interfering/genetics , Cell Line, Tumor , Ifosfamide/pharmacology , Apoptosis/drug effects , Human papillomavirus 16/genetics , Papillomavirus Infections/drug therapy , Papillomavirus Infections/genetics , Papillomavirus Infections/virology , Cell Survival/drug effects , Oncogene Proteins, Viral/genetics , Cell Movement/drug effects , Cell Proliferation/drug effects , Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects
2.
Biomaterials ; 313: 122766, 2025 Feb.
Article in English | MEDLINE | ID: mdl-39180916

ABSTRACT

The immune resistance of tumor microenvironment (TME) causes immune checkpoint blockade therapy inefficient to hepatocellular carcinoma (HCC). Emerging strategies of using chemotherapy regimens to reverse the immune resistance provide the promise for promoting the efficiency of immune checkpoint inhibitors. The induction of cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) in tumor cells evokes the adaptive immunity and remodels the immunosuppressive TME. In this study, we report that mitoxantrone (MIT, a chemotherapeutic drug) activates the cGAS-STING signaling pathway of HCC cells. We provide an approach to augment the efficacy of MIT using a signal transducer and activator of transcription 3 (STAT3) inhibitor called napabucasin (NAP). We prepare an aminoethyl anisamide (AEAA)-targeted polyethylene glycol (PEG)-modified poly (lactic-co-glycolic acid) (PLGA)-based nanocarrier for co-delivery of MIT and NAP. The resultant co-nanoformulation can elicit the cGAS-STING-based immune responses to reshape the immunoresistant TME in the mice orthotopically grafted with HCC. Consequently, the resultant co-nanoformulation can promote anti-PD-1 antibody for suppressing HCC development, generating long-term survival, and inhibiting tumor recurrence. This study reveals the potential of MIT to activate the cGAS-STING signaling pathway, and confirms the feasibility of nano co-delivery for MIT and NAP on achieving HCC chemo-immunotherapy.


Subject(s)
Carcinoma, Hepatocellular , Immunotherapy , Liver Neoplasms , Membrane Proteins , Mitoxantrone , Nucleotidyltransferases , STAT3 Transcription Factor , Mitoxantrone/pharmacology , Mitoxantrone/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Animals , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Humans , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism , STAT3 Transcription Factor/metabolism , Mice , Immunotherapy/methods , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Benzofurans , Naphthoquinones
3.
Biomaterials ; 312: 122736, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39121728

ABSTRACT

The resurgence of influenza viruses as a significant global threat emphasizes the urgent need for innovative antiviral strategies beyond existing treatments. Here, we present the development and evaluation of a novel super-multivalent sialyllactosylated filamentous phage, termed t-6SLPhage, as a potent entry blocker for influenza A viruses. Structural variations in sialyllactosyl ligands, including linkage type, valency, net charge, and spacer length, were systematically explored to identify optimal binding characteristics against target hemagglutinins and influenza viruses. The selected SLPhage equipped with optimal ligands, exhibited exceptional inhibitory potency in in vitro infection inhibition assays. Furthermore, in vivo studies demonstrated its efficacy as both a preventive and therapeutic intervention, even when administered post-exposure at 2 days post-infection, under 4 lethal dose 50% conditions. Remarkably, co-administration with oseltamivir revealed a synergistic effect, suggesting potential combination therapies to enhance efficacy and mitigate resistance. Our findings highlight the efficacy and safety of sialylated filamentous bacteriophages as promising influenza inhibitors. Moreover, the versatility of M13 phages for surface modifications offers avenues for further engineering to enhance therapeutic and preventive performance.


