Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 6.931
1.
Int Ophthalmol ; 44(1): 251, 2024 Jun 22.
Article En | MEDLINE | ID: mdl-38907750

OBJECTIVE: To study the efficacy and side-effect profile of topical 5-Fluorouracil (5-FU) in the treatment of ocular surface squamous neoplasia (OSSN). METHODS: Retrospective study of 101 eyes of 100 patients treated with 5-FU with one week on and 3 weeks off regimen. RESULTS: Of the 100 patients (101 eyes), the mean age at diagnosis of OSSN was 49 (median, 52 years; range, 11-87 years). History of prior intervention was noted in 6 (6%) eyes. Tumor epicenter included bulbar conjunctiva (n = 54; 53%), limbus (n = 27; 27%), and cornea (n = 20;20%). Mean number of cycles of topical 5-FU administered was 3 (median, 3; range, 1-8). Complete tumor regression was achieved with topical 5-FU in 89 (88%) eyes with a mean number of 2 cycles (median, 2; range, 1-6) of 5-FU. The remaining 12 (12%) lesions underwent additional treatment including excisional biopsy (n = 7), extended enucleation (n = 3), and topical Interferon alpha 2b (n = 2) for complete tumor control. Over a mean follow-up period of 6 months (median, 5 months; range, 1-36 months) following treatment, tumor recurrence was noted in 2 (2%) patients, and side-effects were noted in 7 (7%) eyes including conjunctival hyperemia (n = 1), punctal stenosis (n = 1), sterile keratitis (n = 4), and limbal stem cell deficiency (n = 1). CONCLUSION: Topical 5-FU is an effective non-invasive therapy for OSSN with a minimal side-effect profile.


Antimetabolites, Antineoplastic , Carcinoma, Squamous Cell , Fluorouracil , Ophthalmic Solutions , Humans , Fluorouracil/administration & dosage , Retrospective Studies , Aged , Male , Middle Aged , Female , Adult , Aged, 80 and over , Adolescent , Antimetabolites, Antineoplastic/administration & dosage , Young Adult , Ophthalmic Solutions/administration & dosage , Child , Treatment Outcome , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/diagnosis , Administration, Topical , Eye Neoplasms/drug therapy , Eye Neoplasms/diagnosis , Conjunctival Neoplasms/drug therapy , Conjunctival Neoplasms/diagnosis , Conjunctival Neoplasms/pathology , Corneal Diseases/drug therapy , Corneal Diseases/diagnosis , Follow-Up Studies
2.
Arch Dermatol Res ; 316(7): 400, 2024 Jun 15.
Article En | MEDLINE | ID: mdl-38878177

BACKGROUND: Keratoacanthoma (KA) is a benign neoplasm that affects mainly photodamaged skin. It is locally destructive and may rarely spread. Surgery is not always suitable and usually disfiguring. Thus, non-operative modalities represent good alternatives. OBJECTIVE: To assess and compare the efficacy of intralesional methotrexate (MTX) and 5-flurouracil (5-FU) in the treatment of KA. PATIENTS AND METHODS: Randomized controlled trial included 20 patients with biopsy proven KA divided into 2 equal groups; group (A) received intralesional MTX, 25 mg/ml and group (B) received intralesional 5-FU, 50 mg/ml every 2 weeks till complete clearance or for a maximum 5 sessions. RESULTS: In the MTX group, complete clearance was observed in 7 patients (70%) compared to 8 patients (80%) in the 5- FU group with no statistically significant difference. However, the median number of injections needed to achieve complete response in the MTX group was 3 sessions versus only 2 sessions in the 5-FU group. LIMITATIONS: the small sample size due to the relatively low incidence of KAs in our population. CONCLUSION: Intralesional therapy is a good alternative to surgery in selected cases of KA. Both drugs showed comparable efficacy, but 5-FU may give faster results, hence increasing patient satisfaction and compliance.


