Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 491
Filter
1.
Am J Ophthalmol ; 262: 161-169, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38307213

ABSTRACT

PURPOSE: To determine the incidence and type of strabismus in patients with uveal melanoma treated with plaque brachytherapy. DESIGN: Multicenter, retrospective incidence estimation study. METHODS: A total of 438 eyes of 438 patients with uveal melanoma treated with plaque brachytherapy between October 2011 and May 2021. Intervention was Iodine 125, and Palladium 103 plaque brachytherapy. The variables reviewed included incidence of nonresolving strabismus post-plaque brachytherapy, type of strabismus developed, extraocular muscles operated, and modality of treatment received. RESULTS: A total of 438 patients underwent plaque brachytherapy treatment for uveal melanoma. Eleven patients developed strabismus post-plaque brachytherapy (2.5%, n = 11/438). Of these patients, 5 (1.1%, n = 5/438) developed strabismus immediately postoperation. Specifically, 2 patients (0.5%, n = 2/438) developed strabismus immediately postoperation due to slipped muscles, 2 patients (0.5%, n = 2/438) due to decompensated phorias, and 1 patient (0.5%, n = 1/438) due to a fibrotic muscle. Six patients (1.4%, n = 6/438) developed late-onset sensory strabismus. A total of 355 patients (81.1%, n = 355/438) had their extraocular muscles disinserted during surgery, with the lateral rectus being the most common, accounting for 45.4% (n = 161/355), followed by the superior rectus at 26.8% (n = 95/355). Strabismus surgery was the most common treatment modality, comprising 72.7% (n = 8/11) of patients. CONCLUSIONS: The incidence of strabismus after plaque brachytherapy treatment for uveal melanoma was low and primarily classified as late-onset sensory strabismus. Previous studies may underestimate the long-term incidence of strabismus after plaque brachytherapy by focusing primarily on strabismus present immediately postoperatively.


Subject(s)
Brachytherapy , Iodine Radioisotopes , Melanoma , Strabismus , Uveal Neoplasms , Humans , Brachytherapy/adverse effects , Melanoma/radiotherapy , Melanoma/epidemiology , Strabismus/etiology , Strabismus/epidemiology , Incidence , Uveal Neoplasms/radiotherapy , Uveal Neoplasms/epidemiology , Retrospective Studies , Male , Female , Middle Aged , Aged , Iodine Radioisotopes/therapeutic use , Iodine Radioisotopes/adverse effects , Adult , Aged, 80 and over , Oculomotor Muscles/radiation effects , Oculomotor Muscles/surgery , Palladium/therapeutic use , Radioisotopes/therapeutic use , Radiation Injuries/etiology , Radiation Injuries/epidemiology
2.
Int J Hyperthermia ; 41(1): 2299480, 2024.
Article in English | MEDLINE | ID: mdl-38189281

ABSTRACT

OBJECTIVE: To investigate the potential of hybrid Pd/Fe-oxide magnetic nanoparticles designed for thermo-brachytherapy of breast cancer, considering their specific loss power (SLP) and clinical constraints in the applied magnetic field. METHODS: Hybrid nanoparticles consisting of palladium-core and iron oxide shell of increasing thickness, were suspended in water and their SLPs were measured at varying magnetic fields (12-26 mT peak) and frequencies (50-730 kHz) with a commercial alternating magnetic field generator (magneTherm™ Digital, nanoTherics Ltd.). RESULTS: Validation of the heating device used in this study with commercial HyperMag-C nanoparticles showed a small deviation (±4%) over a period of 1 year, confirming the reliability of the method. The integration of dual thermometers, one in the center and one at the bottom of the sample vial, allowed monitoring of homogeneity of the sample suspensions. SLPs measurements on a series of nanoparticles of increasing sizes showed the highest heating for the diameter of 21 nm (SLP = 225 W/g) at the applied frequencies of 346 and 730 kHz. No heating was observed for the nanoparticles with the size <14 nm, confirming the importance of the size-parameter. The heating ability of the best performing Pd/Fe-oxide-21 was calculated to be sufficient to ablate tumors with a radius ±4 and 12 mm using 10 and 1 mg/mL nanoparticle concentration, respectively. CONCLUSIONS: Nanoparticles consisting of non-magnetic palladium-core and magnetic iron oxide shell are suitable for magnetic hyperthermia/thermal ablation under clinically safe conditions of 346 kHz and 19.1 mT, with minimal eddy current effects in combination with maximum SLP.