Subject(s)
Antiviral Agents , Animals , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Humans , Dogs , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/drug therapy , Influenza A virus/drug effects , Influenza A virus/physiology , Madin Darby Canine Kidney Cells , Inovirus/drug effects , Oseltamivir/pharmacology , Oseltamivir/chemistry , Mice , Influenza, Human/virology , Influenza, Human/drug therapy , Mice, Inbred BALB C , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/metabolism , Female
4.
Cancer Lett ; 602: 217214, 2024 Oct 10.
Article in English | MEDLINE | ID: mdl-39218291

ABSTRACT

Protein arginine methylation is a common post-translational modification (PTM) catalyzed by nine protein arginine methyltransferases (PRMTs). As the major symmetric arginine methyltransferase that methylates both histone and non-histone substrates, PRMT5 plays key roles in a number of biological processes critical for development and tumorigenesis. PRMT5 overexpression has been reported in multiple cancer types including prostate cancer (PCa), but the exact biological and mechanistic understanding of PRMT5 in aggressive PCa remains ill-defined. Here, we show that PRMT5 is upregulated in PCa, correlates with worse patient survival, promotes corrupted RNA splicing, and functionally cooperates with an array of pro-tumorigenic pathways to enhance oncogenesis. PRMT5 inhibition via either genetic knockdown or pharmacological inhibition reduces stemness with paralleled differentiation and arrests cell cycle progression without causing appreciable apoptosis. Strikingly, the severity of antitumor effect of PRMT5 inhibition correlates with disease aggressiveness, with AR+ PCa being less affected. Molecular characterization pinpoints MYC, but not (or at least to a lesser degree) AR, as the main partner of PRMT5 to form a positive feedback loop to exacerbate malignancy in both AR+ and AR- PCa cells. Inspired by the surprising finding that PRMT5 negatively correlates with tumor immune infiltration and transcriptionally suppresses an immune-gene program, we further show that although PRMT5 inhibitor (PRMT5i) EPZ015666 or anti-PD-1 immunotherapy alone exhibits limited antitumor effects, combination of PRMT5i with anti-PD-1 displays superior efficacy in inhibiting castration-resistant PCa (CRPC) in vivo. Finally, to expand the potential use of PRMT5i through a synthetic lethality concept, we also perform a global CRISPR/Cas9 knockout screen to unravel that many clinical-grade drugs of known oncogenic pathways can be repurposed to target CRPC when used in combination with PRMT5i at low doses. Collectively, our findings establish a rationale to exploit PRMT5i in combination with immunotherapy or other targeted therapies to treat aggressive PCa.


Subject(s)
Prostatic Neoplasms , Protein-Arginine N-Methyltransferases , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Male , Humans , Animals , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/drug therapy , Cell Line, Tumor , Mice , Cell Proliferation/drug effects , Xenograft Model Antitumor Assays , Immunotherapy/methods , Gene Expression Regulation, Neoplastic
5.
Dig Liver Dis ; 2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39307602

ABSTRACT

BACKGROUND: A significant proportion of inflammatory bowel disease (IBD) patients fail to respond to advanced therapies. Combining advanced therapies may improve treatment outcome. This study aimed to assess the effectiveness, adverse events, and costs associated with combining advanced therapies in IBD patients. METHODS: Combination advanced therapy was defined as the concurrent use of two biological agents or one biological agent with a small molecule therapy. Clinical data, including disease characteristics, treatment regimens, and adverse events, were collected from electronic patient records. Clinical response rates, biochemical markers, and treatment costs were evaluated. RESULTS: The study included 109 IBD patients receiving combination advanced therapies from 9 academic centers in Ireland. Corticosteroid-free clinical response rates at 12 weeks and 52 weeks were 39 % and 38 %, respectively. Adverse events occurred in 26 % of therapeutic trials, with disease-related events being the most common. Notably, there were 3 cases of non-melanomatous skin cancer and 10 infectious complications. The annual cost of maintenance therapy for combination advanced therapies ranged from €17,560 to €30,724 per patient. CONCLUSION: Combination advanced therapies demonstrated effectiveness and acceptable safety profiles in a cohort of treatment-refractory IBD patients. Further large, prospective trials are required to definitively evaluate the role of combination advanced therapies in IBD.