Fluorouracil , Injections, Intralesional , Keratoacanthoma , Methotrexate , Humans , Methotrexate/administration & dosage , Methotrexate/therapeutic use , Fluorouracil/administration & dosage , Fluorouracil/therapeutic use , Keratoacanthoma/drug therapy , Keratoacanthoma/pathology , Female , Male , Middle Aged , Aged , Treatment Outcome , Adult , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/therapeutic use , Aged, 80 and over
3.
Article Ru | MEDLINE | ID: mdl-38881013

Leptomeningeal metastases are lesions of brain and/or spinal cord sheaths by tumor cells. They occur in 5% of patients with solid tumors, although autopsies reveal these lesions much more often (10-20% of cases). Leptomengeal metastases are an unfavorable prognostic factor. Despite the modern NCCN treatment standards, including intrathecal therapy (ITT), such patients receive only irradiation of the entire brain and/or spinal cord in most cases. OBJECTIVE: To evaluate the effectiveness of ITT in patients with leptomeningeal metastases in breast cancer. MATERIAL AND METHODS: Twenty-five patients with breast cancer and leptomeningeal metastases underwent intrathecal administration of methotrexate between 2016 and 2022. Intrathecal chemotherapy was administered through lumbar puncture. We performed an intensive course (intrathecal methotrexate 15 mg 2 times a week for 1 month (8 injections), then intrathecal methotrexate 15 mg 1 time a week (4 injections), and then 15 mg 1 time a month until progression or unacceptable toxicity). RESULTS: The median duration of ITT was 2.5 months. Complete neurological responses were observed in 3 out of 25 (12%) patients, partial neurological response - in 15 out of 25 (60%) patients, progression of neurological symptoms - in 7 (28%) patients. The number of complete cytological responses was observed in 6 out of 25 (24%) patients. The median overall survival after ITT was 6.7 months. CONCLUSION: Effectiveness of ITT is confirmed by higher quality of life (72% of patients), complete cytological responses (24%) and improvement in neuroimaging data. This is an important criterion for severe patients with limited treatment options. First-stage ITT before whole-brain irradiation is preferable, as this approach increases overall survival by 3 months. Undoubtedly, ITT is a treatment option that can be used in routine clinical practice for lesions of brain and spinal cord sheaths.


Breast Neoplasms , Injections, Spinal , Meningeal Neoplasms , Methotrexate , Humans , Female , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Middle Aged , Adult , Meningeal Neoplasms/secondary , Meningeal Neoplasms/drug therapy , Methotrexate/administration & dosage , Aged , Meningeal Carcinomatosis/drug therapy , Meningeal Carcinomatosis/secondary , Antimetabolites, Antineoplastic/administration & dosage
6.
Lancet Haematol ; 11(6): e406-e414, 2024 Jun.
Article En | MEDLINE | ID: mdl-38796193

BACKGROUND: Follicular helper T-cell lymphomas (TFHL) harbour frequent alterations in genes that regulate DNA methylation. Preliminary reports suggest that treatment with 5-azacitidine has clinical activity in patients with relapsed or refractory TFHL. We aimed to compare the oral form of azacitidine with investigator's choice standard therapy (ICT; ie, gemcitabine, bendamustine, or romidepsin) in patients with relapsed or refractory TFHL. METHODS: Patients older than 18 years with relapsed or refractory TFHL (angioimmunoblastic T-cell lymphoma, follicular lymphoma, or nodal T-cell lymphoma with phenotype, ie, positive with two or more markers among CD10, BCL6, CXCL13, PD1, or ICOS) based on the 2017 WHO classification of haematological neoplasms, with an Eastern Cooperative Oncology Group performance status score of 0-3, were recruited in university hospitals from five European countries and from Japan. Patients were randomly assigned 1:1 to treatment with either azacitidine given at a dose of 300 mg once a day (200 mg in Japanese patients) for 14 days in a 28-day cycle or gemcitabine, bendamustine, or romidepsin according to the investigator's choice. Random assignment was stratified by the number of previous lines of therapy and by the presence of previous or concomitant myeloid malignancy. The primary endpoint was investigator-assessed progression-free survival, presented in the intention-to-treat population. This Article is the final analysis of this trial, registered at ClinicalTrials.gov (Europe NCT03593018 and Japan NCT03703375). FINDINGS: 86 patients (median age 69 years [IQR 62-76], 50 patients were male, 36 were female) were enrolled between Nov 9, 2018, to Feb 22, 2021; 42 in the azacitidine group and 44 in the ICT group. With a median follow-up of 27·4 months (IQR 20·2-32·9), the median progression-free survival was 5·6 months (95% CI 2·7 -8·1) in the azacitidine group versus 2·8 months (1·9-4·8) in the ICT group (hazard ratio of 0·63 (95% CI 0·38-1·07); 1-sided p=0·042). Grade 3-4 adverse events were reported in 32 (76%) of 42 patients in the azacitidine group versus 42 (98%) of 43 patients in the ICT group. The most adverse grade 3 or worse adverse events were haematological (28 [67%] of 42 patients vs 40 [93%] of 43 patients), infection (8 [19%] and 14 [33%]), and gastrointestinal (5 [12%] vs 1 [2%] for azacitidine and ICT, respectively). There were two treatment-related deaths in the azacitidine group (one endocarditis and one candidiasis) and three in the ICT group (one heart failure, one COVID-19, and one cause unknown). INTERPRETATION: Although the pre-specified primary outcome of the trial was not met, the favourable safety profile suggests that azacitidine could add to the treatment options in these difficult to treat diseases especially in combination with other drugs. Trials with combination are in preparation in a platform trial. FUNDING: Bristol-Myers Squibb. TRANSLATION: For the French translation of the abstract see Supplementary Materials section.


