Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63.943
Filter
1.
Biomaterials ; 313: 122796, 2025 Feb.
Article in English | MEDLINE | ID: mdl-39226654

ABSTRACT

Chemotherapy-induced cellular senescence leads to an increased proportion of cancer stem cells (CSCs) in breast cancer (BC), contributing to recurrence and metastasis, while effective means to clear them are currently lacking. Herein, we aim to develop new approaches for selectively killing senescent-escape CSCs. High CD276 (95.60%) expression in multidrug-resistant BC cells, facilitates immune evasion by low-immunogenic senescent escape CSCs. CALD1, upregulated in ADR-resistant BC, promoting senescent-escape of CSCs with an anti-apoptosis state and upregulating CD276, PD-L1 to promote chemoresistance and immune escape. We have developed a controlled-released thermosensitive hydrogel containing pH- responsive anti-CD276 scFV engineered biomimetic nanovesicles to overcome BC in primary, recurrent, metastatic and abscopal humanized mice models. Nanovesicles coated anti-CD276 scFV selectively fuses with cell membrane of senescent-escape CSCs, then sequentially delivers siCALD1 and ADR due to pH-responsive MnP shell. siCALD1 together with ADR effectively induce apoptosis of CSCs, decrease expression of CD276 and PD-L1, and upregulate MHC I combined with Mn2+ to overcome chemoresistance and promote CD8+T cells infiltration. This combined therapeutic approach reveals insights into immune surveillance evasion by senescent-escape CSCs, offering a promising strategy to immunotherapy effectiveness in cancer therapy.


Subject(s)
Breast Neoplasms , Cellular Senescence , Drug Resistance, Neoplasm , Neoplastic Stem Cells , Humans , Animals , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/therapy , Drug Resistance, Neoplasm/drug effects , Female , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Cellular Senescence/drug effects , Cell Line, Tumor , Mice , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Genetic Engineering/methods , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Nanoparticles/chemistry , Single-Chain Antibodies/chemistry , Tumor Escape/drug effects , B7-H1 Antigen/metabolism , Apoptosis/drug effects , Biomimetics/methods , B7 Antigens
2.
Theranostics ; 14(14): 5413-5428, 2024.
Article in English | MEDLINE | ID: mdl-39310094

ABSTRACT

Rationale: Oral chemotherapy has been emerging as a hopeful therapeutic regimen for the treatment of various cancers because of its high safety and convenience, lower costs, and high patient compliance. Despite the current advancements in nanoparticle-mediated drug delivery, numerous anticancer drugs susceptible to the hostile gastrointestinal (GI) environment exhibit poor permeability across the intestinal epithelium, rendering them ineffective in providing therapeutic benefits. In this paper, we focus on harnessing milk-derived extracellular vesicles (mEVs) for gut-to-tumor oral drug delivery by leveraging their high bioavailability. Methods: The tumor-activated prodrug (a cathepsin B-specific cleavable FRRG peptide and doxorubicin, FDX) is used as a model drug and is complexed with mEVs, resulting in FDX@mEVs. To verify stability in the GI tract, prolonged intestinal retention, and enhanced trans-epithelial transport via neonatal Fc receptor (FcRn)-mediated transcytosis, intestinal transport evaluation is conducted using in vitro intestinal barrier model and mouse model. Results: FDX@mEVs form a stable nanostructure with an average diameter of 131.1 ± 70.5 nm and complexation processes do not affect the inherent properties of FDX. Orally administered FDX@mEVs show significantly improved bioavailability compared to uncomplexed FDX via FcRn-mediated transcytosis of mEVs resulting in increased tumor accumulation of FDX in tumor-bearing mouse model. Conclusions: After oral administration of FDX@mEVs, it is observed that remarkable antitumor efficacy in colon tumor-bearing mice without adverse effects, such as body weight loss, liver/kidney dysfunction, and cardiotoxicity.


Subject(s)
Doxorubicin , Extracellular Vesicles , Prodrugs , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Doxorubicin/pharmacokinetics , Animals , Prodrugs/administration & dosage , Prodrugs/pharmacology , Mice , Extracellular Vesicles/metabolism , Administration, Oral , Humans , Milk/chemistry , Drug Delivery Systems/methods , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Biological Availability , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Nanoparticles/chemistry
3.
Pan Afr Med J ; 48: 56, 2024.
Article in English | MEDLINE | ID: mdl-39315063

ABSTRACT

Liposarcoma is a rare primitive mesenchymal tumor, developed at the expense of adipose tissue and with a preferential location in the thigh. We report an observation of liposarcoma in the buttock. A 56-year-old man, presented with a tumor of the right buttock for 2 years. Examination revealed an inflammatory, ulcerated tumor in the upper-external quadrant of the right buttock, measuring about 8 cm. Bilateral inguinal adenopathies were associated. The diagnostic hypotheses were: a squamous cell carcinoma, a cutaneous lymphoma, and cutaneous metastases. An anatomical examination confirmed the diagnosis of myxoid round-cell liposarcoma. The extension work-up appeared compatible with secondary pleuropulmonary, hepatic, cutaneous, and lymph node neoplastic localizations. The patient was treated with chemotherapy with the Adriamycin-carboplatin protocol. The evolution was rapidly fatal after a few weeks after the first course of chemotherapy. It should be evoked in front of any ulcerated tumor of the buttock.