6.
Molecules ; 29(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39274943

ABSTRACT

In this study, we report a novel platinum-doxorubicin conjugate that demonstrates superior therapeutic indices to cisplatin, doxorubicin, or their combination, which are commonly used in cancer treatment. This new molecular structure (1) was formed by conjugating an amphiphilic Pt(IV) prodrug of cisplatin with doxorubicin. Due to its amphiphilic nature, the Pt(IV)-doxorubicin conjugate effectively penetrates cell membranes, delivering both cisplatin and doxorubicin payloads intracellularly. The intracellular accumulation of these payloads was assessed using graphite furnace atomic absorption spectrometry and fluorescence imaging. Since the therapeutic effects of cisplatin and doxorubicin stem from their ability to target nuclear DNA, we hypothesized that the amphiphilic Pt(IV)-doxorubicin conjugate (1) would effectively induce nuclear DNA damage toward killing cancer cells. To test this hypothesis, we used flow the cytometric analysis of phosphorylated H2AX (γH2AX), a biomarker of nuclear DNA damage. The Pt(IV)-doxorubicin conjugate (1) markedly induced γH2AX in treated MDA-MB-231 breast cancer cells, showing higher levels than cells treated with either cisplatin or doxorubicin alone. Furthermore, MTT cell viability assays revealed that the enhanced DNA-damaging capability of complex 1 resulted in superior cytotoxicity and selectivity against human cancer cells compared to cisplatin, doxorubicin, or their combination. Overall, the development of this amphiphilic Pt(IV)-doxorubicin conjugate represents a new form of combination therapy with improved therapeutic efficacy.


Subject(s)
Cisplatin , Doxorubicin , Cisplatin/pharmacology , Humans , Doxorubicin/pharmacology , Doxorubicin/chemistry , Cell Line, Tumor , DNA Damage/drug effects , Cell Survival/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Platinum/chemistry , Platinum/pharmacology , Prodrugs/pharmacology , Prodrugs/chemistry , Histones
7.
Int J Biol Macromol ; 280(Pt 1): 135713, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39293631

ABSTRACT

Imaging, silencing cancer-related microRNA, and chemotherapy-phototherapy (CTPT) combination therapy are crucial for cancer diagnosis and drug resistance overcoming. In this study, we designed a multifunctional DNA tetrahedron (MB-MUC1-TD) for the targeted delivery of combined daunorubicin (DAU) + toluidine blue O (TBO). The detection limit of miRNA-21 was determined to be 0.91 nM. The intercalation of DAU and TBO into MB-MUC1-TD was proved by spectroscopic and calorimetric methods. The thermodynamic parameters for the interactions of DAU and/or TBO with MB-MUC1-TD confirmed high drug loading. The first addition of TBO in the ternary system achieved a higher loading of both drugs and a more stable complex structure. Deoxyribonuclease I (DNase I) accelerated the release of DAU and/or TBO loaded in MB-MUC1-TD. Confocal laser scanning microscope demonstrated that MB-MUC1-TD exhibited good imaging ability for miRNA-21 to accurately identify cancer cells, and DAU/TBO was predominantly distributed within the nucleus of cancer cells. In vitro cytotoxicity showed better gene therapy efficacy of MB on MCF-7 cells, better biocompatibility of loaded DAU and TBO on LO2 cells, and stronger synergistic cytotoxicity of DAU + TBO on MCF-7/ADR cells. This study may establish a theoretical foundation for co-loading CTPT combination drugs based on multifunctional DNA nanostructures.