Subject(s)
Brachytherapy , Nanoparticles , Oxides , Palladium/therapeutic use , Reproducibility of Results , Magnetic Fields
3.
Adv Sci (Weinh) ; 10(33): e2304002, 2023 11.
Article in English | MEDLINE | ID: mdl-37807805

ABSTRACT

Ulcerative colitis (UC) is a chronic inflammatory bowel disease mainly involving the colon and rectum, which features recurrent mucosal inflammation. The excessive production of reactive oxygen species (ROS) is a trigger for pathological changes such as cell apoptosis and disordered immune microenvironments, which are crucial for the progression of UC and can be a promising therapeutic target. Nowadays, the development of targeted therapeutic strategies for UC is still in its infancy. Thus, developing effective therapies based on ROS scavenging and elucidating their molecular pathways are urgently needed. Herein, a biomimetic nanoformulation (Pd@M) with cubic palladium (Pd) as the core and macrophage-derived extracellular vesicles (MEVs) as the shell is synthesized for the treatment of UC. These Pd@M nanoformulations exhibit multienzyme-like activities for effective ROS scavenging, excellent targeting ability as well as good biocompatibility. It is verified that Pd@M can regulate the polarization state of macrophages by inhibiting glycolysis, and decrease neutrophil infiltration and recruitment. In this way, the colonic inflammatory and immune microenvironment is remodeled, and apoptosis is prevented, ultimately improving colonic mucosal barrier function and alleviating colitis in the mouse model. This finding provides a promising alternative option for the treatment of UC patients.


Subject(s)
Colitis, Ulcerative , Extracellular Vesicles , Animals , Mice , Humans , Colitis, Ulcerative/drug therapy , Palladium/therapeutic use , Reactive Oxygen Species/metabolism , Macrophages/metabolism , Extracellular Vesicles/metabolism , Homeostasis
4.
BJS Open ; 7(3)2023 05 05.
Article in English | MEDLINE | ID: mdl-37254902

ABSTRACT

INTRODUCTION: Pancreatic cancer has one of the worst prognoses of all cancers. Patients with locally advanced pancreatic cancer have a 12.7-20.2 per cent chance of receiving curative surgery after induction systemic chemotherapy. Intratumoral injection therapies have been studied as complementary treatment options for improved local tumour control. The aim of this systematic review was to provide an overview of intratumoral injection therapies, their safety, and oncological outcome in patients with locally advanced pancreatic cancer. METHODS: A literature search was conducted in PubMed, Embase and the Cochrane Library for articles written in English up to 28 November 2022. All study designs involving at least five patients with locally advanced pancreatic cancer who were treated with an intratumoral injection therapy were included. Critical appraisal of the included studies was performed using the Newcastle-Ottawa scale. RESULTS: After evaluation of the 1680 articles yielded by the systematic search, 52 studies treating 1843 patients were included. Included intratumoral injection treatment modalities comprised iodine-125 (125I) seed brachytherapy (32 studies, 1283 patients), phosphorus-32 (32P) microbrachytherapy (5 studies, 133 patients), palladium-103 (103Pd) seed brachytherapy (2 studies, 26 patients), immunotherapy (9 studies, 330 patients), and chemotherapy (4 studies, 71 patients). Overall survival ranged between 7.0 and 16.0 months for 125I, 5.2 and 15.5 months for 32P, 6.9 and 10.0 months for 103Pd, 5.8 and 13.8 months for immunotherapy, and 9.0 and 16.2 months for chemotherapy. Severe complication (greater than or equal to grade III complications using Clavien-Dindo classification) rates were 6.2 per cent for 125I, 49.2 per cent for 32P, 15 per cent for 103Pd, 57.9 per cent for immunotherapy, and 0 per cent for chemotherapy. CONCLUSION: Five intratumoral injection therapies are described and an overview is reported. Some intratumoral injection therapies for patients with locally advanced pancreatic cancer seem safe, although 32P microbrachytherapy and immunotherapy require additional evidence. Currently available data are insufficient to provide firm conclusions regarding the added value to survival. The potential advantage of intratumoral injection therapies complementary to conventional care should be studied in well designed RCTs.


Subject(s)
Iodine Radioisotopes , Pancreatic Neoplasms , Humans , Iodine Radioisotopes/therapeutic use , Palladium/therapeutic use , Injections, Intralesional , Pancreatic Neoplasms/drug therapy
5.
Radiother Oncol ; 183: 109599, 2023 06.
Article in English | MEDLINE | ID: mdl-36889593

ABSTRACT

BACKGROUND AND PURPOSE: Pd-103 and I-125 are commonly used in low dose rate (LDR) brachytherapy for prostate cancer. Comparisons of outcomes by isotope type are limited, but Pd-103 has distinct radiobiologic advantages over I-125 despite its lesser availability outside the United States. We evaluated oncologic outcomes after Pd-103 vs I-125 LDR monotherapy for prostate cancer. MATERIALS AND METHODS: We retrospectively analyzed databases at 8 institutions for men who received definitive LDR monotherapy with Pd-103 (n = 1,597) or I-125 (n = 7,504) for prostate cancer. Freedom from clinical failure (FFCF) and freedom from biochemical failure (FFBF) stratified by isotope were analyzed by Kaplan-Meier univariate and Cox multivariate analyses. Biochemical cure rates (prostate-specific antigen level ≤ 0.2 ng/mL between 3.5 and 4.5 years of follow-up) by isotype were calculated for men with at least 3.5 years of follow-up and compared by univariate and multivariate logistic regression. RESULTS: Compared with I-125, Pd-103 led to higher 7-year rates of FFBF (96.2% vs 87.6%, P < 0.001) and FFCF (96.5% vs 94.3%, P < 0.001). This difference held after multivariate adjustment for baseline factors (FFBF hazard ratio [HR] = 0.31, FFCF HR = 0.49, both P < 0.001). Pd-103 was also associated with higher cure rates on univariate (odds ratio [OR] = 5.9, P < 0.001) and multivariate (OR = 6.0, P < 0.001) analyses. Results retained significance in sensitivity analyses of data from the 4 institutions that used both isotopes (n = 2,971). CONCLUSIONS: Pd-103 monotherapy was associated with higher FFBF, FFCF, and biochemical cure rates, and suggests that Pd-103 LDR may lead to improved oncologic outcomes compared with I-125.


Subject(s)
Brachytherapy , Prostatic Neoplasms , Male , Humans , Brachytherapy/methods , Iodine Radioisotopes/therapeutic use , Prostate , Palladium/therapeutic use , Retrospective Studies , Radiotherapy Dosage , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/drug therapy , Prostate-Specific Antigen , Follow-Up Studies
6.
Front Cell Infect Microbiol ; 13: 1340135, 2023.
Article in English | MEDLINE | ID: mdl-38292858

ABSTRACT

Staphylococcus aureus leads to diverse infections, and their treatment relies on the use of antibiotics. Nevertheless, the rise of antibiotic resistance poses an escalating challenge and various mechanisms contribute to antibiotic resistance, including modifications to drug targets, enzymatic deactivation of drugs, and increased efflux of antibiotics. Hence, the quest for innovative antimicrobial solutions has intensified in the face of escalating antibiotic resistance and the looming threat of superbugs. The NorA protein of S. aureus, classified as an efflux pump within the major facilitator superfamily, when overexpressed, extrudes various substances, including fluoroquinolones (such as ciprofloxacin) and quaternary ammonium. Addressing this, the unexplored realm of inorganic and organometallic compounds in medicinal chemistry holds promise. Notably, the study focused on investigating two different series of palladium-based metal complexes consisting of QSL_PA and QSL_PB ligands to identify a potent NorA efflux pump inhibitor that can restore the susceptibility to fluoroquinolone antibiotics. QSL_Pd5A was identified as a potent efflux pump inhibitor from the real-time efflux assay. QSL_Pd5A also resensitized SA1199B to ciprofloxacin at a low concentration of 0.125 µg/mL without elucidating cytotoxicity on the NRK-62E cell line. The in vitro findings were substantiated by docking results, indicating favorable interactions between QSL_Pd5A and the NorA efflux pump.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Humans , Staphylococcus aureus , Fluoroquinolones/pharmacology , Fluoroquinolones/metabolism , Palladium/pharmacology , Palladium/metabolism , Palladium/therapeutic use , Ciprofloxacin/pharmacology , Anti-Bacterial Agents/therapeutic use , Staphylococcal Infections/drug therapy , Bacterial Proteins/metabolism , Microbial Sensitivity Tests
7.
J Mater Chem B ; 10(48): 10027-10041, 2022 12 14.
Article in English | MEDLINE | ID: mdl-36458841

ABSTRACT

Reactive oxygen species (ROS) have become an effective "weapon" for cancer therapy due to their strong oxidation and high anti-tumor activity. Photodynamic therapy (PDT) is one of the classical methods to induce reactive oxygen species. Therefore, an ultraminiature palladium ruthenium alloy (sPdRu) and Ru(II) were combined with thermally responsive phase change materials (PCMs). Polypyridyl-complex (RCE) co-encapsulation was performed to obtain thermally responsive nanoparticles (PdRu-RCE@PCMNPs) for multimodal synergistic anti-breast cancer therapy. On the one hand, the thermosensitive PCM protective layer can realize the slow release of sPdRu, and then catalyze the production of oxygen from tumor endogenous H2O2 to perform RCE-mediated PDT. At the same time, sPdRu further increased ROS levels through peroxidase (POD) activity. On the other hand, sPdRu has high photothermal conversion efficiency and can be effectively used for photothermal therapy and photodynamic therapy. Importantly, PdRu-RCE@PCM NPs not only can effectively inhibit primary tumor growth, but also can inhibit tumor metastasis. In addition, due to the effective accumulation of sPdRu and RCE, PdRu-RCE@PCM NPs also show excellent fluorescence and photothermal imaging capabilities of tumors, which can be used for tumor tracing and evaluation of treatment. Accordingly, PdRu-RCE@PCM NPs are useful in treating primary tumors and inhibiting tumor metastasis.


Subject(s)
Breast Neoplasms , Photochemotherapy , Ruthenium , Mice , Animals , Humans , Female , Reactive Oxygen Species , Ruthenium/pharmacology , Palladium/pharmacology , Palladium/therapeutic use , Hydrogen Peroxide/therapeutic use , Cell Line, Tumor , Mice, Inbred BALB C , Breast Neoplasms/pathology
8.
J Nanobiotechnology ; 20(1): 299, 2022 Jun 25.
Article in English | MEDLINE | ID: mdl-35752849