Subject(s)
Liposarcoma, Myxoid , Skin Neoplasms , Humans , Buttocks/pathology , Middle Aged , Male , Liposarcoma, Myxoid/pathology , Liposarcoma, Myxoid/diagnosis , Liposarcoma, Myxoid/drug therapy , Skin Neoplasms/pathology , Skin Neoplasms/diagnosis , Fatal Outcome , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Doxorubicin/administration & dosage
4.
Technol Cancer Res Treat ; 23: 15330338241286755, 2024.
Article in English | MEDLINE | ID: mdl-39311637

ABSTRACT

Breast cancer is the most common malignancy in women worldwide, and major challenges in its treatment include drug resistance and metastasis. Three-dimensional cell culture systems have received widespread attention in drug discovery studies but existing models have limitations, that warrant the development of a simple and repeatable three-dimensional culture model for high-throughput screening. In this study, we designed a simple, reproducible, and highly efficient microencapsulated device to co-culture MCF-7 cells and HUVECs in microcapsules to establish an in vitro vascularized micro-tumor model for chemotherapeutic drug screening. First, to construct a three-dimensional micro-tumor model, cell encapsulation devices were created using three different sizes of flat-mouthed needles. Immunohistochemistry and immunofluorescence assays were conducted to determine vascular lumen formation. Cell proliferation was detected using the Cell Counting Kit-8 assay. Finally, to observe the drug reactions between the models, anticancer drugs (doxorubicin or paclitaxel) were added 12 h after the two-dimensional cultured cells were plated or 7 days after cell growth in the core-shell microcapsules. Vascularized micro-tumors were obtained after 14 days of three-dimensional culture. The proliferation rate in the three-dimensional cultured cells was slower than that of two-dimensional cultured cells. Three-dimensional cultured cells were more resistant to anticancer drugs than two-dimensional cultured cells. This novel sample encapsulation device formed core-shell microcapsules and can be used to successfully construct 3D vascularized micro-tumors in vitro. The three-dimensional culture model may provide a platform for drug screening and is valuable for studying tumor development and angiogenesis.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Coculture Techniques , Drug Screening Assays, Antitumor , Human Umbilical Vein Endothelial Cells , Humans , Cell Proliferation/drug effects , Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor/methods , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Female , MCF-7 Cells , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Paclitaxel/pharmacology , Doxorubicin/pharmacology , Cell Culture Techniques, Three Dimensional/methods
5.
Indian J Public Health ; 68(3): 454-456, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39321241

ABSTRACT

SUMMARY: Malignancies in human immunodeficiency virus (HIV) positive individuals have a larger role in morbidity and mortality. Appropriate clinical acumen is required for a clinician to anticipate the occurrence of lymphoma after starting antiretroviral therapy, especially in patients with CD4 <100 cells/mm3. Here is a 30-year-old man with weight loss and appetite, found to be retroviral disease positive status with low CD 4 counts. He was started on antiretroviral treatment, and following that, he developed Hodgkin's lymphoma of mixed cellularity. He is planned for an ABVD regimen and received one cycle of the same without any complications. To our knowledge, we are reporting the first case of an HIV patient with a mixed cellularity form of classical Hodgkin's lymphoma from India.


Subject(s)
HIV Infections , Hodgkin Disease , Humans , Hodgkin Disease/drug therapy , Male , Adult , HIV Infections/drug therapy , India , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CD4 Lymphocyte Count , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Bleomycin/adverse effects , Bleomycin/administration & dosage , Dacarbazine/adverse effects , Vinblastine/administration & dosage , Vinblastine/adverse effects , Vinblastine/therapeutic use , Anti-Retroviral Agents/therapeutic use , Anti-Retroviral Agents/adverse effects
6.
Cancer Med ; 13(18): e70206, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39324173