8.
Front Pharmacol ; 15: 1401000, 2024.
Article in English | MEDLINE | ID: mdl-39295944

ABSTRACT

Glioblastoma (GBM) is the most common and aggressive malignant brain tumor among adults. Despite advancements in multimodality therapy for GBM, the overall prognosis remains poor, with an extremely high risk of recurrence. Currently, there is no established consensus on the optimal treatment option for recurrent GBM, which may include reoperation, reirradiation, chemotherapy, or a combination of the above. Bevacizumab is considered a first-line treatment option for recurrent GBM, as is temozolomide. However, in recurrent GBM, it is necessary to balance the risks and benefits of reirradiation in combination with bevacizumab and temozolomide. Herein, we report the case of a patient with recurrent GBM after standard treatment who benefited from stereotactic radiotherapy followed by bevacizumab and temozolomide maintenance therapy. Following 16 months of concurrent chemoradiotherapy (CCRT), the patient was diagnosed with recurrent GBM by a 3-T contrast-enhanced magnetic resonance imaging (MRI). The addition of localized radiotherapy to the ongoing treatment regimen of bevacizumab, in combination with temozolomide therapy, prolonged the patient's disease-free survival to over 2 years, achieving a significant long-term outcome, with no notable adverse effects observed. This clinical case may provide a promising new option for patients with recurrent GBM.

9.
MycoKeys ; 108: 227-247, 2024.
Article in English | MEDLINE | ID: mdl-39296990

ABSTRACT

Metacampanella is an important genus in the Marasmiaceae family. We collected specimens during our investigations in China and Mongolia. Through morphological and molecular phylogenetic analyses, we identified two new species of this genus: Metacampanellasubtricolor and Metacampanellacoprophila. In addition, we identified Metacampanellatricolor as a novel combination. Molecular systematic studies support these results. Illustrated descriptions, taxonomic discussions, and keys to the genus are provided.

10.
J Hazard Mater ; 480: 135850, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39298945

ABSTRACT

Advanced oxidation processes (AOPs) using persulfate system can effectively remove organic pollutants. However, dissolved organic matter (DOM) has multiple effects on AOPs efficiency, and the influence of DOMs from natural sources on AOPs is still unclear. In this study, we explored the effects of soil DOM (SDOM) and fertilizer DOM (FDOM) on tetracycline (TC) removal by persulfate systems. DOMs introduction decreased light transmittance, slightly increased the pH of the systems, and destroyed original adsorption-desorption equilibrium. SDOM promoted most reactive species generation in the initial stage, thus improving the initial TC degradation rate. However, introduction of SDOM and FDOM increased the final TC residual rate. FDOM produced more obvious inhibitory effects on TC degradation. The final TC residual rates in systems containing 7.5 and 15 mg L-1 FDOM (F7.5-TC-PS and F15-TC-PS, respectively) were 25.85 % and 25.52 %, respectively. The inhibitory effects of FDOM on TC degradation were related to the combination between TC and FDOM, with humic acid-like component in FDOM being the main contributor. Besides, the main components in DOMs underwent transformation in the persulfate systems. This study sought to provide insights into the regulatory effects of DOM on TC photo-assisted conversion by AOPs.

11.
Virol Sin ; 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39299564

ABSTRACT

Oncolytic virus (OV) is increasingly being recognized as a novel vector in cancer immunotherapy. Increasing evidence suggests that OV has the ability to change the immune status of tumor microenvironment, so called transformation of 'cold' tumors into 'hot' tumors. The improved anti-tumor immunity can be induced by OV and further enhanced through the combination of various immunomodulators. The Neo-2/15 is a newly de novo synthesized cytokine that functions as both IL-2 and IL-15. However, it specifically lacks the binding site of IL-2 receptor α subunit (CD25), therefore unable to induce the Treg proliferation. In present study, a recombinant vesicular stomatitis virus expressing the Neo-2/15 (VSVM51R-Neo-2/15) was generated. Intratumoral delivery of VSVM51R-Neo-2/15 efficiently inhibited tumor growth in mice without causing the IL-2-related toxicity previously observed in clinic. Moreover, treatment with VSVM51R-Neo-2/15 increased the number of activated CD8+ T cells but not Treg cells in tumors. More tumor-bearing mice were survival with VSVM51R-Neo-2/15 treatment, and the surviving mice displayed enhanced protection against tumor cell rechallenge due to the induced anti-tumor immunity. In addition, combination therapy of OV and anti-PD-L1 immune checkpoint inhibitors further enhanced the anti-tumor immune response. These findings suggest that our novel VSVM51R-Neo-2/15 can effectively inhibit the tumor growth and enhance the sensitivity to immune checkpoint inhibitors, providing promising attempts for further clinical trials.

