Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomaterials ; 308: 122570, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38636133

RESUMEN

Metallic biomaterials activate tumor ferroptosis by increasing oxidative stress, but their efficacy is severely limited in tumor microenvironment. Although interferon gamma (IFN-γ) can promote tumor ferroptosis sensitivity by inhibiting the antioxidant system and promoting lipid accumulation, this effect limited by the lack of IFN-γ accumulation in tumors. Herein, we report a near-infrared (NIR)-responsive HCuS nanocomposite (HCuS-PE@TSL-tlyp-1) that can stimulate immunogenic cell death (ICD)-mediated IFN-γ secretion through exogenous oxidative stress, thereby achieving cascaded ferrotherapy by mutually reinforcing ferroptosis and systemic immunity. Upon laser irradiation, the dissolution of the thermal coating, and the introduction of Cu ions and piperazine-erastin (PE) simultaneously induce oxidative stress by reactive oxygen species (ROS)/lipid peroxide (LPO) accumulation and deplete cystine-glutamate transporter (xCT)/GSH. The onset of oxidative stress-mediated ferroptosis is thus achieved, and ICD is triggered, significantly promoting cytotoxic T-cell (CTL) infiltration for IFN-γ secretion. Furthermore, IFN-γ induces immunogenic tumor ferroptosis by inhibiting xCT-antioxidant pathways and enhancing the ACSL4-fatty acid recruitment pathway, which further promotes sensitivity to ferroptosis in cells. These HCuS nanocomposites combined with aPD-L1 effectively in inhibiting tumor metastasis and recurrence. Importantly, these cascade ferrotherapy results broadens the application of HCuS biomaterials.


Asunto(s)
Cobre , Ferroptosis , Interferón gamma , Liposomas , Ferroptosis/efectos de los fármacos , Animales , Cobre/química , Cobre/farmacología , Interferón gamma/metabolismo , Ratones , Liposomas/química , Nanocompuestos/química , Línea Celular Tumoral , Muerte Celular Inmunogénica/efectos de los fármacos , Rayos Infrarrojos , Humanos , Estrés Oxidativo/efectos de los fármacos , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo
2.
Acta Biomater ; 160: 239-251, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36774974

RESUMEN

M2-like tumor-associated macrophages (TAMs) typically exhibit numerous tumor-promoting properties. Reducing the abundance of M2-like TAMs would shed light on the relief of immunosuppressive tumor microenvironment (TME), activation of the host immune system, infiltration of CD8+ T cells into the TME and restoring the function of the infiltrating T cells, which collectively inhibits tumor growth. Therefore, targeted depletion of M2-like TAMs can be a promising immunotherapy approach. In this study, we rationally constructed an M2-like TAMs-targeted nanoliposome, which encapsulates zoledronic acid (ZA) in the core, loads hematoporphyrin monomethyl ether (HMME, a typical sonosensitizer) in the lipid bilayer, and modifies M2pep peptide (the targeting unit) on the surface (designated as M-H@lip-ZA). Our aim is to validate the effectiveness of M-H@lip-ZA nanoliposomes to remodel TME via targeted depletion of M2-like TAMs for cancer immunotherapy. Through the M2pep peptide, M-H@lip-ZA can be efficiently delivered to M2-like TAMs. In the meantime, reactive oxygen species (ROS) resulting from sonodynamic therapy (SDT), together with inner ZA that shows high affinity and cytotoxicity to TAMs, can effectively deplete M2-like TAMs and remodel TME (normalize tumor vasculatures, strengthen intertumoral perfusion, ease tumor hypoxia, increase immune-promoting cytokines and decrease immunosuppressive cytokines). The tumor growth can be effectively inhibited. This work proposed a new paradigm for cancer immunotherapy via targeted depletion of M2-like TAMs. STATEMENT OF SIGNIFICANCE: • M2-like TAMs-targeted nanoliposome (M-H@lip-ZA) was designed and prepared. • Sonodynamic therapy (SDT), together with zoledronic acid (ZA) that shows high affinity and cytotoxicity to tumor-associated macrophages (TAMs), can effectively deplete M2-like TAMs. Subsequently, immune-promoting tumor microenvironment (TME) can be formed, which includes normalized tumor vasculatures, enhanced intertumoral perfusion, relieved tumor hypoxia, increased immune-promoting cytokines, and decreased immunosuppressive cytokines. • The targeted depletion of M2-like TAMs is a promising cancer immunotherapy approach.


Asunto(s)
Neoplasias , Macrófagos Asociados a Tumores , Humanos , Ácido Zoledrónico/farmacología , Macrófagos , Linfocitos T CD8-positivos , Microambiente Tumoral , Neoplasias/patología , Citocinas/farmacología , Péptidos/farmacología , Inmunoterapia/métodos
3.
J Nanobiotechnology ; 21(1): 30, 2023 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-36698190

RESUMEN

BACKGROUND: Combined therapy based on the effects of cascade reactions of nanoplatforms to combat specific solid tumor microenvironments is considered a cancer treatment strategy with transformative clinical value. Unfortunately, an insufficient O2 supply and the lack of a visual indication hinder further applications of most nanoplatforms for solid tumor therapy. RESULTS: A visualizable nanoplatform of liposome nanoparticles loaded with GOD, H(Gd), and PFP and grafted with the peptide tLyP-1, named tLyP-1H(Gd)-GOD@PFP, was constructed. The double-domain peptide tLyP-1 was used to specifically target and penetrate the tumor cells; then, US imaging, starvation therapy and sonodynamic therapy (SDT) were then achieved by the ultrasound (US)-activated cavitation effect under the guidance of MR/PA imaging. GOD not only deprived the glucose for starvation therapy but also produced H2O2, which in coordination with 1O2 produced by H(Gd), enable the effects of SDT to achieve a synergistic therapeutic effect. Moreover, the synergistic therapy was enhanced by O2 from PFP and low-intensity focused ultrasound (LIFU)-accelerated redox effects of the GOD. The present study demonstrated that the nanoplatform could generate a 3.3-fold increase in ROS, produce a 1.5-fold increase in the maximum rate of redox reactions and a 2.3-fold increase in the O2 supply in vitro, and achieve significant tumor inhibition in vivo. CONCLUSION: We present a visualizable nanoplatform with tumor-penetrating ability that can be unlocked by US to overcome the current treatment problems by improving the controllability of the O2 supply, which ultimately synergistically enhanced cascade therapy.


Asunto(s)
Retroalimentación Sensorial , Nanopartículas , Humanos , Peróxido de Hidrógeno , Línea Celular Tumoral , Nanopartículas/química , Péptidos , Hipoxia
4.
Small ; 19(14): e2206174, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36651135

RESUMEN

Multidrug resistance (MDR) and metastasis in cancer have become increasingly serious problems since antitumor efficiency is greatly restricted by a single therapeutic modality and the insensitive tumor microenvironment (TME). Herein, metal-phenolic network-functionalized nanoparticles (t-P@TFP NPs) are designed to realize multiple therapeutic modalities and reshape the TME from insensitive to sensitive under multimodal imaging monitoring. After a single irradiation, a near-infrared laser-activated multistage reaction occurs. t-P@TFP NPs trigger the phase transition of perfluoropentane (PFP) to release tannic acid (TA)/ferric ion (Fe3+ )-coated paclitaxel (PTX) and cause hyperthermia in the tumor region to efficiently kill cancer cells. Additionally, PTX is released after the disassembly of the TA-Fe3+ film by the abundant adenosine triphosphate (ATP) in the malignant tumor, which concurrently inhibits ATP-dependent drug efflux to improve sensitivity to chemotherapeutic agents. Furthermore, hyperthermia-induced immunogenic cell death (ICD) transforms "cold" tumors into "hot" tumors with the assistance of PD-1/PD-L1 blockade to evoke antitumor immunogenicity. This work carefully reveals the mechanisms underlying the abilities of these multifunctional NPs, providing new insights into combating the proliferation and metastasis of multidrug-resistant tumors.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Fototerapia/métodos , Paclitaxel/farmacología , Neoplasias/terapia , Sistemas de Liberación de Medicamentos/métodos , Resistencia a Múltiples Medicamentos , Metales , Línea Celular Tumoral , Microambiente Tumoral
5.
Adv Sci (Weinh) ; 10(4): e2204989, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36494092

RESUMEN

The emergence of aggregation-induced emission luminogens (AIEgens) has attracted substantial scientific attention. However, their antitumor efficacy in photodynamic therapy (PDT) is significantly restricted by the poor water solubility and limited treatment depth. Therefore, a novel AIEgens-involved therapeutic platform with good permeability and bioavailability is urgently required. Herein, supramolecular chemistry is combined with the AIEgen bis-pyrene (BP) to construct a peptide-AIEgen hybrid nanosystem (PAHN). After intravenous injection, the versatile nanoplatform not only improved the hydrophilicity of BP but also achieved stratified targeting from tumor to mitochondrial and induced mitochondrial dysfunction, thus activating caspase-3 upregulation. Then, sonodynamic therapy (SDT), an alternative modality with high tissue penetrability, is performed to evoke reactive oxygen species (ROS) generation for BP. More importantly, since the hydrophilic shell is separated from the nanosystem by the specific cleavage of caspase-3, the resulting decrease in hydrophilicity induced tight self-aggregation of PAHN residues in situ, further allowing more absorbed energy to be used for ROS generation under ultrasound irradiation and enhancing SDT efficacy. Moreover, severe oxidative stress resulting from ROS imbalance in the mitochondria initiates the immunogenic cell death process, thus evoking antitumor immunogenicity. This PAHN provides prospective ideas into AIE-involved antitumor therapy and design of peptide-AIEgens hybrids.


Asunto(s)
Fotoquimioterapia , Caspasa 3 , Especies Reactivas de Oxígeno , Estudios Prospectivos , Fotoquimioterapia/métodos , Péptidos
6.
Int J Nanomedicine ; 17: 4547-4565, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36199475

RESUMEN

Background: Sonodynamic therapy (SDT) has been regarded as a novel therapeutic modality for killing tumors. However, the hypoxic tumor microenvironment, especially deep-seated tumors distant from blood vessels, severely restricts therapeutic efficacy due to the oxygen-dependent manner of SDT. Methods: Herein, we report a novel ultrasonic cavitation effect-based therapeutic modality that is able to facilitate the hypoxia-tolerant SDT for inducing hypoxic tumor death. A tLyP-1 functionalized liposomes is fabricated, composed of hematoporphyrin monomethyl ether gadolinium as the sonosentizer and perfluoropentane (PFP) as the acoustic environment regulator. Moreover, the tLyP-1 functioned liposomes could achieve active tumor homing and effective deep-penetrating into hypoxic tumors. Upon low intensity focused ultrasound (LIFU) irradiation, the acoustic droplet vaporization effect of PFP induced fast liquid-to-gas transition and quick bubbles explosion to generate hydroxyl radicals, efficiently promoting cell death in both normoxic and hypoxic microenvironment (acting as deep-penetration nanobomb, DPNB). Results: The loading of PFP is proved to significantly enhance the therapeutic efficacy of hypoxic tumors. In particular, these DPNB can also act as ultrasound, photoacoustic, magnetic resonance, and near-infrared fluorescence tetramodal imaging agents for guiding the therapeutic process. Conclusion: This study is the first report involving that liquid-to-gas transition based SDT has the potential to combat hypoxic tumors.


Asunto(s)
Neoplasias , Terapia por Ultrasonido , Línea Celular Tumoral , Gadolinio , Humanos , Hipoxia/diagnóstico por imagen , Hipoxia/terapia , Liposomas , Neoplasias/diagnóstico por imagen , Neoplasias/patología , Neoplasias/terapia , Oxígeno , Microambiente Tumoral , Terapia por Ultrasonido/métodos
7.
Adv Healthc Mater ; 11(23): e2201399, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36165612

RESUMEN

The central cells of solid tumors are more proliferative and metastatic than the marginal cells. Therefore, more intelligent strategies for targeting cells with deep spatial distributions in solid tumors remain to be explored. In this work, a biocompatible nanotheranostic agent with a lipid membrane-coated, Fe3 O4 and perfluoropentane (PFP)-loaded, cRGD peptide (specifically targeting the integrin αvß3 receptor)-grafted, magnetic nanodroplets (MNDs) is developed. The MNDs exhibit excellent magnetothermal conversion and controllable magnetic hyperthermia (MHT) through alternating magnetic field regulation. Furthermore, MHT-mediated magnetic droplet vaporization (MDV) induces the expansion of the MNDs to transform them into ultrasonic microbubbles, increasing the permeability of tissue and the cell membrane via the ultrasound-targeted microbubble destruction (UTMD) technique and thereby promoting the deep penetration of MNDs in solid tumors. More importantly, MHT not only causes apoptotic damage by downregulating the expression of the HSP70, cyclin D1, and Bcl-2 proteins in tumor cells but also improves the response rate to T-cell-related immunotherapy by upregulating PD-L1 expression in tumor cells, thus inhibiting the growth of both primary and metastatic tumors. Overall, this work introduces a distinct application of nanoultrasonic biomedicine in cancer therapy and provides an attractive immunotherapy strategy for preventing the proliferation and metastasis of deeply distributed cells in solid tumors.


Asunto(s)
Neoplasias , Humanos , Neoplasias/terapia , Proliferación Celular , Fenómenos Magnéticos
8.
Small ; 18(15): e2106252, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35246943

RESUMEN

In thrombotic diseases, the effects of reactive oxygen species (ROS)-mediated oxidative stress as a "perpetrator" in thrombosis must be resolved. Accordingly, an insufficient understanding of thrombus therapy prompted the authors to pursue a more comprehensive and efficient antithrombotic treatment strategy. A Prussian blue (PB)-based nanodroplet system (PB-PFP@PC) is designed using PB and perfluorinated pentane (PFP) in the core, and a targeting peptide (CREKA, Cys-Arg-Glu-Lys-Ala) is attached to poly(lactic-coglycolic acid) (PLGA) as the delivery carrier shell. Upon near-infrared (NIR) laser irradiation, PB and PFP jointly achieve an unprecedented dual strategy for drug-free thrombolysis: photothermal therapy (PTT) combined with optical droplet vaporization (ODV). PB, a nanoenzyme, also regulates the vascular microenvironment via its antioxidant activity to continuously scavenge abnormally elevated ROS and correspondingly reduce inflammatory factors in the thrombus site. This study provides a demonstration of not only the potential of ODV in thrombus therapy but also the mechanism underlying PTT thrombolysis due to thermal ablation-induced fibrin network structural damage. Moreover, PB catalyzes ROS to generate oxygen (O2 ), which combines with the ODV effect, enhancing the ultrasound signal. Thus, regulation of the thrombosis microenvironment combined with specific nonpharmaceutical thrombolysis by PB nanodroplets provides a more comprehensive and efficient antithrombotic therapeutic strategy.


Asunto(s)
Nanopartículas , Trombosis , Ferrocianuros , Fibrinolíticos/farmacología , Fibrinolíticos/uso terapéutico , Humanos , Nanopartículas/química , Especies Reactivas de Oxígeno , Terapia Trombolítica , Trombosis/terapia
9.
J Nanobiotechnology ; 20(1): 80, 2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-35168608

RESUMEN

BACKGROUND: Comprehensive antitumor therapy through integrated multimodal means has drawn increasing attention owing to its high efficiency and metastasis suppression. RESULTS: We describe a synergistic triple protocol combining photothermal and sonodynamic therapy (PTT and SDT), together with immune checkpoint blockade for the inhibition of breast cancer growth and metastases in the 4T1 mouse model. PTT and SDT are synergistically augmented by a novel multimodal imaging nanoprobe integrated with cancer cell membrane-biomimetic nanoparticles (CHINPs) loaded with superparamagnetic iron oxide (SPIO) and hematoporphyrin monomethyl ether (HMME). CHINPs exhibit excellent homologous tumor targeting, and are sequentially triggered by ultrasound and near infrared (NIR) light under the guidance of magnetic resonance, photoacoustic and photothermal imaging, leading to complete in situ tumor eradication and systemic anti-tumor immune activation. Further combination of this approach with immune checkpoint blockade therapy is shown to suppress tumor metastasis. CONCLUSION: This work provides proof-of-principle for triple therapy using multimodal imaging-guided PTT/SDT based on biomimetic nanoprobes in combination with immunotherapy to eliminate tumors.


Asunto(s)
Nanopartículas , Fototerapia , Animales , Biomimética , Línea Celular Tumoral , Humanos , Inmunoterapia , Ratones
10.
J Nanobiotechnology ; 19(1): 449, 2021 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-34952587

RESUMEN

BACKGROUND: Mono-therapeutic modality has limitations in combating metastatic lesions with complications. Although emerging immunotherapy exhibits preliminary success, solid tumors are usually immunosuppressive, leading to ineffective antitumor immune responses and immunotherapeutic resistance. The rational combination of several therapeutic modalities may potentially become a new therapeutic strategy to effectively combat cancer. RESULTS: Poly lactic-co-glycolic acid (PLGA, 50 mg) nanospheres were constructed with photothermal transduction agents (PTAs)-Prussian blue (PB, 2.98 mg) encapsulated in the core and chemotherapeutic docetaxel (DTX, 4.18 mg)/ immune adjuvant-imiquimod (R837, 1.57 mg) loaded in the shell. Tumor cell membranes were further coated outside PLGA nanospheres (designated "M@P-PDR"), which acted as "Nano-targeted cells" to actively accumulate in tumor sites, and were guided/monitored by photoacoustic (PA)/ magnetic resonance (MR) imaging. Upon laser irradiation, photothermal effects were triggered. Combined with DTX, PTT induced in situ tumor eradication. Assisted by the immune adjuvant R837, the maturation rate of DCs increased by 4.34-fold compared with that of the control. In addition, DTX polarized M2-phenotype tumor-associated macrophages (TAMs) to M1-phenotype, relieving the immunosuppressive TME. The proportion of M2-TAMs decreased from 68.57% to 32.80%, and the proportion of M1-TAMs increased from 37.02% to 70.81%. Integrating the above processes, the infiltration of cytotoxic T lymphocytes (CTLs) increased from 17.33% (control) to 35.5%. Primary tumors and metastasis were significantly inhibited when treated with "Nano-targeted cells"-based cocktail therapy. CONCLUSION: "Nano-targeted cells"-based therapeutic cocktail therapy is a promising approach to promote tumor regression and counter metastasis/recurrence.


Asunto(s)
Antineoplásicos/uso terapéutico , Membrana Celular/química , Docetaxel/química , Nanopartículas/química , Neoplasias/terapia , Adyuvantes Inmunológicos/química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Docetaxel/farmacología , Docetaxel/uso terapéutico , Ferrocianuros/química , Ferrocianuros/farmacología , Ferrocianuros/uso terapéutico , Humanos , Imiquimod/química , Imiquimod/inmunología , Inmunoterapia/métodos , Rayos Infrarrojos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Desnudos , Neoplasias/diagnóstico por imagen , Imagen Óptica , Terapia Fototérmica/métodos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química
11.
Biomaterials ; 277: 121100, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34492584

RESUMEN

Ferroptosis-based nanomedicine has drawn increasing attention in antitumor therapy because of the advantages of this unconventional mode of apoptosis, but the difficulties of delivery to the tumor site and surface-to-core penetration after arrival seriously hinder further clinical transformation and application. Herein, we propose an unprecedented strategy of injecting magnetic nanodroplets (MNDs) to solve these two longstanding problems. MNDs are nanocarriers that can carry multifunctional drugs and imaging materials. MNDs can effectively accumulate in the tumor site by active tumor targeting (multifunctional drugs) and passive tumor targeting (enhanced permeability and retention effect), allowing diffusion of the MNDs from the surface to the core through mild-temperature magnetic fluid hyperthermia (MHT) under multimodal imaging guidance. Finally, the ferroptosis pathway is activated deep within the tumor site through the drug release. This approach was inspired by the ability of mild-temperature MHT to allow MNDs to quickly pass through the blood vessel-tumor barrier and deeply penetrate the tumor tissue from the surface to the core to amplify the antitumor efficacy of ferroptosis. This strategy is termed as "thermoferroptosis sensitization". Importantly, this behavior can be performed under the guidance of multimodal imaging, making the design of MNDs for cancer therapy safer and more reasonable.


Asunto(s)
Hipertermia Inducida , Nanopartículas , Línea Celular Tumoral , Fenómenos Magnéticos , Imagen Multimodal
12.
J Nanobiotechnology ; 19(1): 200, 2021 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-34225744

RESUMEN

BACKGROUND: Recent studies have demonstrated that multidrug resistance (MDR) is a critical factor in the low efficacy of cancer chemotherapy. The main mechanism of MDR arises from the overexpression of P-glycoprotein (P-gp), which actively enhances drug efflux and limits the effectiveness of chemotherapeutic agents. RESULTS: In this study, we fabricated a "combo" nanoagent equipping with triple synergistic strategies for enhancing antitumor efficacy against MDR cells. Tumor homing-penetrating peptide endows the nanosystem with targeting and penetrating capabilities in the first stage of tumor internalization. The abundant amine groups of polyethylenimine (PEI)-modified nanoparticles then trigger a proton sponge effect to promote endo/lysosomal escape, which enhances the intracellular accumulation and retention of anticancer drugs. Furthermore, copper tetrakis(4-carboxyphenyl)porphyrin (CuTCPP) encapsulated in the nanosystem, effectively scavenges endogenous glutathione (GSH) to reduce the detoxification mediated by GSH and sensitize the cancer cells to drugs, while simultaneously serving as a photoacoustic imaging (PAI) contrast agent for image visualization. Moreover, we also verify that these versatile nanoparticles in combination with PD-1/PD-L1 blockade therapy can not only activate immunological responses but also inhibit P-gp expression to obliterate primary and metastatic tumors. CONCLUSION: This work shows a significant enhancement in therapeutic efficacy against MDR cells and syngeneic tumors by using multiple MDR reversing strategies compared to an equivalent dose of free paclitaxel.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Inhibidores de Puntos de Control Inmunológico/farmacología , Nanopartículas/uso terapéutico , Animales , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Cobre , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos , Quimioterapia , Femenino , Compuestos Heterocíclicos , Humanos , Lisosomas , Células MCF-7 , Metaloporfirinas , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanomedicina , Nanopartículas/química , Compuestos Organofosforados , Paclitaxel/farmacología
13.
J Thorac Dis ; 11(9): 3853-3863, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31656658

RESUMEN

BACKGROUND: The amplitude spectrum area (AMSA), a frequency-domain ventricular fibrillation (VF) waveform metric, can predict successful defibrillation and the return of spontaneous circulation (ROSC) after defibrillation attempts. We aimed to investigate the validation of Spectral Energy for the quantitative analysis of the VF waveform to guide defibrillation in a porcine model of cardiac arrest and compare it with the AMSA metric. In addition, we sought to determine the effects of epinephrine and cardiopulmonary resuscitation (CPR) on AMSA and Spectral Energy. METHODS: Sixty male domestic pigs weighing 35 to 45 kg were involved in this study. VF was initially untreated for 10 min followed by 6 min of CPR. Epinephrine was administered to the animals after 2 min of CPR. After the CPR, a single 120-J biphasic shock was applied to the animals. AMSA and Spectral Energy values were measured every minute from the electrocardiogram (ECG) to defibrillation. Receiver operating characteristic (ROC) curves were calculated for both the Spectral Energy and AMSA methods. RESULTS: Spectral Energy and AMSA values gradually decayed during untreated VF in all the animals. However, after the application of CPR and epinephrine, Spectral Energy and AMSA values were significantly increased in animals which were later successfully defibrillated, but did not increase in animals in which defibrillation was unsuccessful. The ROC curves showed that the Spectral Energy and AMSA methods possessed similar levels of sensitivity and specificity in predicting defibrillation success (P<0.001). CONCLUSIONS: Both the Spectral Energy and AMSA methods accurately predict successful defibrillation. Moreover, increases in the value of either Spectral Energy or AMSA after application of CPR and epinephrine may also predict successful defibrillation.

14.
Am J Emerg Med ; 37(7): 1224-1229, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30194021

RESUMEN

PURPOSE: A novel amplitude screening method, termed Optimal Amplitude Spectrum Area (Opt-AMSA) with the aim of improving the performance of the Amplitude Spectrum Area (AMSA) method, was proposed to optimize the timing of defibrillation. We investigated the effects of the Opt-AMSA method on the prediction of successful defibrillation when compared with AMSA in a porcine model of ventricular fibrillation (VF). METHOD: 60 male domestic pigs were untreated in the first 10 min of VF, then received cardiopulmonary resuscitation (CPR) for 6 min. Values of Opt-AMSA and AMSA were calculated every minute before defibrillation. Linear regression was used to evaluate the correlation between Opt-AMSA and AMSA. Receiver Operating Characteristic (ROC) analysis was conducted for the two methods and to compare their predictive values. RESULTS: The values of both AMSA and Opt-AMSA gradually decreased over time during untreated VF in all animals. The values of both methods of defibrillation were slightly increased after the implementation of CPR in animals that were successfully resuscitated, while there were no significant changes in either method in those who ultimately failed to resuscitate. The significant positive correlation between Opt-AMSA and AMSA was shown by Pearson correlation analysis. ROC analysis showed that Opt-AMSA (AUC = 0.87) significantly improved the performance of AMSA (AUC = 0.77) to predict successful defibrillation (Z = 2.27, P < 0.05). CONCLUSION: Both the Opt-AMSA and AMSA methods showed high potential to predict the success of defibrillation. Moreover, the Opt-AMSA method improved the performance of the AMSA method, and may be a promising tool to optimize the timing of defibrillation.


Asunto(s)
Cardioversión Eléctrica , Fibrilación Ventricular/prevención & control , Fibrilación Ventricular/fisiopatología , Animales , Reanimación Cardiopulmonar/métodos , Modelos Animales de Enfermedad , Masculino , Estudios Retrospectivos , Sensibilidad y Especificidad , Procesamiento de Señales Asistido por Computador , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...