ABSTRACT

BACKGROUND: The poor overall survival of osteosarcoma (OS) underscores the need to explore new therapeutic avenues. Tumor necrosis rate (TNR) after neoadjuvant chemotherapy predicts prognosis. AIMS: The study was to investigate safety and activity of neoadjuvant chemotherapy with camrelizumab (a humanized antibody against PD-1) in patients with resectable OS. MATERIALS & METHODS: We conducted a prospective, single-arm, exploratory phase II trial in OS patients. Eligible patients received camrelizumab combined with doxorubicin or liposomal doxorubicin, cisplatin, methotrexate, ifosfamide with mesna. Surgery was performed 12-14 days after neoadjuvant therapy and adjuvant therapy starting 2-3 weeks postoperatively. The primary endpoint was the rate of good tumor necrosis (TNR ≥90%) after neoadjuvant therapy, and the secondary outcomes were safety, 2-year progression free survival and 2-year overall survival. RESULTS: Seventy-five patients were recruited to the study. Subsequently, 64 patients completed neoadjuvant therapy and underwent surgery. Thirty-one patients (48.4%) have a good TNR to neoadjuvant therapy. With a median follow-up of 22.4 months (range 2.2-44.9 months), the estimated 2-year PFS was 69.6% and the estimated 2-year overall survival was 89.4%. Grade 3 or 4 treatment-related adverse events were noticed in 62.7% of the patients. Frequent grade 3 or 4 adverse events were decreased platelet count (45.3%), decreased white blood cell count (36%). No immune-related serious adverse events were observed. DISCUSSION: Our study had limitations. First, it was limited by its non-randomized design. Besides, stromal tumor-infiltrating lymphocytes was comprehensively analyzed in this study. CONCLUSIONS: This study demonstrated that amrelizumab combined with adriamycin, cisplatin, methotrexate, and ifosfamide in the neoadjuvant treatment of resectable OS was safe and tolerable. This combined therapeutic strategy may not increase TNR, but the long-term survival benefit remains to be followed up.


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols , Cisplatin , Doxorubicin , Ifosfamide , Methotrexate , Neoadjuvant Therapy , Osteosarcoma , Humans , Female , Osteosarcoma/drug therapy , Osteosarcoma/mortality , Osteosarcoma/therapy , Osteosarcoma/surgery , Male , Methotrexate/administration & dosage , Methotrexate/therapeutic use , Ifosfamide/administration & dosage , Ifosfamide/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Neoadjuvant Therapy/methods , Cisplatin/administration & dosage , Cisplatin/therapeutic use , Adult , Prospective Studies , Young Adult , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Adolescent , Bone Neoplasms/drug therapy , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Middle Aged
7.
Sci Rep ; 14(1): 22514, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39341955

ABSTRACT

Bioconjugation is one of the most promising strategies to improve drug delivery, especially in cancer therapy. Biomolecules such as bile acids (BAs) have been intensively explored as carriers, due to their peculiar physicochemical properties and biocompatibility. BAs trafficking is regulated by intracellular lipid-binding proteins and their transport in the liver can be studied using chicken liver Bile Acid-Binding Proteins (cL-BABPs) as a reference model. Therefore, we conceived the idea of developing a BA-conjugate with Mirin, an exonuclease inhibitor of Mre11 endowed with different anticancer activities, to direct its transport to the liver. Following computational analysis of various BAs in complex with cL-BABP, we identified cholic acid (CA) as the most promising candidate as carrier, leading to the synthesis of a novel bioconjugate named CA-M11. As predicted by computational data and confirmed by X-ray crystallographic studies, CA-M11 was able to accommodate into the binding pocket of BABP. Hence, it can enter BAs trafficking in the hepatic compartment and here release Mirin. The effect of CA-M11, evaluated in combination with varying concentrations of Doxorubicin on HepG2 cell line, demonstrated a significant increase in cell mortality compared to the use of the cytotoxic drug or Mirin alone, thus highlighting chemo-sensitizing properties. The promising results regarding plasma stability for CA-M11 validate its potential as a valuable agent or adjuvant for hepatic cancer therapy.


Subject(s)
Carrier Proteins , Cholic Acid , Liver Neoplasms , Humans , Cholic Acid/chemistry , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Hep G2 Cells , Animals , Carrier Proteins/metabolism , Doxorubicin/pharmacology , Chickens , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Liver/metabolism , Liver/drug effects , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/chemistry , Membrane Glycoproteins
8.
Int J Mol Sci ; 25(18)2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39337470