Azacitidine , Humans , Male , Female , Aged , Middle Aged , Azacitidine/therapeutic use , Azacitidine/adverse effects , Azacitidine/administration & dosage , Administration, Oral , Bendamustine Hydrochloride/therapeutic use , Bendamustine Hydrochloride/administration & dosage , Bendamustine Hydrochloride/adverse effects , Gemcitabine , Lymphoma, Follicular/drug therapy , Lymphoma, Follicular/mortality , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Depsipeptides/therapeutic use , Depsipeptides/adverse effects , Depsipeptides/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antimetabolites, Antineoplastic/therapeutic use , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/administration & dosage , Aged, 80 and over
7.
Sci Rep ; 14(1): 11431, 2024 05 19.
Article En | MEDLINE | ID: mdl-38763930

Our current study reports the successful synthesis of thiolated chitosan-based nanoparticles for targeted drug delivery of 5-Fluorouracil. This process was achieved through the ionic gelation technique, aiming to improve the efficacy of the chemotherapeutic moiety by modifying the surface of the nanoparticles (NPs) with a ligand. We coated these NPs with hyaluronic acid (HA) to actively target the CD44 receptor, which is frequently overexpressed in various solid malignancies, including breast cancer. XRD, FTIR, SEM, and TEM were used for the physicochemical analysis of the NPs. These 5-Fluorouracil (5-FU) loaded NPs were evaluated on MDA-MB-231 (a triple-negative breast cell line) and MCF-10A (normal epithelial breast cells) to determine their in vitro efficacy. The developed 5-FU-loaded NPs exhibited a particle size within a favorable range (< 300 nm). The positive zeta potential of these nanoparticles facilitated their uptake by negatively charged cancer cells. Moreover, they demonstrated robust stability and achieved high encapsulation efficiency. These nanoparticles exhibited significant cytotoxicity compared to the crude drug (p < 0.05) and displayed a promising release pattern consistent with the basic diffusion model. These traits improve the pharmacokinetic profile, efficacy, and ability to precisely target these nanoparticles, offering a potentially successful anticancer treatment for breast cancer. However, additional in vivo assessments of these formulations are obligatory to confirm these findings.


Chitosan , Fluorouracil , Hyaluronan Receptors , Nanoparticles , Triple Negative Breast Neoplasms , Fluorouracil/administration & dosage , Fluorouracil/pharmacology , Fluorouracil/chemistry , Chitosan/chemistry , Humans , Hyaluronan Receptors/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Nanoparticles/chemistry , Cell Line, Tumor , Female , Drug Carriers/chemistry , Hyaluronic Acid/chemistry , Drug Delivery Systems , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacokinetics , Antimetabolites, Antineoplastic/chemistry , Cell Survival/drug effects , Particle Size
8.
Int J Pharm ; 658: 124206, 2024 Jun 10.
Article En | MEDLINE | ID: mdl-38734276

The constraints associated with current cancer therapies have inspired scientists to develop advanced, precise, and safe drug delivery methods. These delivery systems boost treatment effectiveness, minimize harm to healthy cells, and combat cancer recurrence. To design advanced drug delivery vehicle with these character, in the present manuscript, we have designed a self-healing and injectable hybrid hydrogel through synergistically interacting metal organic framework, CuBTC with the poly(vinyl alcohol) (PVA). This hybrid hydrogel acts as a localized drug delivery system and was used to encapsulate and release the anticancer drug 5-Fluorouracil selectively at the targeted site in response to the physiological pH. The hydrogel was formed through transforming the gaussian coil like matrix of PVA-CuBTC into a three-dimensional network of hydrogel upon the addition of crosslinker; borax. The biocompatible character of the hydrogel was confirmed through cell viability test. The biocompatible hybrid hydrogel then was used to encapsulate and studied for the pH responsive release behavior of the anti-cancer drug, 5-FU. The in vitro cytotoxicity of the drug-loaded hydrogel was evaluated against MCF-7 and HeLa cells. The study confirms that the hybrid hydrogel is effective for targeted and sustained release of anticancer drugs at cancer sites.


