Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 402
Filter
1.
Ther Apher Dial ; 26(1): 185-190, 2022 Feb.
Article in English | MEDLINE | ID: mdl-33891365

ABSTRACT

Central venous stenosis (CVS) is usually a late-diagnosed clinical entity that is common in hemodialysis patients. It causes various problems ranging from hemodialysis difficulty to loss of the arterio-venous (A-V) fistula. In the present study, we aimed to determine the effect of drug eluting balloon while excluding the influence of other variable factors by evaluating the same individuals with plain and paclitaxel-eluting balloons. This research was a prospective study of 18 symptomatic hemodialysis patients (age 50.9 ± 14.0 years, range 32-72 years; 11 male, 7 female) with CVS who underwent treatment by plain balloon angioplasty (PBA) and paclitaxel-eluting balloon angioplasty (PEBA) in our hospital from January 2016 to June 2017. First, third and sixth month central vein patency rates were compared. The median patency rates of central veins were 109.0 (range: 10-324) days after PBA and 238.5 (range: 157-501) days after PEBA (p < 0.001). There was no statistically significant difference between PBA and PEBA angioplasty in one-month patency (p Ëƒ 0.05). By contrast, a statistically significant difference was found between 3- and 6-month patency rates (p = 0.031 and p Ë‚ 0.001, respectively). Kaplan-Meier analysis revealed that the primary cumulative patency rate of PEBA was significantly longer than that of PBA (p ˂ 0.001). In this prospective study, PEBA patency is superior to PBA patency in the treatment of CVS in dialysis patients.


Subject(s)
Angioplasty, Balloon/instrumentation , Angioplasty, Balloon/methods , Graft Occlusion, Vascular/drug therapy , Paclitaxel/administration & dosage , Renal Dialysis/adverse effects , Vascular Patency/drug effects , Adult , Aged , Constriction, Pathologic/complications , Constriction, Pathologic/drug therapy , Female , Graft Occlusion, Vascular/etiology , Humans , Male , Middle Aged , Prospective Studies , Renal Dialysis/instrumentation , Treatment Outcome , Tubulin Modulators/administration & dosage
2.
Eur Rev Med Pharmacol Sci ; 25(22): 7151-7161, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34859881

ABSTRACT

OBJECTIVE: To assess the effectiveness of colchicine, compared with standard of care, for reducing mortality, admission to intensive care, and use of mechanical ventilation. MATERIALS AND METHODS: We performed a systematic review, meta-analysis, and sequential trial analysis. The terms (SARS-CoV-2 OR COVID-19 OR coronavirus) AND (colchicine) were searched in MEDLINE, Scopus, Embase, Cochrane Central Register of Controlled Trials, and preprint repositories (February 2020 to April 2021, extended to June 2021). Risk of bias for randomised controlled trials and observational studies were assessed using the tools RoB 2.0 and ROBINS-I, respectively. We performed subgroup analyses based on study design and sensitivity analyses based on time of colchicine administration. RESULTS: We included six observational studies (1329 patients) and five clinical trials (16,048 patients). All studies but one were conducted in the hospital setting. Colchicine treatment was not associated with a significant decrease in mortality (RR 0.93, 95% CI 0.87 to 1; p=0.06, I2=72%) with a significant subgroup effect (p<0.001) depending on the design of the studies. The drug was effective in observational studies (RR 0.57, 95% CI 0.46 to 0.70, p<0.001, I2=50%) but not in clinical trials (RR 0.99, 95% CI 0.92 to 1.07, p=0.89, I2=21%). The effect of colchicine on intensive care admissions and the need for mechanical ventilation could not be confirmed. Trial sequential boundaries for cumulative meta-analyses of randomised controlled trials suggested no significant effect on mortality (p=0.182) beyond the optimal information size (13,107 patients). CONCLUSIONS: Our results suggest that colchicine treatment has no effect on mortality in hospitalised patients with SARS-CoV-2 infection, and that no further confirmatory clinical trials are needed owing to futility.


Subject(s)
COVID-19 Drug Treatment , COVID-19/mortality , Colchicine/therapeutic use , Tubulin Modulators/therapeutic use , Adult , COVID-19/diagnosis , COVID-19/virology , Case-Control Studies , Clinical Trials as Topic , Colchicine/administration & dosage , Critical Care/statistics & numerical data , Hospitalization/statistics & numerical data , Humans , Middle Aged , Mortality/trends , Observational Studies as Topic , Outcome Assessment, Health Care , Randomized Controlled Trials as Topic , Respiration, Artificial/statistics & numerical data , SARS-CoV-2/drug effects , SARS-CoV-2/genetics , Sensitivity and Specificity , Treatment Outcome , Tubulin Modulators/administration & dosage
3.
Sci Rep ; 11(1): 21722, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34741076