ABSTRACT

The potential of RNA-based liquid biopsy is increasingly being recognized in diffuse large B-cell lymphoma (DLBCL), the most common subtype of non-Hodgkin's lymphoma. This study explores the cell-free transcriptome in a humanized DLBCL patient-derived tumor xenograft (PDTX) model. Blood plasma samples (n = 171) derived from a DLBCL PDTX model, including 27 humanized (HIS) PDTX, 8 HIS non-PDTX, and 21 non-HIS PDTX non-obese diabetic (NOD)-scid IL2Rgnull (NSG) mice were collected during humanization, xenografting, treatment, and sacrifice. The mice were treated with either rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP), CD20-targeted human IFNα2-based AcTaferon combined with CHOP (huCD20-Fc-AFN-CHOP), or phosphate-buffered saline (PBS). RNA was extracted using the miRNeasy serum/plasma kit and sequenced on the NovaSeq 6000 platform. RNA sequencing data of the formalin-fixed paraffin-embedded (FFPE) tissue and blood plasma samples of the original patient were included. Flow cytometry was performed on immune cells isolated from whole blood, spleen, and bone marrow. Bulk deconvolution was performed using the Tabula Sapiens v1 basis matrix. Both R-CHOP and huCD20-Fc-AFN-CHOP were able to control tumor growth in most mice. Xenograft tumor volume was strongly associated with circulating tumor RNA (ctRNA) concentration (p < 0.001, R = 0.89), as well as with the number of detected human genes (p < 0.001, R = 0.79). Abundance analysis identified tumor-specific biomarkers that were dynamically tracked during tumor growth or treatment. An 8-gene signature demonstrated high accuracy for assessing therapy response (AUC 0.92). The tumoral gene detectability in the ctRNA of the PDTX-derived plasma was associated with RNA abundance levels in the patient's tumor tissue and blood plasma (p < 0.001), confirming that tumoral gene abundance contributes to the cell-free RNA (cfRNA) profile. Decomposing the transcriptome, however, revealed high inter- and intra-mouse variability, which was lower in the HIS PDTX mice, indicating an impact of human engraftment on the stability and profile of cfRNA. Immunochemotherapy resulted in B cell depletion, and tumor clearance was reflected by a decrease in the fraction of human CD45+ cells. Lastly, bulk deconvolution provided complementary biological insights into the composition of the tumor and circulating immune system. In conclusion, the blood plasma-derived transcriptome serves as a biomarker source in a preclinical PDTX model, enables the assessment of biological pathways, and enhances the understanding of cfRNA dynamics.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Cyclophosphamide , Lymphoma, Large B-Cell, Diffuse , Mice, Inbred NOD , Mice, SCID , Prednisone , Rituximab , Transcriptome , Vincristine , Xenograft Model Antitumor Assays , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Animals , Humans , Mice , Liquid Biopsy/methods , Rituximab/therapeutic use , Prednisone/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Doxorubicin , Biomarkers, Tumor/genetics , Male , Cell-Free Nucleic Acids/genetics , Cell-Free Nucleic Acids/blood , Female , Gene Expression Regulation, Neoplastic
9.
Molecules ; 29(18)2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39339277

ABSTRACT

The aim of this study was to obtain new, safe, and effective compounds with anticancer activity since cancer is still the leading cause of mortality worldwide. The rational design of new compounds was based on the introduction of differentially substituted phenylpiperazines into the 1,2-benzothiazine scaffold as a reference for the structures of recent topoisomerase II (Topo II) inhibitors such as dexrazoxane and XK-469. The newly designed group of 1,2-benzothiazine derivatives was synthesized and tested on healthy (MCF10A) and cancer (MCF7) cell lines, alone and in combination with doxorubicin (DOX). In addition, molecular docking studies were performed both to the DNA-Topo II complex and to the minor groove of DNA. Most of the tested compounds showed cytotoxic activity comparable to doxorubicin, a well-known anticancer drug. The compound BS230 (3-(4-chlorobenzoyl)-2-{2-[4-(3,4-dichlorophenyl)-1-piperazinyl]-2-oxoethyl}-4-hydroxy-2H-1,2-benzothiazine 1,1-dioxide) showed the best antitumor activity with lower cytotoxicity towards healthy cells and at the same time stronger cytotoxicity towards cancer cells than DOX. Moreover, molecular docking studies showed that BS230 has the ability to bind to both the DNA-Topo II complex and the minor groove of DNA. Binding of the minor groove to DNA was also proven by fluorescence spectroscopy.


Subject(s)
Antineoplastic Agents , Drug Design , Molecular Docking Simulation , Thiazines , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Thiazines/chemistry , Thiazines/pharmacology , Thiazines/chemical synthesis , Cell Line, Tumor , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/chemical synthesis , DNA Topoisomerases, Type II/metabolism , DNA Topoisomerases, Type II/chemistry , Structure-Activity Relationship , Piperazines/chemistry , Piperazines/pharmacology , Piperazines/chemical synthesis , MCF-7 Cells , Doxorubicin/pharmacology , Doxorubicin/chemistry , Molecular Structure , Drug Screening Assays, Antitumor , Cell Proliferation/drug effects
10.
Nutrients ; 16(18)2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39339777

ABSTRACT

It has been demonstrated that filbertone, the principal flavor compound of hazelnuts, exhibits preventive effects against hypothalamic inflammation, obesity, neurodegenerative diseases, and muscle lipid accumulation. However, its influence on muscle aging has yet to be elucidated. The objective of this study was to investigate the effects of filbertone on muscle aging in C2C12 myotubes subjected to senescence induction by either doxorubicin or hydrogen peroxide. To ascertain the mechanisms by which filbertone exerts its effects, we conducted a series of experiments, including Western blot analysis, reverse transcription quantitative polymerase chain reaction (qRT-PCR), and senescence-associated ß-galactosidase (SA-ß-gal) staining. Filbertone was markedly observed to decrease not only the protein levels of p53 (p < 0.01) in senescence-induced skeletal muscle cells, but also the gene expression levels of p21 (p < 0.05), a direct target of p53. The expression of muscle-related genes, including myogenin and muscle RING-finger protein-1 (MuRF1), was found to be significantly enhanced in senescent muscle cells following treatment with filbertone (p < 0.05). In addition, the number of senescent skeletal muscle cells exhibiting ß-galactosidase activity was found to be markedly reduced in the presence of filbertone (p < 0.01). Collectively, these findings suggest that filbertone plays a pivotal role in the regulation of muscle aging.