Breast Neoplasms , Cell Survival , Drug Delivery Systems , Drug Liberation , Fluorouracil , Hydrogels , Metal-Organic Frameworks , Polyvinyl Alcohol , Humans , Fluorouracil/administration & dosage , Fluorouracil/chemistry , Fluorouracil/pharmacology , Breast Neoplasms/drug therapy , MCF-7 Cells , HeLa Cells , Cell Survival/drug effects , Hydrogels/chemistry , Female , Metal-Organic Frameworks/chemistry , Polyvinyl Alcohol/chemistry , Drug Delivery Systems/methods , Hydrogen-Ion Concentration , Drug Carriers/chemistry , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology
9.
Medicine (Baltimore) ; 103(21): e38292, 2024 May 24.
Article En | MEDLINE | ID: mdl-38788030

This study aimed to investigate the prognostic relationship between relative dose intensity (RDI) of adjuvant S-1 chemotherapy and psoas muscle mass volume (PMV) in patients with resected pancreatic ductal adenocarcinoma. We enrolled 105 patients with histologically confirmed pancreatic ductal adenocarcinoma who had undergone pancreatectomy. Adjuvant S-1 chemotherapy was administered to 72 (68.6%) of the 105 patients and not to the remaining 33 patients. Patients who received adjuvant S-1 chemotherapy were stratified into high- and low-RDI groups by the cutoff value for RDI. Five-year overall survival (OS) and relapse-free survival (RFS) rates were significantly higher in the high- than in the low-RDI group. Similarly, both the 5-year OS and RFS rates were significantly greater among patients in the high-PMV group than among patients in the low-PMV group. The RDI was an independent prognostic factor in our study patients. Furthermore, patients who received adjuvant S-1 chemotherapy were stratified into 3 groups: those with both high RDI and high-PMV, Group A; those with either high RDI or high PMV (but not both), Group B; and those with both low RDI and low-PMV, group C. There were statistically significant differences in 5-year OS and RFS between 3 patient groups (5-year overall survival: P = .023, 5-year relapse-free survival: P = .001). The area under the curve for the combination of RDI and PMV (0.674) was greater than that for RDI alone (0.645). A sufficient dosage of adjuvant S-1 chemotherapy is important in improving survival of patients with resected pancreatic ductal adenocarcinoma. A combination of RDI and PMV may predict the prognosis of patients with resected pancreatic ductal adenocarcinoma more effective than RDI alone.


Carcinoma, Pancreatic Ductal , Drug Combinations , Oxonic Acid , Pancreatectomy , Pancreatic Neoplasms , Psoas Muscles , Tegafur , Humans , Male , Female , Oxonic Acid/administration & dosage , Oxonic Acid/therapeutic use , Tegafur/administration & dosage , Tegafur/therapeutic use , Retrospective Studies , Middle Aged , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/surgery , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/pathology , Psoas Muscles/pathology , Chemotherapy, Adjuvant/methods , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Aged , Pancreatectomy/methods , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/therapeutic use , Prognosis , Dose-Response Relationship, Drug , Adult
10.
Clin Ther ; 46(5): 429-432, 2024 May.
Article En | MEDLINE | ID: mdl-38763818

PURPOSE: The case of a 47-year-old female patient who underwent sigmoidectomy for metastatic colorectal cancer is reported. Treatment with capecitabine and 5-fluorouracil induced severe hypertriglyceridemia repeatedly. METHODS: Based on laboratory tests and clinical evaluations, treatment was suggested by specialists. FINDINGS: After treatment with capecitabine, the patient's triglycerides increased from 19.7 mmol/L to 42 mmol/L. It was proposed that the patient had multifactorial chylomicronemia syndrome triggered by secondary factors. Statins, fenofibrate, ezetimib, and metformin were added to the therapy. After metastases appeared, FOLFIRI (leucovorin calcium [folinic acid], 5-fluorouracil, and irinotecan hydrochloride) chemotherapy and biological treatment (cetuximab) followed and triglycerides increased to 55.3 mmol/L. IMPLICATIONS: Monitoring triglyceride levels before and during therapy is suggested.