12.
Infection ; 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300000

ABSTRACT

PURPOSE: Infective endocarditis caused by non-HACEK gram-negative bacilli (GNB-IE) is rare but associated with significant morbidity and case fatality. Evidence on optimal treatment and management is limited. We aimed to describe the characteristics and management of GNB-IE patients, investigating factors associated with disease acquisition and unfavorable outcomes. METHODS: We conducted a retrospective descriptive single-center study (tertiary care and referral hospital) between 2015 and 2021, including adult patients with definite GNB-IE. We reviewed demographic, clinical and microbiological data, focusing on predisposing factors, clinical outcomes and 1-year mortality. RESULTS: Of 1093 patients with probable or definite IE, 19 patients (median age 69 years) had definite GNB-IE, with an increasing incidence throughout the study period. Median age-adjusted Charlson Comorbidity Index score was 4 points. Prosthetic valve IE (PVIE) was present in 7/19 (37%) patients. Nosocomial acquisition occurred in 8/19 (42%) patients. Escherichia coli and Klebsiella pneumoniae were the most common pathogens. Beta-lactam (BL) based combination therapy was applied in 12/19 (63%) patients (58% BL + fluoroquinolone, 42% BL + aminoglycoside). Cardiac surgery was required in 8/19 (42%) patients (PVIE 71%, native valve IE 25%), primarily for embolism prevention and heart failure. Complications occurred in 14/19 (74%) patients. The in-hospital mortality rate was 21% (4/19); the one-year mortality rate was 44% (7/16). One-year mortality did not significantly differ between patients who underwent cardiac surgery and patients managed with anti-infective treatment alone (p = 0.633). CONCLUSIONS: GNB-IE affects elderly patients with high comorbidity levels and recent health-care exposure. GNB-IE was associated with high complication rates and high mortality.

13.
PeerJ Comput Sci ; 10: e2252, 2024.
Article in English | MEDLINE | ID: mdl-39314736

ABSTRACT

The world faces the ongoing challenge of terrorism and extremism, which threaten the stability of nations, the security of their citizens, and the integrity of political, economic, and social systems. Given the complexity and multifaceted nature of this phenomenon, combating it requires a collective effort, with tailored methods to address its various aspects. Identifying the terrorist organization responsible for an attack is a critical step in combating terrorism. Historical data plays a pivotal role in this process, providing insights that can inform prevention and response strategies. With advancements in technology and artificial intelligence (AI), particularly in military applications, there is growing interest in utilizing these developments to enhance national and regional security against terrorism. Central to this effort are terrorism databases, which serve as rich resources for data on armed organizations, extremist entities, and terrorist incidents. The Global Terrorism Database (GTD) stands out as one of the most widely used and accessible resources for researchers. Recent progress in machine learning (ML), deep learning (DL), and natural language processing (NLP) offers promising avenues for improving the identification and classification of terrorist organizations. This study introduces a framework designed to classify and predict terrorist groups using bidirectional recurrent units and self-attention mechanisms, referred to as BiGRU-SA. This approach utilizes the comprehensive data in the GTD by integrating textual features extracted by DistilBERT with features that show a high correlation with terrorist organizations. Additionally, the Synthetic Minority Over-sampling Technique with Tomek links (SMOTE-T) was employed to address data imbalance and enhance the robustness of our predictions. The BiGRU-SA model captures temporal dependencies and contextual information within the data. By processing data sequences in both forward and reverse directions, BiGRU-SA offers a comprehensive view of the temporal dynamics, significantly enhancing classification accuracy. To evaluate the effectiveness of our framework, we compared ten models, including six traditional ML models and four DL algorithms. The proposed BiGRU-SA framework demonstrated outstanding performance in classifying 36 terrorist organizations responsible for terrorist attacks, achieving an accuracy of 98.68%, precision of 96.06%, sensitivity of 96.83%, specificity of 99.50%, and a Matthews correlation coefficient of 97.50%. Compared to state-of-the-art methods, the proposed model outperformed others, confirming its effectiveness and accuracy in the classification and prediction of terrorist organizations.