Subject(s)
Cellular Senescence , Doxorubicin , Hydrogen Peroxide , Muscle Fibers, Skeletal , Muscle Proteins , Myogenin , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Animals , Cellular Senescence/drug effects , Mice , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Myogenin/metabolism , Myogenin/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Muscle Proteins/metabolism , Muscle Proteins/genetics , Doxorubicin/pharmacology , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Cell Line , Tumor Suppressor Protein p53/metabolism , beta-Galactosidase/metabolism
11.
Phytomedicine ; 134: 155989, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39217656

ABSTRACT

BACKGROUND: Doxorubicin (DOX) is a potent anticancer medication, but its significant cardiotoxicity poses a challenge in clinical practice. Galangin (Gal), a flavonoid compound with diverse pharmacological activities, has shown potential in exerting cardioprotective effects. However, the related molecular mechanism has not been fully elucidated. PURPOSE: Combined with bioinformatics and experimental verification methods to investigate Gal's potential role and underlying mechanisms in mitigating DOX-induced cardiotoxicity (DIC). METHODS: C57BL/6 mice received a single dose of DOX via intraperitoneal injection 4 days before the end of the gavage period with Gal. Myocardial injury was evaluated using echocardiography, myocardial injury biomarkers, Sirius Red and H&E staining. H9c2 cells were stimulated with DOX to mimic DIC in vitro. The potential therapeutic target of Gal was identified through network pharmacology, molecular docking and cellular thermal shift assay (CETSA), complemented by an in-depth exploration of the GSTP1/JNK signaling pathway using immunofluorescence. Subsequently, the GSTP1 inhibitor Ezatiostat (Eza) substantiated the signaling pathway. RESULTS: Gal administration considerably raised DOX-inhibited the left ventricular ejection fractions (LVEF), reduced levels of myocardial injury markers (c-TnI, c-TnT, CKMB, LDH, and AST), and alleviated DOX-induced myocardial histopathological injury and fibrosis in mice, thereby improving cardiac dysfunction. The ferroptosis induced by DOX was inhibited by Gal treatment. Gal remarkably ameliorated the DOX-induced lipid peroxidation, accumulation of iron and Ptgs2 expression both in H9c2 cells and cardiac tissue. Furthermore, Gal effectively rescued the DOX-inhibited crucial regulators of ferroptosis such as Gpx4, Nrf2, Fpn, and Slc7a11. The mechanistic investigations revealed that Glutathione S-transferase P1 (GSTP1) may be a potential target for Gal in attenuating DIC. Gal act on GSTP1 by stimulating its expression, thereby enhancing the interaction between GSTP1 and c-Jun N-terminal kinase (JNK), leading to the deactivation of JNK/c-Jun pathway. Furthermore, interference of GSTP1 with inhibitor Eza abrogated the cardioprotective and anti-ferroptotic effects of Gal, as evidenced by decreased cell viability, reduced expression of GSTP1 and Gpx4, elevated MDA levels, and promoted phosphorylation of JNK and c-Jun compared with Gal treatment. CONCLUSION: Gal could inhibit ferroptosis and protect against DIC through regulating the GSTP1/JNK pathway. Our research has identified a novel pathway through which Gal regulates DIC, providing valuable insights into the potential therapeutic efficacy of Gal in mitigating cardiotoxic effects.


Subject(s)
Cardiotoxicity , Doxorubicin , Ferroptosis , Flavonoids , Animals , Male , Mice , Rats , Cardiotoxicity/drug therapy , Cell Line , Doxorubicin/adverse effects , Ferroptosis/drug effects , Flavonoids/pharmacology , Glutathione S-Transferase pi/metabolism , MAP Kinase Signaling System/drug effects , Mice, Inbred C57BL , Molecular Docking Simulation , Myocardium , Signal Transduction/drug effects
12.
N Engl J Med ; 391(9): 789-799, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39231341