ABSTRACT

Spinal cord regeneration is limited due to various obstacles and complex pathophysiological events after injury. Combination therapy is one approach that recently garnered attention for spinal cord injury (SCI) recovery. A composite of three-dimensional (3D) collagen hydrogel containing epothilone B (EpoB)-loaded polycaprolactone (PCL) microspheres (2.5 ng/mg, 10 ng/mg, and 40 ng/mg EpoB/PCL) were fabricated and optimized to improve motor neuron (MN) differentiation efficacy of human endometrial stem cells (hEnSCs). The microspheres were characterized using liquid chromatography-mass/mass spectrometry (LC-mas/mas) to assess the drug release and scanning electron microscope (SEM) for morphological assessment. hEnSCs were isolated, then characterized by flow cytometry, and seeded on the optimized 3D composite. Based on cell morphology and proliferation, cross-linked collagen hydrogels with and without 2.5 ng/mg EpoB loaded PCL microspheres were selected as the optimized formulations to compare the effect of EpoB release on MN differentiation. After differentiation, the expression of MN markers was estimated by real-time PCR and immunofluorescence (IF). The collagen hydrogel containing the EpoB group had the highest HB9 and ISL-1 expression and the longest neurite elongation. Providing a 3D permissive environment with EpoB, significantly improves MN-like cell differentiation and maturation of hEnSCs and is a promising approach to replace lost neurons after SCI.


Subject(s)
Adult Stem Cells/drug effects , Cell Differentiation/drug effects , Epothilones/administration & dosage , Motor Neurons/cytology , Tubulin Modulators/administration & dosage , Adult Stem Cells/ultrastructure , Cell Culture Techniques, Three Dimensional , Collagen/chemistry , Collagen/pharmacology , Endometrium/cytology , Female , Hedgehog Proteins/administration & dosage , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Microspheres , Polyesters , Primary Cell Culture , Tretinoin/administration & dosage
4.
Drug Des Devel Ther ; 15: 4195-4211, 2021.
Article in English | MEDLINE | ID: mdl-34675483

ABSTRACT

BACKGROUND: Fenugreek, also known as Trigonella foenum-graecum L, is a natural plant that belongs to the Fabaceae family and has been known as a promising source of bioactive compounds. It has been widely used as traditional medicine since it has shown to lower blood glucose, manage cholesterol levels and further aid in the prevention and treatment of cancer. Herein, we aim to evaluate the anticancer activity of methanolic fenugreek seed extract against several cancer cell lines. METHODS: We sought to investigate the phytochemical classes present in multiple fenugreek seeds extracts using HPLC-DAD followed by LC/MS, predict and investigate anticancer activity using PASS online webserver, the CellTiter-Glo assay, evaluate ADME properties, and perform molecular docking for all bioactive compounds via Maestro software. RESULTS: Multiple extracts exhibited distinct phytochemical classes that demonstrated different biological activities. Fenugreek methanolic extract contains flavonoid chemical class, which showed the highest anticancer activity against the HCT8 cell line of colorectal cancer (IC50 of 8.83 µg/mL), followed by KAIMRC1 breast cancer cell line (IC50 of 35.06 µg/mL), HL60 leukemia cell line (37.80 µg/mL), MDA-MB-231 breast cancer cell line (38.51 µg/mL), and lastly, HCT116 colorectal cancer cell line with IC50 of 56.03 µg/mL. In contrast, the chloroform extract was inactive. The molecular docking study for all the bioactive compounds suggested that flavonoids F6 (-9.713 and -12.132), F7 (-10.166 and -12.411), and F11 (-10.084 and -13.516) possess the highest docking scores through SP and XP scores, respectively. CONCLUSION: The obtained results confirm that the bioactive compounds present in fenugreek seeds exhibit anticancer activity against several cancer cells that can mediate via tubulin polymerization inhibition. Although our study has evaluated the anticancer potential of Trigonella foenum-graecum as a promising natural source for new anticancer agents, fenugreek biological activity needs further research and investigations on their mechanism of action and toxicity profile.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Neoplasms/drug therapy , Plant Extracts/pharmacology , Tubulin Modulators/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Cell Line, Tumor , Chromatography, High Pressure Liquid , Chromatography, Liquid , Humans , Inhibitory Concentration 50 , Mass Spectrometry , Molecular Docking Simulation , Neoplasms/pathology , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Trigonella/chemistry , Tubulin/drug effects , Tubulin/metabolism , Tubulin Modulators/administration & dosage , Tubulin Modulators/chemistry
5.
Molecules ; 26(19)2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34641324