14.
Ther Adv Musculoskelet Dis ; 16: 1759720X241274055, 2024.
Article in English | MEDLINE | ID: mdl-39314822

ABSTRACT

Psoriatic arthritis (PsA) is an immune-mediated inflammatory disease with heterogeneity regarding its clinical features, mainly affecting the skin and the musculoskeletal system; additionally, extra-musculoskeletal manifestations and comorbidities are common, adding complexity to its treatment. In the last decades, a plethora of therapeutic options have been available, including conventional synthetic disease-modifying anti-rheumatic drugs (DMARDs), biological DMARDs (bDMARDs), and targeted synthetic DMARDs (tsDMARDs), and many recommendations have been published regarding the proper use of them in patients with PsA. In rheumatoid arthritis, the combination of conventional with bDMARDs or tsDMARDs is a common and recommended practice, whereas in PsA there is scarce data about the benefit of this combination. This review summarizes all the available data from randomized clinical trials, observational studies, and registries about the value of this therapeutic strategy.


Use of b/tsDMARDs in PsA: with or without csDMARDs Over the last years, many different b/ts DMARDs have been porven to be efficacious in psoriatic arthritis (PsA). Although in rheumatoid arthritis, it is established that most of these drugs work better in combination with conventional synthetic DMARDs (e.g methotrexate), this seems to be slightly different in PsA. Herein, we review the current literature about the combination therapy versus monotherapy of b/ts DMARDs in PsA. We present the results of this narrative review in a structured (per drug category) way, so that it is easier for the reader to find relevant information. There is no doubt that the currently available treatment options in PsA have changed the course of the disease and improved the functional status of the patients. However, as there is still a substantial proportion of patients who do not achieve remission or low disease activity, the need to find effective therapeutic regimens or follow different strategies is growing. In this direction, the combination of a conventional synthetic with biological or targeted synthetic DMARD does not seem to be more effective than the monotherapy of the latter. This seems to be more pronounced in the newer drug categories (anti-IL-17, anti-IL23 and JAKi) compared to the TNFi, where the co-administration of a csDMARD improves their survival.

15.
Chem Biol Drug Des ; 104(3): e14627, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39317691

ABSTRACT

Breast cancer (BC) is one of the leading causes of high mortality rates in women worldwide. Although advancements have been made in the design of therapeutic strategies and drug discovery, drug resistance remains one of the key challenges. One of the ways to overcome drug resistance is finding potential drug combinations since the efficacy of combined drugs is higher than their individual efficacies if the combination is a synergistic pair. Therefore, the current study uses a BC patient-derived xenograft (PDX) dataset to evaluate the effects of various cancer drugs on breast cancer in vivo models. The drug effects are further validated by four machine learning models, namely Elastic Net, Least Absolute Shrinkage and Selection (LASSO), Support Vector Machine (SVM), Random Forests (RF), as well as exploring the shortlisted drugs in combination with paclitaxel, a baseline drug for enhanced efficacy on tumor volume reduction. Additionally, the study also shortlists the top 50 in vivo biomarkers correlated with the effects of the drugs. The outcomes could be significantly important for the design of an effective anti-breast cancer therapy.