ABSTRACT

BACKGROUND: The addition of trabectedin to doxorubicin, followed by trabectedin maintenance, may have superior efficacy to doxorubicin alone as first-line treatment in patients with advanced leiomyosarcoma. METHODS: We conducted a phase 3 trial involving patients with metastatic or unresectable leiomyosarcoma who had not received chemotherapy previously. Patients were randomly assigned to receive either single-agent doxorubicin (six cycles) or doxorubicin plus trabectedin (six cycles), with continued trabectedin as maintenance therapy in patients in the doxorubicin-trabectedin group who did not have disease progression. Surgery to resect residual disease was allowed in each group after six cycles of therapy. Analyses of progression-free survival (primary end point) and overall survival (secondary end point) were adjusted for two stratification factors: tumor origin site (uterine vs. soft tissue) and disease stage (locally advanced vs. metastatic). The primary end-point results were reported previously. RESULTS: A total of 150 patients underwent randomization. At a median follow-up of 55 months (interquartile range, 49 to 63), a total of 107 patients had died (47 in the doxorubicin-trabectedin group and 60 in the doxorubicin group). The median overall survival was longer in the doxorubicin-trabectedin group (33 months; 95% confidence interval [CI], 26 to 48) than in the doxorubicin group (24 months; 95% CI, 19 to 31); the adjusted hazard ratio for death was 0.65 (95% CI, 0.44 to 0.95). In a finding consistent with earlier reports, progression-free survival was longer in the doxorubicin-trabectedin group (12 months; 95% CI, 10 to 16) than in the doxorubicin group (6 months; 95% CI, 4 to 7); the adjusted hazard ratio for progression or death was 0.37 (95% CI, 0.26 to 0.53). The incidence of adverse events and the percentage of patients with dose reductions were higher with doxorubicin plus trabectedin than with doxorubicin alone. CONCLUSIONS: Combination therapy with doxorubicin and trabectedin induction, followed by trabectedin maintenance, was associated with improved overall survival and progression-free survival, as compared with doxorubicin alone, among patients with metastatic or surgically unresectable uterine or soft-tissue leiomyosarcoma. (Funded by PharmaMar and others; LMS04 ClinicalTrials.gov number, NCT02997358.).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Doxorubicin , Leiomyosarcoma , Soft Tissue Neoplasms , Trabectedin , Uterine Neoplasms , Aged , Female , Humans , Male , Middle Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Kaplan-Meier Estimate , Leiomyosarcoma/drug therapy , Leiomyosarcoma/mortality , Leiomyosarcoma/pathology , Maintenance Chemotherapy , Progression-Free Survival , Soft Tissue Neoplasms/drug therapy , Soft Tissue Neoplasms/mortality , Soft Tissue Neoplasms/pathology , Survival Analysis , Trabectedin/administration & dosage , Trabectedin/adverse effects , Uterine Neoplasms/drug therapy , Uterine Neoplasms/mortality , Uterine Neoplasms/pathology , Neoplasm Staging
14.
J Nanobiotechnology ; 22(1): 533, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39223666

ABSTRACT

The poor prognosis of hepatocellular carcinoma (HCC) is still an urgent challenge to be solved worldwide. Hence, assembling drugs and targeted short peptides together to construct a novel medicine delivery strategy is crucial for targeted and synergy therapy of HCC. Herein, a high-efficiency nanomedicine delivery strategy has been constructed by combining graphdiyne oxide (GDYO) as a drug-loaded platform, specific peptide (SP94-PEG) as a spear to target HCC cells, sorafenib, doxorubicin-Fe2+ (DOX-Fe2+), and siRNA (SLC7A11-i) as weapons to exert a three-path synergistic attack against HCC cells. In this work, SP94-PEG and GDYO form nanosheets with HCC-targeting properties, the chemotherapeutic drug DOX linked to ferrous ions increases the free iron pool in HCC cells and synergizes with sorafenib to induce cell ferroptosis. As a key gene of ferroptosis, interference with the expression of SLC7A11 makes the ferroptosis effect in HCC cells easier, stronger, and more durable. Through gene interference, drug synergy, and short peptide targeting, the toxic side effects of chemotherapy drugs are reduced. The multifunctional nanomedicine GDYO@SP94/DOX-Fe2+/sorafenib/SLC7A11-i (MNMG) possesses the advantages of strong targeting, good stability, the ability to continuously induce tumor cell ferroptosis and has potential clinical application value, which is different from traditional drugs.


Subject(s)
Carcinoma, Hepatocellular , Doxorubicin , Ferroptosis , Liver Neoplasms , Nanomedicine , Peptides , Sorafenib , Ferroptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Humans , Liver Neoplasms/drug therapy , Doxorubicin/pharmacology , Doxorubicin/chemistry , Nanomedicine/methods , Sorafenib/pharmacology , Sorafenib/chemistry , Cell Line, Tumor , Animals , Peptides/chemistry , Peptides/pharmacology , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Drug Synergism , Amino Acid Transport System y+/metabolism , Mice, Nude , RNA, Small Interfering , Mice, Inbred BALB C , Drug Delivery Systems/methods
16.
J Nanobiotechnology ; 22(1): 531, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39218878