Colorectal Neoplasms , Fluorouracil , Hypertriglyceridemia , Humans , Female , Middle Aged , Fluorouracil/adverse effects , Hypertriglyceridemia/chemically induced , Colorectal Neoplasms/drug therapy , Capecitabine/adverse effects , Capecitabine/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/administration & dosage , Triglycerides/blood , Leucovorin/therapeutic use , Leucovorin/adverse effects , Leucovorin/administration & dosage
11.
Investig Clin Urol ; 65(3): 248-255, 2024 May.
Article En | MEDLINE | ID: mdl-38714515

PURPOSE: This study investigated the efficacy of intravesical gemcitabine as an alternative to bacillus Calmette-Guérin (BCG) therapy. MATERIALS AND METHODS: Data were retrospectively collected across seven institutions from February 1999 to May 2023. Inclusion criteria included patients with intermediate- or high-risk non-muscle invasive bladder cancer (NMIBC) who underwent transurethral resection of bladder tumors (TURBT) and received at least four sessions of intravesical gemcitabine or BCG induction therapy. Patient characteristics, complete remission (CR), occurrence, and progression rates were compared. RESULTS: In total, 149 patients were included in this study (gemcitabine, 63; BCG, 86). No differences were apparent between the two groups in baseline characteristics, except for the follow-up period (gemcitabine, 9.2±5.9 months vs. BCG, 43.9±41.4 months, p<0.001). There were no consistent significant differences observed between the two groups in the 3-month (gemcitabine, 98.4% vs. BCG, 95.3%; p=0.848), 6-month (94.9% vs. 90.0%, respectively; p=0.793) and 1-year CR rates (84.2% vs. 83.3%, respectively; p=0.950). Also, there was no significant statistical difference in progression-free survival between the two groups (p=0.953). The occurrence rates of adverse events were similar between the groups (22.2% vs. 22.1%; p=0.989); however, the rate of Clavien-Dindo grade 2 or higher was significantly higher in the BCG group (1.6% vs. 16.3%, respectively; p<0.001). CONCLUSIONS: Intravesical gemcitabine demonstrated efficacy comparable to BCG therapy for the first year in patients with intermediate- and high-risk NMIBC. However, long-term follow-up studies are warranted.


Adjuvants, Immunologic , Antimetabolites, Antineoplastic , BCG Vaccine , Deoxycytidine , Gemcitabine , Neoplasm Recurrence, Local , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/surgery , Urinary Bladder Neoplasms/therapy , Deoxycytidine/analogs & derivatives , Deoxycytidine/administration & dosage , Retrospective Studies , BCG Vaccine/administration & dosage , BCG Vaccine/therapeutic use , Male , Female , Administration, Intravesical , Aged , Antimetabolites, Antineoplastic/administration & dosage , Middle Aged , Adjuvants, Immunologic/administration & dosage , Cystectomy/methods , Risk Assessment , Urethra
12.
Eur J Pharm Biopharm ; 200: 114326, 2024 Jul.
Article En | MEDLINE | ID: mdl-38759897

Glioblastoma (GBM) is a highly deadly brain tumor that does not respond satisfactorily to conventional treatment. The non-alkylating agent gemcitabine (GEM) has been proposed for treating GBM. It can overcome MGMT protein-mediated resistance, a major limitation of conventional therapy with the alkylating agent temozolomide (TMZ). However, GEM's high systemic toxicity and poor permeability across the blood-brain barrier (BBB) pose significant challenges for its delivery to the brain. Thus, mucoadhesive poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) coated with chitosan (CH), suitable for intranasal GEM delivery, were proposed in this work. A central composite design (CCD) was implemented for NPs optimization, and NPs with appropriate characteristics for intranasal administration were obtained. in vitro studies revealed that the NPs possess excellent mucoadhesive properties and the ability to selectively release GEM in the simulated tumor tissue environment. in vitro studies using two human GBM cell lines (U215 and T98G) revealed the NPs' ability to promote GEM's antiproliferative activity to sensitize cells to the effect of TMZ. The findings of this work demonstrate that the developed CH-GEM-NPs are suitable delivery systems for GEM, both as a single therapy and as a chemosensitizer to the GBM gold standard therapy.