ABSTRACT

Traumatic brain injury (TBI) is a cause of disability and death worldwide, but there are currently no specific treatments for this condition. Release of excess reactive oxygen species (ROS) in the injured brain leads to a series of pathological changes; thus, eliminating ROS could be a potential therapeutic strategy. Herein, we synthesized insulin-incubated ultrasmall palladium (Pd@insulin) clusters via green biomimetic chemistry. The Pd@insulin clusters, which were 3.2 nm in diameter, exhibited marked multiple ROS-scavenging ability testified by the theoretical calculation. Pd@insulin could be rapidly excreted via kidney-urine metabolism and induce negligible adverse effects after a long-time treatment in vivo. In a TBI mouse model, intravenously injected Pd@insulin clusters aggregated in the injured cortex, effectively suppressed excessive ROS production, and significantly rescued motor function, cognition and spatial memory. We found that the positive therapeutic effects of the Pd@insulin clusters were mainly attributed to their ROS-scavenging ability, as they inhibited excessive neuroinflammation, reduced cell apoptosis, and prevented neuronal loss. Therefore, the ability of Pd@insulin clusters to effectively eliminate ROS, as well as their simple structure, easy synthesis, low toxicity, and rapid metabolism may facilitate their clinical translation for TBI treatment.


Subject(s)
Brain Injuries, Traumatic , Brain Injuries , Animals , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Insulin , Mice , Palladium/pharmacology , Palladium/therapeutic use , Reactive Oxygen Species/metabolism
9.
Brachytherapy ; 21(5): 599-604, 2022.
Article in English | MEDLINE | ID: mdl-35725549

ABSTRACT

PURPOSE/OBJECTIVE: To compare toxicity profiles of low-dose rate (LDR) and high-dose rate (HDR) brachytherapy boost combined with ultra-hypofractionated external beam radiation therapy (UH-EBRT). MATERIALS/METHODS: 99 patients with intermediate-risk prostate cancer underwent an HDR (n = 59) or LDR (n = 40) boost combined with UH-EBRT (5 Gy x 5) . HDR (Ir-192) was delivered a single dose (15 Gy) and LDR (Pd-103) prescription dose was 100 Gy. Median baseline IPSS was 5 for both cohorts. Median follow-up was 29.3mos. Cumulative incidences were calculated for toxicity. Fisher exact tests were used to evaluate associations. RESULTS: Overall incidence of grade 2 genitourinary toxicity for the entire cohort at 12 and 24 months was 21% and 29%, respectively. The incidence of grade 2 genitourinary toxicity at 12 and 24 months was higher for LDR cohort compared with HDR cohort (45% vs 5.1% and 55% vs 11%; p<0.001). On MVA, only treatment regimen (LDR versus HDR) was associated with grade 2+ genitourinary toxicity (p<0.001). Two patients experienced grade 2 rectal toxicity in each cohort. No grade > 3 toxicities were observed. CONCLUSIONS: Both LDR and HDR brachytherapy combined with UH-EBRT had favorable toxicity profiles, but significantly less grade 2+ genitourinary toxicity was observed in patients receiving HDR.


Subject(s)
Brachytherapy , Prostatic Neoplasms , Brachytherapy/methods , Humans , Male , Palladium/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , Radioisotopes/therapeutic use , Radiotherapy Dosage
10.
Technol Cancer Res Treat ; 21: 15330338221106852, 2022.
Article in English | MEDLINE | ID: mdl-35712977

ABSTRACT

Introduction: Brachytherapy using permanently implantable collagen tiles containing cesium-131 (Cs-131) is indicated for treatment of malignant intracranial neoplasms. We quantified Cs-131 source migration and modeled the resulting dosimetric impact for Cs-131, iodine-125 (I-125), and palladium-103 (Pd-103). Methods and Materials: This was a retrospective analysis of a subgroup of patients enrolled in a prospective, single-center, nonrandomized, clinical trial (NCT03088579) of Cs-131 collagen tile brachytherapy. Postimplant Cs-131 plans and hypothetical I-125 and Pd-103 calculations were compared for 20 glioblastoma patients for a set seed geometry. Dosimetric impact of decay and seed migration was calculated for 2 hypothetical scenarios: Scenario 1, assuming seed positions on a given image set were unchanged until acquisition of the subsequent set; Scenario 2, assuming any change in seed positions occurred the day following acquisition of the prior images. Seed migration over time was quantified for a subset of 7 patients who underwent subsequent image-guided radiotherapy. Results: Mean seed migration was 1.7 mm (range: 0.7-3.1); maximum seed migration was 4.3 mm. Mean dose to the 60 Gy volume differed by 0.4 Gy (0.6%, range 0.1-1.0) and 0.9 Gy (1.5%, range 0.2-1.7) for Cs-131, 1.2 Gy (2.0%, range 0.1-2.1) and 1.6 Gy (2.6%, range 1.2-2.6) for I-125, and 0.8 Gy (1.3%, range 0.2-1.5) and 1.4 Gy (2.3%, range 0.3-1.9) for Pd-103, for Scenarios 1 and 2, respectively, compared with the postimplant plan. For a set seed geometry mean implant dose was higher for Pd-103 (1.3 times) and I-125 (1.1 times) versus Cs-131. Dose fall-off was steepest for Pd-103: gradient index 1.88 versus 2.23 (I-125) and 2.40 (Cs-131). Conclusions: Dose differences due to source migration were relatively small, suggesting robust prevention of seed migration from Cs-131-containing collagen tiles. Intratarget heterogeneity was greater with Pd-103 and I-125 than Cs-131. Dose fall-off was fastest with Pd-103 followed by I-125 and then Cs-131.


Subject(s)
Brachytherapy , Brain Neoplasms , Brachytherapy/methods , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Cesium Radioisotopes/therapeutic use , Humans , Iodine Radioisotopes/therapeutic use , Palladium/therapeutic use , Prospective Studies , Radioisotopes , Radiotherapy Dosage , Retrospective Studies
11.
J Nanobiotechnology ; 20(1): 186, 2022 Apr 12.
Article in English | MEDLINE | ID: mdl-35413839

ABSTRACT

BACKGROUND: Sonodynamic therapy (SDT) is a burgeoning modality for cancer therapy owing to its high tissue-penetrating capability, controllability and safety. Whereas, the undesirable reactive oxygen species (ROS) yield of sonosensitizers and tumor hypoxia are two vulnerable spots of SDT. Therefore, it is an advisable strategy to augment ROS level and simultaneously relieve hypoxia for SDT to arrive its full potential in cancer treatment. RESULTS: In this work, the defected two-dimensional (2D) Pd/H-TiO2 nanosheets (NSs) with triple antineoplastic properties were dexterously elaborated and engineered using a facile one-pot Pd-catalyzed hydrogenation tactic by loading a tiny amount of Pd and then inletting hydrogen flow at atmospheric pressure and temperature. The 2D black Pd/H-TiO2 NSs with oxygen defects exerted eximious SDT effect based on the decreased bandgap that made it easier for the separation of electrons and holes when triggered by ultrasound as theoretically guided by density functional theory calculations. Additionally, Pd/H-TiO2 NSs could serve as Fenton-like agents because of the presence of oxygen defects, facilitating the conversion of hydrogen peroxide into hydroxyl radicals for exerting the chemodynamic therapy (CDT). Simultaneously, the introduced tiny Pd component possessed catalase-like activity responsible for oxygen production to ameliorate hypoxic condition and thus contributed to improving SDT and CDT efficacies. Both in vitro and in vivo results provided compelling evidences of high ROS yield and aggrandized sono-chemodynamic effect of Pd/H-TiO2 nanosonosensitizers with the detailed underlying mechanism investigation by RNA sequencing. CONCLUSION: This work delves the profound potential of Pd-catalyzed hydrogenated TiO2 on oncotherapy, and the effective antineoplastic performance and ignorable therapeutic toxicity make it a powerful competitor among a cornucopia of nanosonosensitizers.


Subject(s)
Antineoplastic Agents , Neoplasms , Ultrasonic Therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Humans , Hypoxia/drug therapy , Neoplasms/drug therapy , Oxygen , Palladium/pharmacology , Palladium/therapeutic use , Reactive Oxygen Species , Titanium , Ultrasonic Therapy/methods
12.
J Healthc Eng ; 2022: 4993405, 2022.
Article in English | MEDLINE | ID: mdl-35345664

ABSTRACT

Objective: Prostate cancer is one of the most common types of cancer found to occur in males and is ranked as the second-highest cause of cancer-associated deaths among male patients. In this study, we have shown the influence of a new palladium-based anticancer agent in contrast to the six distinct prostate cancer lines and the primary cultures. Methods: In this study, we have used six distinct prostate cell lines, that is, PNT2-C2, LNCaP, BPH-1, PC-3, PNT1A, and P4E6. The MTP and ATP assay were performed to evaluate the growth of the cell and the flow cytometry to investigate the status of the cell cycle. The antigrowth effect of the palladium complex was evaluated against different cell lines at three time zones 24 h, 48 h, and 72 h. [PdCl(terpy)] (capsule)-2H2O is synthesized by direct encapsulation of equimolar amounts of capsule ions into [Pd (terpy) Cl] Cl-2H2O. Results: A comparative analysis was done on 25 mM etoposide and 12 mM cisplatin, cytotoxic agents. The lowest IC50 value at 72 hours was 0.128 mM for BPH-1 cell lines with 0.139 mM, whereas PNT2-C2 cells were found to be most resistant with IC50 values of 0.829 mM. The antigrowth effect of palladium complex on cell lines was measured using the MTS assay at 24, 48, and 72 hours. BPH-1, PNT2-C2, and PNT1A either possess normal tissues or have benign prostatic hyperplasia tissues whereas P4E6, PC-3, and LNCaP cell lines possess malignant origin. The Pd complex exhibited significant cytotoxic action in stem cells when compared against etoposide. An antigrowth effect was reported for Pd complex at lower concentration, but it was more cytotoxic than etoposide with significant cytotoxicity (P=0.001). Conclusion: The palladium complex experienced a substantial antigrowth influence over most of the prostate tumor cell lines and the primary cultures, eventually, leading to the implementation of this Pd complex in the treating procedure of metastatic prostate cancer, which is tremendously resistant to the traditional treatment.


Subject(s)
Antineoplastic Agents , Prostatic Hyperplasia , Prostatic Neoplasms , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Epithelial Cells/metabolism , Epithelial Cells/pathology , Etoposide/metabolism , Etoposide/pharmacology , Etoposide/therapeutic use , Humans , Male , Palladium/metabolism , Palladium/pharmacology , Palladium/therapeutic use , Prostate , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Stem Cells
13.
Nano Lett ; 22(2): 751-760, 2022 01 26.
Article in English | MEDLINE | ID: mdl-35030010

ABSTRACT

In recent years, targeted therapies and immunotherapeutics, along with conventional chemo- and radiotherapy, have greatly improved cancer treatments. Unfortunately, in cancer patients, anemia, either as a complication of cancer progression or as the result of cancer treatment, undermines the expected therapeutic efficacy. Here, we developed a smart nanosystem based on the palladium nanoplates (PdPLs) to deliver tocilizumab (TCZ, a widely used IL-6R antibody) to the liver for specific blockade of IL-6/IL-6R signaling to correct anemia. With chemical modifications, this nanosystem delivered a large mass of TCZ and enhanced liver delivery, inducing a marked suppression of hepcidin expression as a result of diminished IL-6 signaling. Through this mechanism, significant suppression of tumor progression was realized (at least in part) because of the corrected anemia after treatment.


Subject(s)
Anemia , Neoplasms , Anemia/drug therapy , Anemia/etiology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Neoplasms/complications , Neoplasms/drug therapy , Palladium/pharmacology , Palladium/therapeutic use , Receptors, Interleukin-6/antagonists & inhibitors , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism
14.
Brachytherapy ; 21(2): 255-259, 2022.
Article in English | MEDLINE | ID: mdl-35031255

ABSTRACT

The treatment of borderline resectable (BR) pancreatic cancer is challenging and requires a multidisciplinary approach with chemotherapy, radiation and surgical resection. Despite using chemotherapy and radiotherapy in the neoadjuvant setting, achievement of negative surgical margins remains technically challenging. Positive margins are associated with increased local recurrences and worse overall survival and there are no standard options for treatment. The CivaSheet is an FDA-cleared implantable sheet with a matrix of unidirectional planar low-dose-rate (LDR) Palladium-103 (Pd-103) sources. The sources are shielded on one side with gold to spare radio-sensitive structures such as the bowel. The sheet can easily be customized and implanted at the time of surgery when there is concern for close or positive margins. The CivaSheet provides an interesting solution to target the region of close/positive margins after pancreatectomy. Here we discuss the physical properties, the dosimetry, clinical workflow and early patient outcomes with the CivaSheet in pancreatic cancer.


Subject(s)
Brachytherapy , Pancreatic Neoplasms , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brachytherapy/methods , Humans , Neoadjuvant Therapy , Palladium/therapeutic use , Pancreatic Neoplasms/radiotherapy , Pancreatic Neoplasms/surgery , Radioisotopes/therapeutic use
15.
J Asthma ; 59(8): 1627-1637, 2022 08.
Article in English | MEDLINE | ID: mdl-34346266

ABSTRACT

OBJECTIVE: PALLADIUM and IRIDIUM studies demonstrated efficacy and safety of indacaterol/mometasone (IND/MF) versus salmeterol/fluticasone (SAL/FLU). This post hoc analysis of pooled data from PALLADIUM and IRIDIUM studies evaluated efficacy and safety of IND/MF versus SAL/FLU in Asian patients with inadequately controlled asthma. METHODS: Both studies were Phase III, 52-week, randomized, double-blind, active-controlled that included patients with predicted pre-bronchodilator FEV1 (PALLADIUM, ≥50%-<85%; IRIDIUM, <80%), ACQ-7 score ≥1.5. Patients treated with IND/MF high- (150/320 µg) or medium-dose (150/160 µg) or SAL/FLU high-dose (50/500 µg) were included. Lung function, asthma control, and asthma exacerbations were evaluated. RESULTS: In total, 323 patients (IND/MF high-dose, n = 107; IND/MF medium-dose, n = 106, SAL/FLU high-dose, n = 110) were included. IND/MF high-dose showed improvement in trough FEV1 versus SAL/FLU high-dose at Weeks 26 (Δ, 42 mL; 95% CI, -35 to 120 mL), and 52 (Δ, 41 mL; 95% CI, -37 to 120 mL). IND/MF high-dose exhibited numerically greater improvement in ACQ-7 score versus SAL/FLU high-dose at Weeks 26 (Δ, -0.215; 95% CI, -0.385 to -0.044) and 52 (Δ, -0.176; 95% CI, -0.348 to -0.005). Improvements in trough FEV1 and ACQ-7 score were comparable between IND/MF medium-dose and SAL/FLU high-dose. IND/MF high- and medium-dose showed reductions in moderate or severe (45%; 30%), severe (39%; 41%), and all (9%; 25%) exacerbations, respectively, versus SAL/FLU high-dose over 52 weeks. All treatments were well tolerated. CONCLUSIONS: Once-daily, single-inhaler IND/MF high-dose improved lung function with better asthma control, reduced asthma exacerbations with comparable safety versus twice-daily SAL/FLU high-dose. IND/MF medium-dose showed comparable outcomes to SAL/FLU high-dose at a reduced steroid dose.


Subject(s)
Asthma , Palladium , Administration, Inhalation , Bronchodilator Agents , Double-Blind Method , Drug Combinations , Fluticasone-Salmeterol Drug Combination , Forced Expiratory Volume , Humans , Indans , Iridium/pharmacology , Iridium/therapeutic use , Mometasone Furoate/therapeutic use , Nebulizers and Vaporizers , Palladium/pharmacology , Palladium/therapeutic use , Quinolones , Treatment Outcome
16.
Biomater Sci ; 10(1): 216-226, 2021 Dec 21.
Article in English | MEDLINE | ID: mdl-34843611

ABSTRACT

Multifunctional phototherapy nanoagents for imaging-guided synergistic photothermal therapy (PTT) and photodynamic therapy (PDT) are highly desirable in the field of solid tumor therapy. Nevertheless, the tumor microenvironment (TME) inherently associated with hypoxia significantly hampers the photodynamic effect of these multifunctional nanoagents. Herein, Pd nanocubes coated with an ultrathin Pt shell were prepared and further conjugated with fluorescein labeled and thiol functionalized polyethylene glycol (FITC-PEG-SH) (denoted as Pd@Pt-PEG). The deposition of a Pt shell on Pd nanocubes not only enhances the photothermal performance, exhibiting excellent hyperthermia outcomes and impressive photothermal (PT) imaging quality, but also leads to the formation of singlet oxygen (1O2) induced by plasmonic excitation. In the meantime, the catalytic activity of the Pt layer is enhanced by electronic coupling and the plasmonic effect, which induces the decomposition of endogenous overexpressed hydrogen peroxide (H2O2) in tumors to generate O2 for conquering TME and augmenting 1O2 generation for efficacious tumor cell apoptosis. The modification of FITC-PEG-SH improves the biocompatibility and provides outstanding fluorescence (FL) imaging properties. Upon NIR laser irradiation, Pd@Pt-PEG allows in situ O2 generation and dual-mode imaging-guided synergistic PTT/PDT that effectively kills hypoxic tumor cells, which makes it a promising nanotherapeutic agent for enhanced tumor therapy.


Subject(s)
Hyperthermia, Induced , Neoplasms , Photochemotherapy , Cell Line, Tumor , Humans , Hydrogen Peroxide , Hypoxia , Neoplasms/drug therapy , Palladium/therapeutic use , Photosensitizing Agents/therapeutic use , Photothermal Therapy , Surface Plasmon Resonance , Tumor Microenvironment
17.
J Nanobiotechnology ; 19(1): 321, 2021 Oct 14.
Article in English | MEDLINE | ID: mdl-34649589

ABSTRACT

BACKGROUND: Photothermal therapy (PTT) frequently cause thermal resistance in tumor cells by inducing the heat shock response, limiting its therapeutic effect. Hydrogen sulfide (H2S) with appropriate concentration can reverse the Warburg effect in cancer cells. The combination of PTT with H2S gas therapy is expected to achieve synergistic tumor treatment. METHODS: Here, sulourea (Su) is developed as a thermosensitive/hydrolysable H2S donor to be loaded into Pd nanocubes through in-depth coordination for construction of the Pd-Su nanomedicine for the first time to achieve photo-controlled H2S release, realizing the effective combination of photothermal therapy and H2S gas therapy. RESULTS: The Pd-Su nanomedicine shows a high Su loading capacity (85 mg g-1), a high near-infrared (NIR) photothermal conversion efficiency (69.4%), and NIR-controlled H2S release by the photothermal-triggered hydrolysis of Su. The combination of photothermal heating and H2S produces a strong synergetic effect by H2S-induced inhibition of heat shock response, thereby effectively inhibiting tumor growth. Moreover, high intratumoral accumulation of the Pd-Su nanomedicine after intravenous injection also enables photothermal/photoacoustic dual-mode imaging-guided tumor treatment. CONCLUSIONS: The proposed NIR-responsive heat/H2S release strategy provides a new approach for effective cancer therapy.


Subject(s)
Hydrogen Sulfide/chemistry , Metal Nanoparticles/chemistry , Nanomedicine/methods , Palladium/chemistry , Photothermal Therapy/methods , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Female , Hydrogen Sulfide/therapeutic use , Infrared Rays , Metal Nanoparticles/therapeutic use , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Palladium/therapeutic use
18.
ACS Appl Mater Interfaces ; 13(24): 27963-27971, 2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34110773

ABSTRACT

1T-phase transition-metal dichalcogenides (TMDs) nanomaterials are one type of emerging and promising near-infrared II (NIR-II) photothermal agents (PTAs) derived from their distinct metallic electronic structure, but it is still challenging to synthesize these nanomaterials. Herein, PdTe2 nanoparticles (PTNs) with a 1T crystal symmetry and around 50 nm in size are prepared by an electrochemical exfoliation method, and the corresponding photothermal performances irradiated under a NIR-II laser have been explored. The encapsulation of 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-poly(ethylene glycol) (DSPE-PEG) endows PTNs with water solubility, enhanced photothermal stability, and high biocompatibility. Notably, PTN/DSPE-PEG displays a potent absorbance through the NIR-II zone and considerable photothermal conversion efficiency, which is up to 68% when irradiated with a 1060 nm laser. With these unique photothermal properties, excellent in vitro and in vivo tumor inhibition effects of PTN/DSPE-PEG have been achieved under the irradiation of a NIR-II (1060 nm) laser without visible toxicity to normal tissues, suggesting that it is an efficient NIR-II photothermal nanoagent.


Subject(s)
Antineoplastic Agents/therapeutic use , Metal Nanoparticles/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/radiation effects , Antineoplastic Agents/toxicity , Cell Line, Tumor , Female , Hydrophobic and Hydrophilic Interactions , Infrared Rays , Metal Nanoparticles/chemistry , Metal Nanoparticles/radiation effects , Metal Nanoparticles/toxicity , Mice, Inbred BALB C , Palladium/chemistry , Palladium/radiation effects , Palladium/therapeutic use , Palladium/toxicity , Phosphatidylethanolamines/chemistry , Photothermal Therapy , Polyethylene Glycols/chemistry , Tellurium/chemistry , Tellurium/radiation effects , Tellurium/therapeutic use , Tellurium/toxicity
19.
Biochem Pharmacol ; 190: 114598, 2021 08.
Article in English | MEDLINE | ID: mdl-33979647

ABSTRACT

In women globally, breast cancer is responsible for most cancer-related deaths and thus, new effective therapeutic strategies are required to treat this malignancy. Platinum-based compounds like cisplatin are widely used to treat breast cancer, however, they come with limitations such as poor solubility, adverse effects, and drug resistance. To overcome these limitations, complexes containing other platinum group metals such as palladium have been studied and some have already entered clinical trials. Here we investigated the anti-cancer activity of a palladium complex, BTC2, in MCF-7 oestrogen receptor positive (ER+) and MDA-MB-231 triple negative (TN) human breast cancer cells as well as in a human breast cancer xenograft chick embryo model. BTC2 exhibited an average IC50 value of 0.54 µM, a desirable selectivity index of >2, inhibited the migration of ER+ and TN breast cancer cells, and displayed anti-cancer stem cell activity. We demonstrate that BTC2 induced DNA double strand breaks (increased levels of γ-H2AX) and activated the p-ATM/p-CHK2 and p-p38/MAPK pathways resulting in S- and G2/M-phase cell cycle arrests. Importantly, BTC2 sensitised breast cancer cells by triggering the intrinsic (cleaved caspase 9) and extrinsic (cleaved caspase 8) apoptotic as well as necroptotic (p-RIP3 and p-MLKL) cell death pathways and inhibiting autophagy and its pro-survival role. Furthermore, in the xenograft in vivo model, BTC2 displayed limited toxicity and arrested the tumour growth of breast cancer cells over a 9-day period in a manner comparable to that of the positive control drug, paclitaxel. BTC2 thus displayed promising anti-breast cancer activity.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Breast Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Palladium/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chick Embryo , Female , Humans , MCF-7 Cells , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Paclitaxel/chemistry , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Palladium/chemistry , Palladium/pharmacology , Xenograft Model Antitumor Assays/methods
20.
J Cancer Res Ther ; 17(1): 130-135, 2021.
Article in English | MEDLINE | ID: mdl-33723143

ABSTRACT

OBJECTIVE: Retinoblastoma is the most common cancer among children under 5 years of age. The common conventional methods for the treatment of retinoblastoma include chemotherapy and brachytherapy (BT). This study investigated the concurrent use of BT with 125I and 103Pd sources and chemotherapy with platinum-based chemotherapy drugs for retinoblastoma. MATERIALS AND METHODS: The absorbed doses in different parts of the eye were measured with and without platinum. Platinum concentrations of 5, 7.5, 10, and 15 mg/g were evaluated, and the dose enhancement factors (DEFs) were calculated for different cases. RESULTS: For the 125I source, the DEFs at the tumor apex were 1.49, 1.67, 1.81, and 1.97 at concentrations of 5, 7.5, 10, and 15 mg/g, respectively. The DEF decreased dramatically beyond the apex at 0.85 cm from tumor base and was 0.87, 0.82, 0.76, and 0.63 for the abovementioned concentrations, respectively. For the 103Pd source, the DEFs were 1.15, 1.24, 1.21, and 1.07, respectively, at the apex and 0.76, 0.65, 0.56, and 0.39, respectively, beyond the apex. CONCLUSIONS: Our results showed that the concurrent use of low-dose-rate plaque BT and platinum-based chemotherapy significantly increased the tumor-absorbed dose and decreased the absorbed dose in areas outside the tumor and the treatment time.


Subject(s)
Brachytherapy/methods , Computer Simulation , Iodine Radioisotopes/therapeutic use , Organoplatinum Compounds/administration & dosage , Palladium/therapeutic use , Radioisotopes/therapeutic use , Retinal Neoplasms/therapy , Retinoblastoma/therapy , Humans , Injections, Intra-Arterial , Monte Carlo Method , Phantoms, Imaging , Radiotherapy Dosage , Retinal Neoplasms/pathology , Retinoblastoma/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...