ABSTRACT

Microtubule targeting agents (MTAs) that interfere with the dynamic state of the mitotic spindle are well-known and effective chemotherapeutic agents. These agents interrupt the microtubule network via polymerization or depolymerization, halting the cell cycle progression and leading to apoptosis. We report two novel pyrrole-based carboxamides (CAs) (CA-61 and -84) as the compounds exhibiting potent anti-cancer properties against a broad spectrum of epithelial cancer cell lines, including breast, lung, and prostate cancer. The anti-cancer activity of CAs is due to their ability to interfere with the microtubules network and inhibit tubulin polymerization. Molecular docking demonstrated an efficient binding between these ligands and the colchicine-binding site on the tubulin. CA-61 formed two hydrogen bond interactions with THR 179 (B) and THR 353 (B), whereas two hydrogen bonds with LYS 254 (B) and 1 with ASN 101 (A) were identified for CA-84. The binding energy for CA-84 and CA-61 was -9.910 kcal/mol and -9.390 kcal/mol. A tubulin polymerization assay revealed a strong inhibition of tubulin polymerization induced by CA-61 and -84. The immunofluorescence data revealed the disruption of the tubulin assembly in CA-treated cancer cells. As an outcome of the tubulin inhibition, these compounds halted the cell cycle progression in the G2/M phase, leading to the accumulation of the mitotic cells, and further induced apoptosis. Lastly, the in vivo study indicated that CAs significantly inhibited the HCC1806 breast cancer xenograft tumor growth in a nude mouse model. Collectively, we identified the novel CAs as potent MTAs, inhibiting tubulin polymerization via binding to the colchicine-binding site, disrupting the microtubule network, and exhibiting potent pro-apoptotic activities against the epithelial cancer cell lines both in vitro and in vivo.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Colchicine/metabolism , Pyrroles/administration & dosage , Tubulin Modulators/administration & dosage , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Female , Mice , Mice, Nude , Molecular Conformation , Molecular Docking Simulation , Molecular Structure , Pyrroles/chemical synthesis , Pyrroles/chemistry , Pyrroles/pharmacology , Structure-Activity Relationship , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Xenograft Model Antitumor Assays
6.
PLoS One ; 16(10): e0256783, 2021.
Article in English | MEDLINE | ID: mdl-34634057

ABSTRACT

BACKGROUND/OBJECTIVES: Drug-coated balloon therapy for diseased superficial femoral arteries remains controversial. Despite its clinical relevance, only a few computational studies based on simplistic two-dimensional models have been proposed to investigate this endovascular therapy to date. This work addresses the aforementioned limitation by analyzing the drug transport and kinetics occurring during drug-coated balloon deployment in a three-dimensional geometry. METHODS: An idealized three-dimensional model of a superficial femoral artery presenting with a calcific plaque and treated with a drug-coated balloon was created to perform transient mass transport simulations. To account for the transport of drug (i.e. paclitaxel) released by the device, a diffusion-reaction equation was implemented by describing the drug bound to specific intracellular receptors through a non-linear, reversible reaction. The following features concerning procedural aspects, pathologies and modelling assumptions were investigated: (i) balloon application time (60-180 seconds); (ii) vessel wall composition (healthy vs. calcified wall); (iii) sequential balloon application; and (iv) drug wash-out by the blood stream vs. coating retention, modeled as exponential decay. RESULTS: The balloon inflation time impacted both the free and specifically-bound drug concentrations in the vessel wall. The vessel wall composition highly affected the drug concentrations. In particular, the specifically-bound drug concentration was four orders of magnitude lower in the calcific compared with healthy vessel wall portions, primarily as a result of reduced drug diffusion. The sequential application of two drug-coated balloons led to modest differences (~15%) in drug concentration immediately after inflation, which became negligible within 10 minutes. The retention of the balloon coating increased the drug concentration in the vessel wall fourfold. CONCLUSIONS: The overall findings suggest that paclitaxel kinetics may be affected not only by the geometrical and compositional features of the vessel treated with the drug-coated balloon, but also by balloon design characteristics and procedural aspects that should be carefully considered.


Subject(s)
Angioplasty, Balloon/instrumentation , Drug Delivery Systems/instrumentation , Femoral Artery/surgery , Vascular Calcification/therapy , Coated Materials, Biocompatible , Drug Liberation , Equipment Design , Femoral Artery/drug effects , Humans , Models, Biological , Paclitaxel/administration & dosage , Paclitaxel/pharmacokinetics , Tubulin Modulators/administration & dosage , Tubulin Modulators/pharmacokinetics , Vascular Calcification/surgery
8.
J Pharm Pharm Sci ; 24: 344-362, 2021.
Article in English | MEDLINE | ID: mdl-34224665

ABSTRACT

PURPOSE: The ultimate goal of this study is to develop a novel delivery system for a new potent cytotoxic compound, CCI-001, with anti-b tubulin activity, so that the drug can be effectively administered and at the same time its harmful side effects can be reduced. METHODS: In the current study, CCI-001 was loaded into serum albumin (SA), using a modified desolvation method, generating CCI-001-SA nanoparticles. Both bovine and human SA were used for the encapsulation of this drug candidate. Optimum conditions for drug loading were achieved when already formed and crosslinked albumin nanoparticles were incubated overnight at 37°C with CCI-001 solutions. The CCI-001-loaded albumin nanoparticles were assessed for average particle diameter and polydispersity, zeta potential, drug loading, in vitro release, morphology and cell toxicity against SW620 and HCT116 colorectal cancer cells. RESULTS: The spherical nanoparticles obtained were negatively charged (~ -30 mV) and had an average diameter of ~ 130 nm, with a narrow size distribution. The in vitro release of CCI-001 from the albumin nanoparticles showed a sustained release pattern over 24 hours without any initial burst release, compared to the fast release of the free drug under experimental conditions. No difference between the SA from the two species in terms of CCI-001 loading was observed. However, a significant difference was observed between the release profiles of CCI-001 from drug-loaded HSA and drug-loaded BSA nanoparticles with HSA nanoparticles showing slower drug release (mean release time, MRT, values of 5.14 ± 0.33 h and 6.88 ± 0.15 h for BSA-NPs and HSA-NPs, respectively, P < 0.01). Cellular toxicity studies showed higher cytotoxicity for CCI-001-SA compared to the free drug (IC50s of 0.62 ± 0.31 nM vs 2.06 ± 0.29 nM in SW620 cells and 0.9 ± 0.1 nM vs 4.2 ± 0.2 nM in HCT116 cells, for CCI-001-HSA NPs and free drug, respectively). Therefore, despite the low drug content level in the HSA nanoparticles of CCI-001, the formulation provides relevant concentrations for further in vivo studies in animal models due to high drug potency. CONCLUSIONS: The data support the potential use of albumin as a nanocarrier for CCI-001 in biological systems.


Subject(s)
Colorectal Neoplasms/drug therapy , Drug Delivery Systems , Nanoparticles , Tubulin Modulators/pharmacology , Animals , Cattle , Cell Line, Tumor , Chemistry, Pharmaceutical , Drug Carriers/chemistry , Drug Liberation , HCT116 Cells , Humans , Particle Size , Serum Albumin, Bovine/chemistry , Serum Albumin, Human/chemistry , Tubulin Modulators/administration & dosage , Tubulin Modulators/chemistry
9.
Radiology ; 300(3): 715-724, 2021 09.
Article in English | MEDLINE | ID: mdl-34227886

ABSTRACT

Background There is a paucity of randomized trials demonstrating superior efficacy of drug-coated balloon angioplasty (DCBA) compared with conventional percutaneous transluminal angioplasty (PTA) for below-the-knee arterial disease in patients with -critical limb ischemia. Purpose To compare DCBA versus PTA for below-the-knee lesions in participants with critical limb ischemia through 12 months. Materials and Methods In this prospective, randomized, two-center, double-blind superiority study, participants with critical limb ischemia with rest pain or tissue loss with atherosclerotic disease in the native below-the-knee arteries were randomly assigned (in a one-to-one ratio) to DCBA or PTA after stratification for diabetes and renal failure between November 2013 and October 2017. The primary efficacy end point was angiographic primary patency at 6 months analyzed on an intention-to-treat basis. Secondary end points through 12 months were composed of major adverse events including death and major amputations, wound healing, limb salvage, clinically driven target-lesion revascularization, and amputation-free survival. Primary and binary secondary end points, analyzed by using generalized-linear model and time-to-event analyses, were estimated with Kaplan-Meier survival curves and hazard ratios (Cox regression). Results Seventy participants (mean age, 61 years ± 10 [standard deviation]; 43 men) in the DCBA group and 68 (mean age, 64 years ± 10; 50 men) in the PTA group were evaluated. The percentage of patients with angiographic primary patency at 6 months was 43% (30 of 70) in the DCBA group and 38% (26 of 68) in the PTA group (P = .48). Through 12 months, the percentage of deaths was similar: 21% in the DCBA group and 16% in the PTA group (P = .43). Amputation-free survival rate assessed with Kaplan-Meier curves differed through 12 months: 59% (41 of 70) in the DCBA group compared with 78% (53 of 68) in the PTA group (P = .01). Conclusion In participants with critical limb ischemia, the drug-coated balloon angioplasty group and the conventional percutaneous transluminal angioplasty group had similar primary patency rates at 6 months after treatment of below-the-knee arteries. Amputation-free survival rates through 12 months were higher in the percutaneous transluminal angioplasty group. © RSNA, 2021 Online supplemental material is available for this article.


Subject(s)
Angioplasty, Balloon/instrumentation , Ischemia/surgery , Lower Extremity/blood supply , Paclitaxel/administration & dosage , Peripheral Arterial Disease/surgery , Tubulin Modulators/administration & dosage , Angiography , Contrast Media , Double-Blind Method , Drug Delivery Systems , Female , Humans , Iohexol , Ischemia/diagnostic imaging , Limb Salvage , Lower Extremity/diagnostic imaging , Male , Middle Aged , Peripheral Arterial Disease/diagnostic imaging , Prospective Studies
10.
Int J Mol Sci ; 22(8)2021 Apr 16.
Article in English | MEDLINE | ID: mdl-33923421

ABSTRACT

Vincristine-induced peripheral neurotoxicity (VIPN) is a very common side effect of vincristine chemotherapy among pediatric patients with cancer. Neuropathy may be sensory, motor and/or autonomic, with consequent reduction, delay or discontinuation of vincristine-chemotherapy, but also pain, disability, reduced quality of life of patients and an increase in medical costs. Vincristine acts out its antineoplastic function by altering the normal assembly and disassembly of microtubules, with their consequent mitosis block and death. Vincristine leads to VIPN through a complex mechanism of damage, which occurs not only on the microtubules, but also on the endothelium and the mitochondria of nerve cells. Furthermore, both patient-related risk factors (age, race, ethnicity and genetic polymorphisms) and treatment-related risk factors (dose, time of infusion and drug-drug interactions) are involved in the pathogenesis of VIPN. There is a lack of consensus about the prophylaxis and treatment of VIPN among pediatric oncologic patients, despite several molecules (such as gabapentin, pyridoxine and pyridostigmine, glutamic acid and glutamine) having been already investigated in clinical trials. This review describes the molecular mechanisms of VIPN and analyzes the risk factors and the principal drugs adopted for the prophylaxis and treatment of VIPN in pediatric patients with cancer.


Subject(s)
Antineoplastic Agents/toxicity , Neurotoxicity Syndromes/etiology , Peripheral Nervous System Diseases/etiology , Tubulin Modulators/toxicity , Vincristine/toxicity , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Child , Humans , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/prevention & control , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/prevention & control , Tubulin Modulators/administration & dosage , Tubulin Modulators/pharmacokinetics , Vincristine/pharmacokinetics
11.
Breast Cancer ; 28(4): 945-955, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33677779

ABSTRACT

BACKGROUND: It was reported that eribulin regulates the tumor microenvironment, including the immune system, by inducing vascular remodeling. Lymphocyte counts are a critical index of immune response in patients. The non-Asian, global EMBRACE study has suggested that baseline absolute lymphocyte count (ALC) may be a predictor of the survival benefit of eribulin in breast cancer patients. We examined whether the baseline ALC is a potential predictor of overall survival (OS) in Japanese patients with HER2-negative advanced breast cancer treated with eribulin. METHODS: This was a post hoc analysis of data from a post-marketing observational study of eribulin in Japan. The OS by baseline ALC was estimated using the Kaplan-Meier method, with the cut-off value of 1500/µL for ALC. The OS by baseline neutrophil-to-lymphocyte ratio (NLR), a general prognostic index in breast cancer patients, was also estimated, with the cut-off value of 3. RESULTS: The median OS was longer in patients with an ALC of ≥ 1500/µL than in those with an ALC of < 1500/µL (19.4 vs. 14.3 months; hazard ratio [HR]: 0.628; 95% confidence interval [CI]: 0.492, 0.801). Patients with an NLR of ≥ 3 showed shorter OS than those with an NLR of < 3 (13.2 vs. 18.8 months; HR: 1.552; 95% CI 1.254, 1.921), and NLR also separated OS in patients with an ALC of < 1500/µL. CONCLUSIONS: Consistent with the findings of a previous study involving a non-Asian, Western population, our study suggested that baseline ALC may be a predictive factor for the survival benefit of eribulin in Japanese patients.


Subject(s)
Breast Neoplasms/drug therapy , Furans/administration & dosage , Ketones/administration & dosage , Tubulin Modulators/administration & dosage , Administration, Intravenous , Adult , Aged , Breast Neoplasms/blood , Breast Neoplasms/mortality , Female , Follow-Up Studies , Humans , Japan/epidemiology , Lymphocyte Count , Middle Aged , Neutrophils , Proportional Hazards Models , Receptor, ErbB-2
13.
J Vasc Surg ; 73(1): 311-322, 2021 01.
Article in English | MEDLINE | ID: mdl-32890719

ABSTRACT

BACKGROUND: Peripheral vascular devices (stents and balloons) coated with paclitaxel were developed to address suboptimal outcomes associated with percutaneous revascularization procedures of the femoral-popliteal arteries. In randomized controlled trials (RCT), paclitaxel-coated devices (PCD) provided increased long-term patency and a decreased need for repeat revascularization procedures compared with uncoated devices. This finding resulted in the adoption of their use for endovascular lower extremity revascularization procedures. However, in late 2018 a study-level meta-analysis showed increased all-cause mortality at 2 years or more after the procedure in patients treated with PCDs. This review examines the subsequent data evaluation following the publication of the meta-analysis. METHODS: We review the published responses of physicians, regulatory agencies, and patient advocates during 15-month period after the meta-analysis. We present the additional data gathered from RCTs that comprised the meta-analysis and safety outcomes from large insurance databases in both the United States and Europe. RESULTS: Immediately after the publication of the meta-analysis, concern for patient safety resulted in less PCD use, the suspension of large RCTs evaluating their use, and the publication of a letter from the U.S. Food and Drug Administration informing physicians that there was uncertainty in the benefit-risk profile of these devices for indicated patients and that the potential risk should be assessed before the use of PCDs. Review of the meta-analysis found that a mortality signal was present, but criticisms included that the evaluation was performed on study-level, not patient-level data, and the studies in the analysis were heterogenous in device type, paclitaxel doses, and patient characteristics. Further, the studies were not designed to be pooled nor were they powered for evaluating long-term safety. Clinical characteristics associated with a drug effect or causal relationship were also absent. Specifically, there was no dose response, no clustering of causes of death, and a lack of signal consistency across geographic regions. As more long-term data became available in the RCTs the strength of the mortality signal diminished and analysis of real-world use in large insurance databases, showed that there was no significant increase in all-cause mortality associated with PCD use. CONCLUSIONS: The available data do not provide definitive proof for increased mortality with PCD use. A key observation is that trial design improvements will be necessary to better evaluate the risk-benefit profile of PCDs.


Subject(s)
Angioplasty, Balloon/methods , Arterial Occlusive Diseases/therapy , Femoral Artery , Paclitaxel/administration & dosage , Popliteal Artery , Arterial Occlusive Diseases/mortality , Coated Materials, Biocompatible , Dose-Response Relationship, Drug , Follow-Up Studies , Global Health , Humans , Survival Rate/trends , Time Factors , Tubulin Modulators/administration & dosage
14.
J Atheroscler Thromb ; 28(9): 954-962, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-33100279

ABSTRACT

AIM: The predictors of restenosis after endovascular therapy (EVT) with paclitaxel drug-coated balloons (DCBs) have not been clearly established. The present study aimed to investigate the association of post-procedural dissection, as evaluated using intravascular ultrasound (IVUS), with the risk of restenosis following femoropopliteal EVT with paclitaxel DCBs. METHODS: In the present single-center retrospective study, 60 de novo femoropopliteal lesions (44 patients) that underwent EVT with DCBs, without bail-out stenting, were enrolled. The primary outcome was 1-year primary patency. Risk factors for restenosis were evaluated using a Cox proportional hazards regression model and random survival forest analysis. RESULTS: The 1-year primary patency rate was 57.2% [95% confidence interval, 45%-72%]. IVUS-evaluated post-procedural dissection was significantly associated with the risk of restenosis (P=0.002), with the best cutoff point of 64º [range, 39º-83º]. The random survival forest analysis showed that the variable importance measure of IVUS-evaluated dissection was significantly lower than that of the reference vessel diameter (P<0.001), not different from that of the lesion length (P=0.41), and significantly higher than that of any other clinical feature (all P<0.05). CONCLUSION: IVUS-evaluated post-procedural dissection was associated with 1-year restenosis following femoropopliteal EVT with DCB.


Subject(s)
Endovascular Procedures , Paclitaxel/therapeutic use , Peripheral Arterial Disease/therapy , Tubulin Modulators/therapeutic use , Vascular Access Devices , Aged , Aged, 80 and over , Constriction, Pathologic/diagnosis , Constriction, Pathologic/etiology , Endovascular Procedures/adverse effects , Female , Humans , Male , Paclitaxel/administration & dosage , Peripheral Arterial Disease/diagnosis , Prognosis , Retrospective Studies , Treatment Outcome , Tubulin Modulators/administration & dosage , Vascular Access Devices/adverse effects
15.
Am Heart J ; 232: 71-83, 2021 02.
Article in English | MEDLINE | ID: mdl-33157067

ABSTRACT

The Registry Assessment of Peripheral Devices (RAPID) convened a multidisciplinary group of stakeholders including clinicians, academicians, regulators and industry representatives to conduct an in-depth review of limitations associated with the data available to assess the paclitaxel mortality signal. Available studies were evaluated to identify strengths and limitations in the study design and data quality, which were translated to lessons learned to help guide the design, execution, and analyses of future studies. We suggest numerous actionable responses, such as the development and use of harmonized data points and outcomes in a consensus lean case report form. We advocate for reduction in missing data and efficient means for accrual of larger sample sizes in Peripheral arterial disease studies or use of supplemental datasets. Efforts to share lessons learned and working collaboratively to address such issues may improve future data in this device area and ultimately benefit patients. Condensed Abstract: Data sources evaluating paclitaxel-coated devices were evaluated to identify strengths and limitations in the study design and data quality, which were translated to lessons learned to help guide the design, execution, and analyses of future studies. We suggest numerous actionable responses, which we believe may improve future data in this device area and ultimately benefit patients.


Subject(s)
Angioplasty , Drug-Eluting Stents , Mortality , Paclitaxel/administration & dosage , Peripheral Arterial Disease/surgery , Tubulin Modulators/administration & dosage , Advisory Committees , Angioplasty, Balloon , Atherectomy , Common Data Elements , Data Accuracy , Data Collection , Femoral Artery/surgery , Humans , Meta-Analysis as Topic , Popliteal Artery , Randomized Controlled Trials as Topic , Reproducibility of Results , Risk Assessment , Stents
16.
Bioorg Med Chem Lett ; 30(24): 127634, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33148516

ABSTRACT

Gold nanoparticles are promising drug delivery agents with the potential to deliver chemotherapeutic agents to tumour sites. The highly cytotoxic maytansinoid tubulin inhibitor DM1 has been attached to gold nanoparticles and shows tumour growth inhibition in mouse models of hepatocellular carcinoma. Attempting to improve the stability of the gold-cytotoxin bond led to the design and synthesis of novel maytansinoids with improved potency in cell viability assays and improved in vivo tolerability compared to the DM1 analogues. These novel maytansines may also have applications in other methods of drug delivery, for example as the cytotoxic component of antibody drug conjugates.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Gold/chemistry , Liver Neoplasms/drug therapy , Maytansine/administration & dosage , Nanoconjugates/chemistry , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Humans , Liver Neoplasms/pathology , Maytansine/analogs & derivatives , Maytansine/pharmacology , Mice , Models, Molecular , Tubulin Modulators/administration & dosage , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology
17.
Drugs ; 80(15): 1607-1613, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32936437

ABSTRACT

Belantamab mafodotin (BLENREP™; belantamab mafodotin-blmf) is a first-in-class monoclonal antibody-drug conjugate (ADC) that has been developed for the treatment of multiple myeloma by GlaxoSmithKline. The ADC comprises an antibody targeting B-cell maturation antigen (BCMA) conjugated to the microtubule inhibitor monomethyl auristatin F (MMAF). The antibody moiety binds to BCMA on the tumour cell surface, delivering the cytotoxic microtubule inhibitor MMAF to the therapeutic target. Based on preliminary results from the multinational DREAMM-2 trial, belantamab mafodotin was approved in early August 2020 in the USA for the treatment of relapsed or refractory multiple myeloma in adult patients who have received at least four prior therapies, including an anti CD38 monoclonal antibody, a proteasome inhibitor, and an immunomodulatory agent. The ADC was also approved in the EU for this indication in late August 2020. This article summarizes the milestones in the development of belantamab mafodotin leading to this first approval.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Drug Approval , Immunoconjugates/administration & dosage , Multiple Myeloma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Adult , Animals , Antibodies, Monoclonal, Humanized/adverse effects , B-Cell Maturation Antigen/antagonists & inhibitors , B-Cell Maturation Antigen/metabolism , Cell Line, Tumor , Clinical Trials as Topic , Disease Progression , Drug Resistance, Neoplasm , Humans , Immunoconjugates/adverse effects , Infusions, Intravenous , Multiple Myeloma/diagnosis , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Oligopeptides/administration & dosage , Oligopeptides/adverse effects , Progression-Free Survival , Tubulin Modulators/administration & dosage , Tubulin Modulators/adverse effects , Xenograft Model Antitumor Assays
18.
Gynecol Oncol ; 159(2): 539-545, 2020 11.
Article in English | MEDLINE | ID: mdl-32912664

ABSTRACT

OBJECTIVE: We sought to evaluate the role of intrinsic chromosomal aberrations in determining favorable outcome to weekly paclitaxel (WP) in patients with epithelial ovarian cancer (EOC). METHODS: We evaluated the common genomic aberrations of two patients with EOC and exceptional WP response in the GENIUS study (NCT03740503). We then searched for potential markers of unusual outcomes to WP in a validation cohort. We performed shallow whole genome sequencing (sWGS) in the tumor tissue of women with EOC considered as short-responders (SR; progression with ≤3 cycles) and long-responders (LR; response at ≥8 cycles) to WP monotherapy. RESULTS: We identified two women with exceptional response to WP, lasting over four years, who shared chromosome 8 gain as a common genomic aberration. In order to validate our findings, we reviewed 188 patients with EOC treated with WP and selected 61 women (39 SR, 22 LR) with unusual responses. By sWGS, there was no differential alterations in the copy number changes in chromosome 8, or in genes related to angiogenesis, tubulin superfamily, cell-cycle, apoptosis and paclitaxel metabolism or transportation pathways. Amongst the LR group, we identified six exceptionally long responders (ExLR), with responses lasting over a year. In an exploratory analysis, there was increased amplification of angiogenesis (VEGFB, MMP9), tubulin superfamily (TSC2) and apoptosis related genes (BCL2L1, BAD) in ExLR compared to SR. We identified one patient with a complete response to WP for over 7 years. Molecular profiling identified unique amplifications in interleukin related genes (CXCR1, CXCR2, IL1A, IL1B), not detected in other patients. CONCLUSION: Intrinsic tumor pathways may impact outcome with weekly paclitaxel monotherapy and further investigations are required.


Subject(s)
Carcinoma, Ovarian Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Paclitaxel/administration & dosage , Tubulin Modulators/administration & dosage , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Carcinoma, Ovarian Epithelial/genetics , DNA Copy Number Variations/immunology , Drug Administration Schedule , Drug Resistance, Neoplasm/genetics , Female , Humans , Middle Aged , Ovarian Neoplasms/genetics , Progression-Free Survival , Exome Sequencing/methods
19.
Cancer Lett ; 495: 76-88, 2020 12 28.
Article in English | MEDLINE | ID: mdl-32920198

ABSTRACT

Lung cancer is the most common cause of cancer associated mortality. Chemotherapeutic agents, such as paclitaxel, are important treatment options but drug resistance often develops upon prolonged use. We report here the preclinical evaluation of a new orally available tubulin inhibitor, VERU-111, which can overcome several ABC-transporters mediated multi-drug resistance associated with taxane treatment. In vitro, VERU-111 prevents cell proliferation, invasion, migration and colony formation in both paclitaxel-sensitive and paclitaxel-resistant A549 lung cancer cells. VERU-111 effectively inhibits tubulin polymerization, arrests cells in G2/M phase, and induces cancer cell apoptosis. Further evaluation of various apoptotic proteins revealed that treatment of VERU-111 increases the expression of cleaved-PARP, cleaved-caspase-3 and p-histone H3 proteins. In vivo, orally administered VERU-111 in a paclitaxel-sensitive A549 xenograft model strongly inhibits tumor growth in a dose-dependent manner and is equally potent with paclitaxel. When tested in a highly paclitaxel-resistant A549/TxR tumor model, VERU-111 is as effective as the parental A549 model in significantly reducing the tumor volume, whereas paclitaxel is essentially ineffective. Collectively, this study showed that VERU-111 is a promising new generation of anti-tubulin agent for the treatment of taxane-resistant lung cancer.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Imidazoles/administration & dosage , Indoles/administration & dosage , Lung Neoplasms/drug therapy , Paclitaxel/administration & dosage , Tubulin Modulators/administration & dosage , A549 Cells , Administration, Oral , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Lung Neoplasms/metabolism , Mice , Paclitaxel/pharmacology , Tubulin Modulators/pharmacology , Xenograft Model Antitumor Assays
20.
Cancer Lett ; 495: 22-32, 2020 12 28.
Article in English | MEDLINE | ID: mdl-32931884

ABSTRACT

The tubulin colchicine binding site has been recognized as an attractive drug target to combat cancer, but none of the candidate drugs have been approved for medical treatment. We recently identified a structurally distinct small molecule S-40 as an oral potent tubulin destabilizing agent. Crystal structure analysis of S-40 in a complex with tubulin at a resolution of 2.4 Å indicated that S-40 occupies all 3 zones in the colchicine pocket with interactions different from known microtubule inhibitors, presenting unique effects on assembly and curvature of tubulin dimers. S-40 overcomes paclitaxel resistance and lacks neurotoxicity, which are the main obstacles limiting clinical applications of paclitaxel. Moreover, S-40 harbors the ability to inhibit growth of cancer cell lines as well as patient-derived organoids, induce mitotic arrest and cell apoptosis. Xenograft mouse models of human prostate cancer DU145, non-small cell lung cancer NCI-H1299 and paclitaxel-resistant A549 were strongly restrained without apparent side effects by S-40 oral administration once daily. These findings provide evidence for the development of S-40 as the next generation of orally effective microtubule inhibitors for cancer therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Tubulin Modulators/administration & dosage , Tubulin/chemistry , Tubulin/metabolism , A549 Cells , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colchicine/chemistry , Colchicine/pharmacology , Crystallography, X-Ray , Humans , Male , Mice , Models, Molecular , Neoplasms/metabolism , Paclitaxel/pharmacology , Protein Conformation , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...