ABSTRACT

Ferroptosis, triggered by iron overload and excessive lipid peroxidation, plays a pivotal role in the progression of DOX-induced cardiomyopathy (DIC), and thus limits the use of doxorubicin (DOX) in clinic. Here, we further showed that cardiac ferroptosis induced by DOX in mice was attributed to up-regulation of Hmox1, as knockdown of Hmox1 effectively inhibited cardiomyocyte ferroptosis. To targeted delivery of siRNA into cardiomyocytes, siRNA-encapsulated exosomes were injected followed by ultrasound microbubble targeted destruction (UTMD) in the heart region. UTMD greatly facilitated exosome delivery into heart. Consistently, UTMD assisted exosomal delivery of siHomox1 nearly blocked the ferroptosis and the subsequent cardiotoxicity induced by doxorubicin. In summary, our findings reveal that the upregulation of HMOX1 induces ferroptosis in cardiomyocytes and UTMD-assisted exosomal delivery of siHmox1 can be used as a potential therapeutic strategy for DIC.


Subject(s)
Doxorubicin , Exosomes , Ferroptosis , Heme Oxygenase-1 , Microbubbles , Myocytes, Cardiac , RNA, Small Interfering , Ferroptosis/drug effects , Animals , Doxorubicin/pharmacology , Exosomes/metabolism , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Heme Oxygenase-1/metabolism , RNA, Small Interfering/pharmacology , Mice, Inbred C57BL , Male , Drug Delivery Systems , Cardiomyopathies/metabolism , Membrane Proteins
17.
J Med Econ ; 27(1): 1157-1167, 2024.
Article in English | MEDLINE | ID: mdl-39254695

ABSTRACT

AIMS: To understand treatment patterns, healthcare resource utilization (HCRU), and the economic burden of diffuse large B-cell lymphoma (DLBCL) in elderly adults in the US. MATERIALS AND METHODS: This retrospective database analysis utilized US Centers for Medicare and Medicaid Services Medicare fee-for-service administrative claims data from 2015 to 2020 to describe DLBCL patient characteristics, treatment patterns, HCRU, and costs among patients aged ≥66 years. Patients were indexed at DLBCL diagnosis and required to have continuous enrollment from 12 months pre-index until 3 months post-index. HCRU and costs (USD 2022) are reported as per-patient per-month (PPPM) estimates. RESULTS: A total of 11,893 patients received ≥1-line (L) therapy; 1,633 and 391 received ≥2 L and ≥3 L therapies, respectively. Median (Q1, Q3) age at 1 L, 2 L, and 3 L initiation, respectively, was 76 (71, 81), 77 (72, 82), and 77 (72, 82) years. The most common therapy was R-CHOP (70.9%) for 1 L and bendamustine ± rituximab for 2 L (18.7%) and 3 L (17.4%). CAR T was used by 14.8% of patients in 3 L. Overall, 39.6% (1 L), 42.1% (2 L), and 47.8% (3 L) of patients had all-cause hospitalizations. All-cause mean (median [Q1-Q3]) costs PPPM during each line were $22,060 ($20,121 [$16,676-$24,597]) in 1 L, $30,027 ($20,868 [$13,416-$31,016]) in 2 L, and $47,064 ($25,689 [$15,555-$44,149]) in 3 L, with increasing costs driven primarily by inpatient expenses. Total all-cause 3 L mean (median [Q1-Q3]) costs PPPM for patients with and without CAR T were $153,847 ($100,768 [$26,534-$253,630]) and $28,466 ($23,696 [$15,466-$39,107]), respectively. CONCLUSIONS: No clear standard of care exists in 3 L therapy for older adults with relapsed/refractory DLBCL. The economic burden of DLBCL intensifies with each progressing line of therapy, thus underscoring the need for additional therapeutic options.


Subject(s)
Insurance Claim Review , Lymphoma, Large B-Cell, Diffuse , Medicare , Humans , Lymphoma, Large B-Cell, Diffuse/economics , Lymphoma, Large B-Cell, Diffuse/drug therapy , United States , Retrospective Studies , Aged , Male , Female , Aged, 80 and over , Medicare/economics , Antineoplastic Combined Chemotherapy Protocols/economics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Patient Acceptance of Health Care/statistics & numerical data , Health Resources/economics , Health Resources/statistics & numerical data , Health Expenditures/statistics & numerical data , Age Factors , Doxorubicin/therapeutic use , Doxorubicin/economics , Rituximab/economics , Rituximab/therapeutic use
18.
Cancer Med ; 13(18): e70239, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39300969