Subject(s)
Biomarkers, Tumor , Breast Neoplasms , Paclitaxel , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Humans , Female , Animals , Biomarkers, Tumor/metabolism , Mice , Support Vector Machine , Xenograft Model Antitumor Assays , Machine Learning , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
16.
Article in English | MEDLINE | ID: mdl-39318019

ABSTRACT

BACKGROUND: Glucose-regulated protein 78 (GRP78), as a chaperone protein, can protect the endoplasmic reticulum of cells and is expressed to influence chemoresistance and prognosis in cancer. Deoxypodophyllotoxin (DPT) is a compound with antitumor effects on cancers. DPT inhibits the proliferation of osteosarcoma by inducing apoptosis, necrosis, or cell cycle arrest. OBJECT: This study was performed to demonstrate the molecular mechanism by which DPT attenuates osteosarcoma progression through GRP78. METHODS: Natural compound libraries and western blot (WB) were used to screen the inhibitors of osteosarcoma GRP78. The expression of mitochondria-related genes in cancer cells of the treatment group was detected by quantitative real-time PCR (qPCR) and WB. 3-(4,5)- Dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide (MTT) and 5-ethynyl-2'- deoxyuridine (EDU) were used to discover the activity and proliferation of osteosarcoma cells treated with DPT. We constructed an in vivo mouse model of DPT drug therapy and carried out immunohistochemical detection of xenografts. The treated osteosarcoma cells were analyzed using bioinformatics and electron microscopy. The data were analyzed finally. RESULTS: DPT inhibited osteosarcoma cell survival and the growth of tumor xenografts. It promoted up-regulation of BCL2-associated X protein (Bax) and B-cell CLL/lymphoma 2 (Bcl-2), which serves to mediate and attenuate, respectively, the killing activities of DPT through mitochondria dysfunction. The effect of DPT against cancer cells could be attenuated by the overexpression of GRP78, characterized by the inactivation of the caspase cascade. The loss of GRP78 in osteosarcoma cells negatively mediated the basal level of autophagyassociated genes. DPT stimulated autophagy via the phosphoinositide 3-kinase (PI3K)-v-akt murine thymoma viral oncogene homolog (AKT), a mechanistic target of rapamycin (mTOR) axis. The autophagy caused by DPT played an active role in the osteosarcoma of humans and blocked the apoptotic cascade. CONCLUSION: Combination treatment with the GRP78 inhibitor DPT and pharmacological autophagy inhibitors will be a meaningful method of obviating osteosarcoma cells.

17.
Eur J Neurol ; 31 Suppl 2: e16434, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39318200

ABSTRACT

BACKGROUND: Varied responses to acute migraine medications have been observed, with over one-third (34.5%) of patients reporting insufficient headache relief. Sumatriptan-naproxen sodium, a single, fixed-dose combination tablet comprising sumatriptan 85 mg and naproxen sodium 500 mg, was developed with the rationale of targeting multiple putative mechanisms involved in the pathogenesis of migraine to optimise acute migraine care. METHODS: A narrative review of clinical trials investigating sumatriptan-naproxen sodium for both adults and adolescents was performed in March 2024. RESULTS: Across a total of 14 clinical trials in nine publications, sumatriptan-naproxen sodium offered greater efficacy for 2-h pain freedom (14/14) and sustained pain-free response up to 24 h (13/14) compared with monotherapy and/or placebo for both adult and adolescent study participants with an acceptable and well-tolerated adverse effect profile. Clinical trial data also demonstrates the effectiveness of sumatriptan-naproxen sodium in participants with allodynia, probable migraine, menstrual-related migraine and those with poor responses to acute, non-specific, migraine medication. CONCLUSIONS: Multi-mechanistic therapeutic agents offer an opportunity to optimise acute medications by targeting multiple mediators involved in the pathogenesis of migraine. Sumatriptan-naproxen sodium resulted in greater initial and sustained pain freedom, compared with either sumatriptan, naproxen-sodium and/or placebo, for the treatment of single or multiple attacks of migraine across both adult and adolescent study populations.


Subject(s)
Drug Combinations , Migraine Disorders , Naproxen , Sumatriptan , Humans , Sumatriptan/administration & dosage , Sumatriptan/pharmacology , Sumatriptan/therapeutic use , Naproxen/therapeutic use , Naproxen/administration & dosage , Migraine Disorders/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Serotonin 5-HT1 Receptor Agonists/pharmacology , Serotonin 5-HT1 Receptor Agonists/administration & dosage , Serotonin 5-HT1 Receptor Agonists/therapeutic use
18.
ACS Nano ; 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39303096

ABSTRACT

Herein, we constructed a paclitaxel (PTX) prodrug (PA) by conjugating PTX with acrylic acid as a cysteine-depleting agent. The as-synthesized PA can assemble with diacylphosphatidylethanolamine-PEG2000 to form stable nanoparticles (PA NPs). After endocytosis into cells, PA NPs can specifically react with cysteine and trigger release of PTX for chemotherapy. On the other hand, the depletion of cysteine can greatly downregulate the intracellular content of glutathione and lead to oxidative stress outburst-provoking ferroptosis. The released PTX can elicit antitumor immune response by inducing immunogenic cell death, thus promoting dendritic cells maturation and cascaded cytotoxic T lymphocytes activation, which not only produces a robust immunotherapy effect but also synergizes the ferroptosis therapy by inhibiting cysteine transport via the release of interferon-γ in the activated immune system. As a result, PA NPs exhibit favorable in vitro and in vivo antitumor performance with reduced systemic toxicity. Our work highlights the potential of simple molecular design of prodrugs for enhancing the therapeutic efficacy toward malignant cancer.

19.
Adv Sci (Weinh) ; : e2408598, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39303165

ABSTRACT

In the previous studies, anti-TGF-ß/PD-L1 bispecific antibody YM101 is demonstrated, with superior efficacy to anti-PD-L1 monotherapy in multiple tumor models. However, YM101 therapy can not achieve complete regression in most tumor-bearing mice, suggesting the presence of other immunosuppressive elements in the tumor microenvironment (TME) beyond TGF-ß and PD-L1. Thoroughly exploring the TME is imperative to pave the way for the successful translation of anti-TGF-ß/PD-L1 BsAb into clinical practice. In this work, scRNA-seq is employed to comprehensively profile the TME changes induced by YM101. The scRNA-seq analysis reveals an increase in immune cell populations associated with antitumor immunity and enhances cell-killing pathways. However, the analysis also uncovers the presence of immunosuppressive CCR5+ T cells in the TME after YM101 treatment. To overcome this hurdle, YM101 is combined with Maraviroc, a widely used CCR5 antagonist for treating HIV infection, suppressing CCR5+ T cell accumulation, and optimizing the immune response. Mechanistically, YM101-induced neutrophil activation recruits immunosuppressive CCR5+ T cells via CCR5 ligand secretion, creating a feedback loop that diminishes the antitumor response. Maraviroc then cleared these infiltrating cells and offset YM101-mediated immunosuppressive effects, further unleashing the antitumor immunity. These findings suggest selectively targeting CCR5 signaling with Maraviroc represents a promising and strategic approach to enhance YM101 efficacy.

20.
Cancer Radiother ; 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39304400

ABSTRACT

Radiotherapy is widely used to treat various cancers. Its combination with immune checkpoint inhibitors is intensively studied preclinically and clinically. Although the first results were very encouraging, the number of patients who respond positively remains low, and the therapeutic benefit is often temporary. This review summarizes how radiation can stimulate an antitumor immune response and its combination with immunotherapy based on inhibiting immune checkpoints. We will provide an overview of radiotherapy parameters that should be better controlled to avoid downregulating the antitumor immune response. The low response rate of combining radiotherapy and immunotherapy could, at least in part, be caused by the stimulation of cancer cell invasion and metastasis development that occur at similar doses and number of radiation fractions. To end on a positive note, we explore how a targeted inhibition of the inflammatory cytokines induced by radiation with a cyclooxygenase-2 inhibitor could both support an antitumor immune response and block radiation-induced metastasis formation.

SELECTION OF CITATIONS
SEARCH DETAIL