Brain Neoplasms , Chitosan , Deoxycytidine , Drug Repositioning , Gemcitabine , Glioblastoma , Nanoparticles , Polylactic Acid-Polyglycolic Acid Copolymer , Glioblastoma/drug therapy , Glioblastoma/pathology , Deoxycytidine/analogs & derivatives , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Deoxycytidine/chemistry , Humans , Chitosan/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Nanoparticles/chemistry , Cell Line, Tumor , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Drug Repositioning/methods , Temozolomide/administration & dosage , Temozolomide/pharmacology , Temozolomide/pharmacokinetics , Administration, Intranasal , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacokinetics , Drug Carriers/chemistry , Blood-Brain Barrier/metabolism , Drug Liberation
13.
Eur J Pharm Biopharm ; 200: 114328, 2024 Jul.
Article En | MEDLINE | ID: mdl-38763329

Aggressive colon cancer treatment poses significant challenges. This study investigates the potential of innovative carbohydrate-based nanoparticles for targeted Capecitabine (CTB) delivery. CTB nanoparticles were synthesized by conjugating CTB with potato starch and chitosan using ultrasonication, hydrolysis, and ionotropic gelation. Characterization included drug loading, rheology, Surface-Enhanced Raman Spectroscopy (SERS), Fourier-Transform Infrared Spectroscopy (FTIR), Differential Scanning Calorimetry (DSC), X-ray Diffraction (XRD), and Thermogravimetric Analysis (TGA). In vitro and in vivo antitumor activity was evaluated using HT-29 cells and N, N-dimethylhydrazine-induced Balb/c mice, respectively. Cellular assays assessed angiogenesis, migration, proliferation, and apoptosis. Nanoparticles exhibited a mean size of 245 nm, positive zeta potential (+30 mV), high loading efficacy (76 %), and sustained drug release (92 % over 100 h). CTB-loaded nanoparticles displayed superior colon histology, reduced tumour scores, and inhibited VEGD and CD31 expression compared to free CTB. Cellular assays confirmed significant antitumor effects, including reduced tube formation, migration, and proliferation, and increased apoptosis. This study demonstrates the promise of CTB-loaded potato starch-chitosan nanoparticles for aggressive colon cancer treatment. These findings highlight the potential of these nanoparticles for further evaluation in diverse cancer models.


Capecitabine , Chitosan , Colonic Neoplasms , Mice, Inbred BALB C , Nanoparticles , Solanum tuberosum , Starch , Animals , Chitosan/chemistry , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Starch/chemistry , Solanum tuberosum/chemistry , Capecitabine/administration & dosage , Capecitabine/pharmacology , Humans , Mice , Nanoparticles/chemistry , HT29 Cells , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/chemistry , Drug Liberation , Drug Carriers/chemistry , Apoptosis/drug effects , Drug Delivery Systems/methods , Cell Proliferation/drug effects , Male
15.
Mol Pharm ; 21(6): 2699-2712, 2024 Jun 03.
Article En | MEDLINE | ID: mdl-38747900

This study aims to encapsulate gemcitabine (GEM) using a phospholipid complex (PLC) in lipid nanoparticles (NPs) to achieve several desirable outcomes, including high drug loading, uniform particle size, improved therapeutic efficacy, and reduced toxicities. The successful preparation of GEM-loaded lipid NPs (GEM-NPs) was accomplished using the emulsification-solidification method, following optimization through Box-Behnken design. The size of the GEM-NP was 138.5 ± 6.7 nm, with a low polydispersity index of 0.282 ± 0.078, as measured by a zetasizer and confirmed by transmission electron and atomic force microscopy. GEM-NPs demonstrated sustained release behavior, surpassing the performance of the free GEM and phospholipid complex. Moreover, GEM-NPs exhibited enhanced cytotoxicity, apoptosis, and cell uptake in Panc-2 and Mia PaCa cells compared to the free GEM. The in vivo pharmacokinetics revealed approximately 4-fold higher bioavailability of GEM-NPs in comparison with free GEM. Additionally, the pharmacodynamic evaluation conducted in a DMBA-induced pancreatic cancer model, involving histological examination, serum IL-6 level estimation, and expression of cleaved caspase-3, showed the potential of GEM-NPs in the management of pancreatic cancer. Consequently, the lipid NP-based approach developed in our investigation demonstrates high stability and uniformity and holds promise for enhancing the therapeutic outcomes of GEM.


Deoxycytidine , Gemcitabine , Nanoparticles , Pancreatic Neoplasms , Phospholipids , Deoxycytidine/analogs & derivatives , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Deoxycytidine/pharmacokinetics , Deoxycytidine/administration & dosage , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Nanoparticles/chemistry , Animals , Humans , Cell Line, Tumor , Phospholipids/chemistry , Mice , Particle Size , Apoptosis/drug effects , Drug Carriers/chemistry , Lipids/chemistry , Drug Liberation , Male , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacokinetics , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacology , Drug Stability , Rats , Liposomes
16.
Expert Opin Drug Metab Toxicol ; 20(5): 399-406, 2024 May.
Article En | MEDLINE | ID: mdl-38706380

BACKGROUND: Methotrexate (MTX) is partially metabolized by aldehyde oxidase (AOX) in the liver and its clinical impact remains unclear. In this study, we aimed to demonstrate how AOX contributes to MTX-induced hepatotoxicity in vitro and clarify the relationship between concomitant AOX inhibitor use and MTX-associated liver injury development using the U.S. Food and Drug Administration Adverse Event Reporting System (FAERS). METHODS: We assessed intracellular MTX accumulation and cytotoxicity using HepG2 cells. We used the FAERS database to detect reporting odds ratio (ROR)-based MTX-related hepatotoxicity event signals. RESULTS: AOX inhibition by AOX inhibitor raloxifene and siRNA increased the MTX accumulation in HepG2 cells and enhanced the MTX-induced cell viability reduction. In the FAERS analysis, the ROR for MTX-related hepatotoxicity increased with non-overlap of 95% confidence interval when co-administered with drugs with higher Imax, u (maximum unbound plasma concentration)/IC50 (half-maximal inhibitory concentration for inhibition of AOX) calculated based on reported pharmacokinetic data. CONCLUSION: AOX inhibition contributed to MTX accumulation in the liver, resulting in increased hepatotoxicity. Our study raises concerns regarding MTX-related hepatotoxicity when co-administered with drugs that possibly inhibit AOX activity at clinical concentrations.


Adverse Drug Reaction Reporting Systems , Aldehyde Oxidase , Chemical and Drug Induced Liver Injury , Methotrexate , Methotrexate/adverse effects , Methotrexate/administration & dosage , Humans , Aldehyde Oxidase/metabolism , Chemical and Drug Induced Liver Injury/etiology , Hep G2 Cells , Cell Survival/drug effects , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/administration & dosage , United States , United States Food and Drug Administration , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacology , Inhibitory Concentration 50
17.
BMC Immunol ; 25(1): 28, 2024 May 06.
Article En | MEDLINE | ID: mdl-38710996

INTRODUCTION: Metronomic capecitabine used as an adjuvant therapy improves survival in patients with locoregionally advanced nasopharyngeal carcinoma (LA-NPC). This therapeutic approach may also contribute to improving immune function, consequently enhancing overall therapeutic efficacy. AIM: We aimed to evaluate the effect of metronomic capecitabine as adjuvant therapy on immune function and survival in cases of LA-NPC. SUBJECTS AND METHODS: 28 patients with LA-NPC were enrolled in the study and equally assigned to two groups of 14 each: experimental and control group. The experimental group received induction chemotherapy + concurrent chemotherapy + adjuvant chemotherapy as well as oral capecitabine at a dose of 650 mg/m² of body surface area twice daily for 1 year, with the option to discontinue in case of intolerance. The control group did not receive additional chemotherapy or targeted drugs after the induction chemotherapy + concurrent chemoradiotherapy; however, they were followed up regularly. Changes in immune function and survival were compared between the two groups. RESULTS: The median follow-up time was 43.5 months. One year after adjuvant chemotherapy, the experimental group showed higher levels of CD8 + cells, CD28 + CD8 + cells, and activated CD8 + cells compared to the control group (P < 0.05). The CD4/CD8 ratio and proportion of monocyte-derived dendritic cells were also higher in the experimental group than in the control group, but the difference was not statistically significant (P ≥ 0.05). Comparisons of 3-year overall survival, local-regional recurrence-free survival, progression-free survival, and distant metastasis-free survival between the two groups showed percentages of 92.9% vs. 78.6%, 92.9% vs. 92.9%, 78.6% vs. 71.4%, and 85.7% vs. 0.78 0.6% respectively, but these differences were not significant (P > 0 0.05 ). CONCLUSION: Metronomic capecitabine chemotherapy was observed to induce an immunomodulatory effect in LA-NPC. TRIAL REGISTRATION: NCT02958111, date of registration 04-11-2016.


Administration, Metronomic , Capecitabine , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Humans , Capecitabine/administration & dosage , Capecitabine/therapeutic use , Male , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Carcinoma/immunology , Nasopharyngeal Carcinoma/mortality , Female , Middle Aged , Adult , Chemotherapy, Adjuvant/methods , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Aged , Neoplasm Staging , Treatment Outcome , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/therapeutic use , Follow-Up Studies
18.
Arch Dermatol Res ; 316(6): 220, 2024 May 24.
Article En | MEDLINE | ID: mdl-38787403

Cutaneous squamous cell carcinoma in-situ (SCCis) is an intraepithelial tumor with a good prognosis. Standard treatment includes both surgical and non-surgical interventions. We determined the clearance rate for SCCis and residual SCCis identified on frozen section during Mohs micrographic surgery (MMS) after treatment with topical fluorouracil 5% cream (5-FU). All MMS cases were initiated for biopsy-proven invasive squamous cell carcinoma (SCC). A retrospective chart review was conducted from January 2017-February 2024 at Columbia University Irving Medical Center (CUIMC) to identify patients with SCCis who were treated with topical 5-FU as primary therapy or adjuvant therapy (AT) for residual SCCis post-MMS for invasive SCC. 41 patients were included (80% males, 70.1 ± 11.8 years). The average follow-up time for the primary therapy group was 25.4 ± 12.8 months, and for the post-MMS AT group 22.5 ± 11.1 months. In the group treated with topical 5-FU as primary therapy (n = 28), 27 patients (96.43%, 95% confidence interval: 81.65-99.91%) achieved complete clearance. One patient had recurrence at 8 months post-treatment. Of the patients in the post-MMS adjuvant treatment group (n = 13), 12 (92.3% clearance, 95% confidence interval 63.97-99.81%) achieved complete clearance. One patient had recurrence at 8 months post-treatment. This study found that topical 5-FU cream is effective as both primary therapy for SCCis and as adjuvant therapy for residual SCCis following MMS of invasive SCC.


Carcinoma, Squamous Cell , Fluorouracil , Skin Neoplasms , Humans , Fluorouracil/administration & dosage , Fluorouracil/therapeutic use , Male , Female , Aged , Retrospective Studies , Middle Aged , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Skin Neoplasms/diagnosis , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/diagnosis , Chemotherapy, Adjuvant/methods , Aged, 80 and over , Treatment Outcome , Mohs Surgery , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/therapeutic use , Carcinoma in Situ/drug therapy , Carcinoma in Situ/pathology , Administration, Topical , Follow-Up Studies , Neoplasm Recurrence, Local/prevention & control , Administration, Cutaneous
19.
Sci Rep ; 14(1): 9695, 2024 04 27.
Article En | MEDLINE | ID: mdl-38678107

High-dose methotrexate (HD-MTX) is a widely used chemotherapy regimen for hematologic malignancies such as lymphomas and acute lymphoblastic leukemia, but its use can lead to adverse effects, including acute kidney injury (AKI), impaired liver function, and mucositis, causing extended hospital stays and delayed subsequent chemotherapy. Our study aimed to investigate the predictive factors for renal toxicities associated with HD-MTX in Thai patients undergoing treatment for hematologic malignancies. We enrolled 80 patients who underwent MTX-containing regimens, analyzing 132 chemotherapy cycles. The most common disease was primary central nervous system lymphoma (33%). Genetic polymorphisms were examined using the MassARRAY® system, identifying 42 polymorphisms in 25 genes. Serum creatinine and MTX levels were measured 24 and 48 h after MTX administration. For the primary outcome, we found that the allele A of MTRR rs1801394 was significantly related to renal toxicity (odds ratio 2.084 (1.001-4.301), p-value 0.047). Patients who exceeded the MTX threshold levels at 24 h after the dose had a significantly higher risk of renal toxicity (OR (95%CI) = 6.818 (2.350-19.782), p < 0.001). Multivariate logistic regression analysis with a generalized estimated equation revealed hypertension and age as independent predictors of increased MTX levels at 24 h after the given dose.


Hematologic Neoplasms , Methotrexate , Humans , Male , Methotrexate/adverse effects , Methotrexate/administration & dosage , Female , Middle Aged , Thailand/epidemiology , Aged , Adult , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/genetics , Acute Kidney Injury/chemically induced , Acute Kidney Injury/genetics , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/administration & dosage , Polymorphism, Single Nucleotide , Young Adult , Southeast Asian People
...