ABSTRACT

BACKGROUND: Osteosarcoma (OS) survival rates and outcome have not improved in 50 years since the advent of modern chemotherapeutics. Thus, there is a critical need for an improved understanding of the tumor microenvironment to identify better therapies. Extracellular matrix (ECM) deposition and hypoxia are known to abrogate the efficacy of various chemical and cell-based therapeutics. Here, we aim to mechanistically investigate the combinatorial effects of hypoxia and matrix deposition with the use of OS spheroids. METHODS: We use two murine OS cell lines with differential metastatic potential to form spheroids. We form spheroids of two sizes, use ascorbate-2-phosphate supplementation to enhance ECM deposition, and study cell response under standard (21% O2) and physiologic (5% O2) oxygen tensions. Finally, we examine chemotherapeutic responses to doxorubicin treatment. RESULTS: ECM production and oxygen tension are key determinants of spheroid size through cell organization based on nutrient and oxygen distribution. Interestingly, highly metastatic OS is more susceptible to chemotherapeutics compared to less metastatic OS when matrix production increases. Together, these data suggest that dynamic interactions between ECM production and oxygen diffusion may result in distinct chemotherapeutic responses despite inherent tumor aggressiveness. CONCLUSION: This work establishes OS spheroids as a valuable tool for early OS tumor formation investigation and holds potential for novel therapeutic target and prognostic indicator discovery.


Subject(s)
Extracellular Matrix , Osteosarcoma , Oxygen , Spheroids, Cellular , Tumor Microenvironment , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Osteosarcoma/pathology , Spheroids, Cellular/drug effects , Extracellular Matrix/metabolism , Animals , Mice , Oxygen/metabolism , Cell Line, Tumor , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
19.
Biomater Sci ; 12(19): 5076-5090, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39219371

ABSTRACT

Metabolic disorders of cancer cells create opportunities for metabolic interventions aimed at selectively eliminating cancer cells. Nevertheless, achieving this goal is challenging due to cellular plasticity and metabolic heterogeneity of cancer cells. This study presents a dual-drug-loaded, macrophage membrane-coated polymeric nanovesicle designed to reprogram cancer metabolism with high specificity through integrated extracellular and intracellular interventions. This nanoformulation can target cancer cells and largely reduce their glucose intake, while the fate of intracellular glucose internalized otherwise is redirected at the specially introduced oxidation reaction instead of inherent cancer glycolysis. Meanwhile, it inhibits cellular citrate intake, further reinforcing metabolic intervention. Furthermore, the nanoformulation causes not only H2O2 production, but also NADPH down-regulation, intensifying redox damage to cancer cells. Consequently, this nanoformulation displays highly selective toxicity to cancer cells and minimal harm to normal cells mainly due to metabolic vulnerability of the former. Once administered into tumor-bearing mice, this nanoformulation is found to induce the transformation of pro-tumor tumor associated macrophages into the tumor-suppressive phenotype and completely inhibit tumor growth with favourable biosafety.


Subject(s)
Nanoparticles , Polymers , Animals , Mice , Humans , Polymers/chemistry , Polymers/pharmacology , Polymers/administration & dosage , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Glucose/metabolism , RAW 264.7 Cells , Doxorubicin/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Drug Carriers/chemistry , Cell Proliferation/drug effects , Metabolic Reprogramming
20.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(8): 1441-1449, 2024 Aug 20.
Article in Chinese | MEDLINE | ID: mdl-39276039

ABSTRACT

OBJECTIVE: To investigate the effect of Kaixinsan (KXS, a traditional Chinese medicine formula) for alleviating adriamycin-induced depression-like behaviors in mice bearing breast cancer xenografts and explore the pharmacological mechanism. METHODS: Forty female BALB/c mice were randomized equally into control group, model group, and low- and high-dose KXS treatment groups, and in the latter 3 groups, mouse models bearing orthotopic breast cancer 4T1 cell xenografts were established and treated with adriamycin along with saline or KXS via gavage. Depression-like behaviors of the mice were assessed using open field test and elevated plus-maze test, and the changes in serum levels of depression-related factors were examined. RNA-seq analysis and transmission electron microscopy were used and ferroptosis-related factors were detected to explore the mechanisms of adriamycin-induced depression and the therapeutic mechanism of KXS. The results were verified in SH-SY5Y cells using ferroptosis inhibitor Fer-1 as the positive control. RESULTS: KXS significantly alleviated depression-like behaviors and depression-related serological changes induced by adriamycin in the mouse models. RNA-seq results suggested that KXS alleviated chemotherapy-induced depression by regulating oxidative stress, lipid metabolism and iron ion binding in the prefrontal cortex. Pathological analysis and detection of ferroptosis-related factors showed that KXS significantly reduced ferroptosis in the prefrontal cortex of adriamycin-treated mice. In SH-SY5Y cells, both KXS-medicated serum and the ferroptosis inhibitor were capable of attenuating adriamycin-induced cell ferroptosis. CONCLUSION: KXS alleviates adriamycininduced depression-like behaviors in mice by reducing ferroptosis in the prefrontal cortex of breast cancer-bearing mice.


Subject(s)
Depression , Doxorubicin , Ferroptosis , Mice, Inbred BALB C , Prefrontal Cortex , Animals , Ferroptosis/drug effects , Mice , Depression/drug therapy , Depression/chemically induced , Doxorubicin/adverse effects , Female , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Cell Line, Tumor , Behavior, Animal/drug effects , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL