Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
J Cancer ; 15(18): 6002-6015, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39440065

RESUMO

Background: Triple-negative breast cancer (TNBC) is a poor prognostic subtype of breast cancer due to limited treatment. Macrophage plays a critical role in tumor growth and survival. Our study intends to explore the heterogeneity of macrophage in TNBC and establish a macrophage-related prognostic model for TNBC prognostic stratification. Materials and Methods: Seurat package was conducted to analyze the single-cell RNA expression profilers. The cell types were identified by the markers derived from public research and online database. The cell-cell interactions were calculated by the CellChat package. Monocle package was used to visualize the cell trajectory of macrophages. The prognostic model was constructed by six macrophage-related genes after a series of selections. The expression of six genes were validated in normal and TNBC tissues. And several potential agents for high-risk TNBC patients were analyzed by Connectivity Map analysis. Results: Nine cell types were identified, and the macrophages were highly enriched in TNBC samples. five distinct subgroups of macrophage were identified. Notably, SPP1+ tumor-associated macrophages exhibited a poor prognosis. The prognostic model was constructed by HSPA6, LPL, IDO1, ALDH2, TK1, and QPCT with good predictive accuracy at 3-, 5- years overall survival for TNBC patients in both training and external test cohorts. Finally, several drugs were identified for the high-risk TNBC patients decided by model. Conclusion: Our study provides a valuable source for clarifying macrophage heterogeneity in TNBC, and a promising tool for prognostic risk stratification of TNBC.

3.
Oncoimmunology ; 13(1): 2416558, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39429516
4.
Methods Cell Biol ; 189: 153-168, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39393881

RESUMO

Dendritic cells (DCs), and especially so conventional type I DCs (cDC1s), are fundamental regulators of anticancer immunity, largely reflecting their superior ability to engulf tumor-derived material and process it for cross-presentation on MHC Class I molecules to CD8+ cytotoxic T lymphocytes (CTLs). Thus, investigating key DC functions including (but not limited to) phagocytic capacity, expression of CTL-activating ligands on the cell surface, and cross-presentation efficacy is an important component of multiple immuno-oncology studies. Unfortunately, DCs are terminally differentiated cells, implying that they cannot be propagated indefinitely in vitro and hence must be generated ad hoc from circulating or bone marrow-derived precursors, which presents several limitations. Here, we propose a simple, cytofluorometric method to quantify phenotypic activation markers including CD80, CD86 and MHC class II molecules on the surface of a conditionally immortalized immature DC line that can be indefinitely propagated in vitro but also driven into maturation at will with a simple change in culture conditions. Upon appropriate scaling and automatization, this approach is compatible with high-throughput screening programs for the discovery of novel DC activators that do not suffer from batch variability and other limitations associated with the generation of fresh DCs.


Assuntos
Diferenciação Celular , Células Dendríticas , Citometria de Fluxo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Citometria de Fluxo/métodos , Humanos , Animais , Fenótipo , Antígeno B7-2/metabolismo , Biomarcadores/metabolismo , Camundongos , Linhagem Celular , Antígeno B7-1/metabolismo , Linhagem Celular Transformada
5.
Oncoimmunology ; 13(1): 2412874, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39376580

RESUMO

Recent findings revealed that neoantigen-specific cytotoxic type 1 regulatory T (TR1) CD4 T cells can subvert cancer immunotherapy by killing type 1 conventional dendritic cells (cDC1s) that present tumor antigens bound to MHC class II. This underlines the importance of cDC1s for eliciting anticancer immunity but poses a novel clinical challenge.


Assuntos
Células Dendríticas , Imunoterapia , Neoplasias , Linfócitos T Reguladores , Animais , Humanos , Antígenos de Neoplasias/imunologia , Células Dendríticas/imunologia , Imunoterapia/métodos , Neoplasias/imunologia , Neoplasias/terapia , Linfócitos T Reguladores/imunologia
6.
Mol Cancer ; 23(1): 187, 2024 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-39242519

RESUMO

BACKGROUND: The plasma concentrations of acyl coenzyme A binding protein (ACBP, also known as diazepam-binding inhibitor, DBI, or 'endozepine') increase with age and obesity, two parameters that are also amongst the most important risk factors for cancer. METHODS: We measured ACBP/DBI in the plasma from cancer-free individuals, high-risk patients like the carriers of TP53 or BRCA1/2 mutations, and non-syndromic healthy subjects who later developed cancer. In mice, the neutralization of ACBP/DBI was used in models of non-small cell lung cancer (NSCLC) and breast cancer development and as a combination treatment with chemoimmunotherapy (chemotherapy + PD-1 blockade) in the context of NSCLC and sarcomas. The anticancer T cell response upon ACBP/DBI neutralization was characterized by flow cytometry and single-cell RNA sequencing. RESULTS: Circulating levels of ACBP/DBI were higher in patients with genetic cancer predisposition (BRCA1/2 or TP53 germline mutations) than in matched controls. In non-syndromic cases, high ACBP/DBI levels were predictive of future cancer development, and especially elevated in patients who later developed lung cancer. In preclinical models, ACBP/DBI neutralization slowed down breast cancer and NSCLC development and enhanced the efficacy of chemoimmunotherapy in NSCLC and sarcoma models. When combined with chemoimmunotherapy, the neutralizing monoclonal antibody against ACBP/DBI reduced the frequency of regulatory T cells in the tumor bed, modulated the immune checkpoint profile, and increased activation markers. CONCLUSION: These findings suggest that ACBP/DBI acts as an endogenous immune suppressor. We conclude that elevation of ACBP/DBI constitutes a risk factor for the development of cancer and that ACBP/DBI is an actionable target for improving cancer immunosurveillance.


Assuntos
Biomarcadores Tumorais , Animais , Feminino , Humanos , Camundongos , Neoplasias da Mama/imunologia , Neoplasias da Mama/diagnóstico , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/diagnóstico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Linhagem Celular Tumoral , Vigilância Imunológica , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/metabolismo , Neoplasias/diagnóstico , Neoplasias/imunologia , Neoplasias/etiologia , Fatores de Risco
7.
Eur Radiol ; 2024 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-39214892

RESUMO

OBJECTIVES: Implementing personalization protocol in clinical routine necessitates diverse low-dose PET/CT scan protocols. This study explores the clinical feasibility of one-third (1/3) dose regimen and evaluates the diagnostic image quality and lesion detectability of BMI-based 1/3-injection doses for 2-[18F]FDG PET/CT imaging. METHODS: Seventy-four cancer patients underwent total-body 2-[18F]FDG PET/CT examination, with 37 retrospectively enrolled as full-dose group (3.7 MBq/kg) and 37 prospectively enrolled as the 1/3-dose group (1.23 MBq/kg). The 1/3-dose group was stratified by BMI, with an acquisition time of 5 min (G5), 6 min (G6), and 8 min (G8) for BMI < 25, 25 ≤ BMI ≤ 29, and BMI > 29, respectively. Image quality was subjectively and objectively assessed, and lesion detectability was quantitatively analyzed. RESULTS: Subjective assessments of 1/3-dose and full-dose PET images showed strong agreement among readers (κ > 0.88). In the 1/3-dose group, the Likert scores were above 4. G5, G6, and G8 showed comparable image quality, with G5 demonstrating higher lesion conspicuity than G6 and G8 (p = 0.045). Objective evaluation showed no significant differences in SUVmax, liver SUVmean and TBR between 1/3- and full-dose groups (p > 0.05). No statistical differences were observed in the SUVmax of primary tumor, SUVmean of liver and TBR across all BMI categories between the 1/3-dose and full-dose groups. Lesion detection rates showed no significant difference between the 1/3-dose (93.24%, 193/207) and full-dose groups (94.73%, 198/209) (p = 0.520). CONCLUSION: A BMI-stratified 1/3-dose regimen is a feasible low-dose alternative with clinically acceptable lesion detectability equivalent to full-dose protocol, potentially expanding the applicability of personalized protocols. CLINICAL RELEVANCE STATEMENT: This study demonstrated that BMI-stratified 1/3-dose regimens for [18F]FDG total-body PET/CT yielded equivalent outputs compared to the full-dose regimen, which aligns with clinical needs for personalization in dose and BMI. KEY POINTS: Currently, limited personalized low-dose total-body PET/CT protocols are available, particularly for patients with varied BMI. Reducing the radiotracer dose to 1/3 the standard demonstrated comparable image quality and lesion detectability equivalent to full dose. BMI-stratified 1/3-dose regimen is a clinically feasible low-dose alternative.

8.
Oncoimmunology ; 13(1): 2360275, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38812570
9.
Oncoimmunology ; 13(1): 2338951, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38590800

RESUMO

Recently, we showed that an autologous DC-based vaccine induces an increase in immunosuppressive PD-L1+ tumor-associated macrophages (TAM) both in the tumor and the tumor draining lymph nodes, thereby blunting the efficacy of therapeutic immunization. Only the combination of the DC vaccine with anti-PD-L1 immune checkpoint inhibition, but not the use of antibodies targeting PD-1 alone, was able to set off CD8+ cytotoxic T lymphocyte (CTL)-mediated tumor suppression in mice. In sum, we delineated a PD-L1 checkpoint blockade-based strategy to avoid TAM-induced T cell exhaustion during DC vaccine therapy.


Assuntos
Antígeno B7-H1 , Vacinas , Animais , Camundongos , Linfócitos T Citotóxicos , Linfócitos T CD8-Positivos , Macrófagos
10.
Int Urol Nephrol ; 56(8): 2779-2791, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38530583

RESUMO

PURPOSE: Diabetic bladder fibrosis is a common comorbidity. Altered expression of some long non-coding RNAs (LncRNAs) has been associated with bladder fibrosis. LncRNA H19 has been reported to regulate bladder cancer through miR-29b. However, the action mechanism of LncRNA H19 in bladder fibrosis is unclear. METHODS: In vitro, human bladder smooth muscle cells (HBSMCs) were cultured with transforming growth factor-ß1 (TGF-ß1) for 48 h to construct cell model of bladder fibrosis. HBSMCs were then transfected with si-LncRNA H19, si-NC, miR-29b-mimic, mimic-NC, or miR-29b-inhibitor. In vivo, Sprague-Dawley (SD) rats were given a high-sucrose-high-fat (HSHF) diet for 4 weeks and injected with streptozotocin (STZ, 50 mg/kg) to induce bladder fibrosis model in diabetic rats, followed by injection of lentiviral particles knocking down LncRNA H19 expression, empty vector, or miR-29b-inhibitor, respectively. RESULTS: LncRNA H19 was up-regulated in TGF-ß1-induced HBSMC fibrosis and STZ-induced diabetic rat bladder fibrosis, whereas miR-29b was down-regulated. si-LncRNA H19 reduced blood glucose levels and improved histopathological damage of bladder tissue in rats. In addition, si-LncRNA H19 or miR-29b-mimic increased the expression of E-cadherin, but decreased the expression of N-cadherin, vimentin, fibronectin (FN) in bladder tissues, and HBSMCs. si-LncRNA H19 reduced TGF-ß1/p-drosophila mothers against decapentaplegic 3 (Smad3) protein in HBSMCs and in rat bladder tissues, while miR-29b-inhibitor reversed the effect of si-LncRNA H19. CONCLUSION: This study indicated that LncRNA H19 may inhibit bladder fibrosis in diabetic rats by targeting miR-29b via the TGF-ß1/Smad3 signalling pathway.


Assuntos
Diabetes Mellitus Experimental , Fibrose , MicroRNAs , RNA Longo não Codificante , Ratos Sprague-Dawley , Transdução de Sinais , Fator de Crescimento Transformador beta1 , Animais , RNA Longo não Codificante/metabolismo , RNA Longo não Codificante/genética , MicroRNAs/metabolismo , MicroRNAs/genética , Ratos , Fator de Crescimento Transformador beta1/metabolismo , Diabetes Mellitus Experimental/complicações , Humanos , Células Cultivadas , Bexiga Urinária/patologia , Bexiga Urinária/metabolismo , Masculino , Doenças da Bexiga Urinária/metabolismo , Doenças da Bexiga Urinária/etiologia , Proteína Smad3/metabolismo
11.
Cell Rep Med ; 5(1): 101377, 2024 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-38232703

RESUMO

Current immunotherapies provide limited benefits against T cell-depleted tumors, calling for therapeutic innovation. Using multi-omics integration of cancer patient data, we predict a type I interferon (IFN) responseHIGH state of dendritic cell (DC) vaccines, with efficacious clinical impact. However, preclinical DC vaccines recapitulating this state by combining immunogenic cancer cell death with induction of type I IFN responses fail to regress mouse tumors lacking T cell infiltrates. Here, in lymph nodes (LNs), instead of activating CD4+/CD8+ T cells, DCs stimulate immunosuppressive programmed death-ligand 1-positive (PD-L1+) LN-associated macrophages (LAMs). Moreover, DC vaccines also stimulate PD-L1+ tumor-associated macrophages (TAMs). This creates two anatomically distinct niches of PD-L1+ macrophages that suppress CD8+ T cells. Accordingly, a combination of PD-L1 blockade with DC vaccines achieves significant tumor regression by depleting PD-L1+ macrophages, suppressing myeloid inflammation, and de-inhibiting effector/stem-like memory T cells. Importantly, clinical DC vaccines also potentiate T cell-suppressive PD-L1+ TAMs in glioblastoma patients. We propose that a multimodal immunotherapy and vaccination regimen is mandatory to overcome T cell-depleted tumors.


Assuntos
Glioblastoma , Vacinas , Humanos , Animais , Camundongos , Linfócitos T CD8-Positivos , Antígeno B7-H1 , Macrófagos , Células Dendríticas , Linfonodos/metabolismo , Vacinas/metabolismo
12.
Genes Immun ; 25(4): 348-350, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38267541

RESUMO

Recently we developed a dendritic cell (DC) genotype-phenotype screening platform that is based on CRISPR/Cas9-mediated gene editing of immortalized DC precursors. Whole genome screening for gain-of-function phenotypes led to the identification of BCL2 as a DC-specific immune checkpoint. Genetic or pharmacological inhibition of BCL2 similarly enhanced the antigen presentation capacity of conventional type-1 dendritic cells (cDC1) and mediated T cell-dependent anticancer immunity. The therapeutic anticancer efficacy of the BCL2 inhibitor venetoclax in mice was further increased when combined with a PD-1-targeted immune checkpoint inhibitor. In sum, we delineated a novel strategy of dual checkpoint blockade for cancer immunotherapy in which improvement of DC antigen presentation and avoidance of T cell exhaustion can be advantageously combined.


Assuntos
Células Dendríticas , Imunoterapia , Proteínas Proto-Oncogênicas c-bcl-2 , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Animais , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Humanos , Imunoterapia/métodos , Camundongos , Neoplasias/imunologia , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Apresentação de Antígeno , Sulfonamidas/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes
13.
J Laparoendosc Adv Surg Tech A ; 34(4): 313-317, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38294894

RESUMO

Background: Ureteral polyps are rare benign ureteral tumor. No guideline recommends that open or minimally invasive surgery is best for treating ureteral polyps. This article aims to provide a comprehensive review of the minimally invasive techniques currently available for treating ureteral polyps. Materials and Methods: We performed a comprehensive search of articles published in PubMed, using the keywords "ureteral" and "polyp," or "polyps." Results: A total of 275 studies were obtained from the literature search but 96 articles were excluded. Conclusions: Several minimally invasive approaches were developed with the advancement of medical technology, including endoscopic, laparoscopic, and robotic approaches; however, the best surgical technique was yet to be decided. Due to the advantages and disadvantages of these approaches, the best surgical approach should be tailored to each patient's needs and the surgeon's preferences and experience.


Assuntos
Pólipos , Humanos , Pólipos/cirurgia , Procedimentos Cirúrgicos Minimamente Invasivos/métodos , Procedimentos Cirúrgicos Robóticos/métodos , Laparoscopia/métodos , Doenças Ureterais/cirurgia , Ureteroscopia/métodos , Neoplasias Ureterais/cirurgia
15.
Immunol Rev ; 321(1): 7-19, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37596984

RESUMO

The search for immunostimulatory drugs applicable to cancer immunotherapy may profit from target-agnostic methods in which agents are screened for their functional impact on immune cells cultured in vitro without any preconceived idea on their mode of action. We have built a synthetic mini-immune system in which stressed and dying cancer cells (derived from standardized cell lines) are confronted with dendritic cells (DCs, derived from immortalized precursors) and CD8+ T-cell hybridoma cells expressing a defined T-cell receptor. Using this system, we can identify three types of immunostimulatory drugs: (i) pharmacological agents that stimulate immunogenic cell death (ICD) of malignant cells; (ii) drugs that act on DCs to enhance their response to ICD; and (iii) drugs that act on T cells to increase their effector function. Here, we focus on strategies to develop drugs that enhance the perception of ICD by DCs and to which we refer as "ICD enhancers." We discuss examples of ICD enhancers, including ligands of pattern recognition receptors (exemplified by TLR3 ligands that correct the deficient function of DCs lacking FPR1) and immunometabolic modifiers (exemplified by hexokinase-2 inhibitors), as well as methods for target deconvolution applicable to the mechanistic characterization of ICD enhancers.


Assuntos
Linfócitos T CD8-Positivos , Morte Celular Imunogênica , Humanos , Imunoterapia , Células Dendríticas , Percepção
16.
Semin Cell Dev Biol ; 156: 11-21, 2024 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-37977108

RESUMO

The successful treatment of oncological malignancies which results in long-term disease control or the complete eradication of cancerous cells necessitates the onset of adaptive immune responses targeting tumor-specific antigens. Such desirable anticancer immunity can be triggered via the induction of immunogenic cell death (ICD) of cancer cells, thus converting malignant cells into an in situ vaccine that elicits T cell mediated adaptive immune responses and establishes durable immunological memory. The exploration of ICD for cancer treatment has been subject to extensive research. However, functional heterogeneity among ICD activating therapies in many cases requires specific co-medications to achieve full-blown efficacy. Here, we described the hallmarks of ICD and classify ICD activators into three distinct functional categories namely, according to their mode of action: (i) ICD inducers, which increase the immunogenicity of malignant cells, (ii) ICD sensitizers, which prime cellular circuitries for ICD induction by conventional cytotoxic agents, and (iii) ICD enhancers, which improve the perception of ICD signals by antigen presenting dendritic cells. Altogether, ICD induction, sensitization and enhancement offer the possibility to convert well-established conventional anticancer therapies into immunotherapeutic approaches that activate T cell-mediated anticancer immunity.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Neoplasias/patologia , Antineoplásicos/farmacologia , Morte Celular , Antígenos de Neoplasias , Linfócitos T
17.
Biol Direct ; 18(1): 71, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37907944

RESUMO

Cancer immunotherapy, alone or in combination with conventional therapies, has revolutionized the landscape of antineoplastic treatments, with dendritic cells (DC) emerging as key orchestrators of anti-tumor immune responses. Among the distinct DC subsets, conventional type 1 dendritic cells (cDC1) have gained prominence due to their unique ability to cross-present antigens and activate cytotoxic T lymphocytes. This review summarizes the distinctive characteristics of cDC1, their pivotal role in anticancer immunity, and the potential applications of cDC1-based strategies in immunotherapy.


Assuntos
Células Dendríticas , Neoplasias , Humanos , Células Dendríticas/patologia , Neoplasias/terapia , Neoplasias/patologia , Imunoterapia , Linfócitos T CD8-Positivos/patologia
18.
Cell Death Differ ; 30(12): 2447-2451, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37845384

RESUMO

BCL2 is an apoptosis-inhibitory oncoprotein that also possesses apoptosis-unrelated activities. Pharmacological BCL2 inhibitors have been developed with the scope of driving BCL2-dependent cancer cells into apoptosis, and one BCL2 antagonist, venetoclax, has been clinically approved for the treatment of specific leukemias and lymphomas. Nonetheless, it appears that venetoclax, as well as genetic BCL2 inhibition, can mediate anticancer effects through an indirect action. Such an indirect effect relies on the enhancement of the immunostimulatory function of dendritic cells, hence increasing tumor immunosurveillance. Mechanistically, BCL2 inhibition involves improved antigen presentation by conventional type-1 dendritic cells (cDC1s) due to the activation of an interferon response, leading to a T cell-mediated anticancer immune response that can be further enhanced by PD-1 blockade. These findings support the emerging hypothesis that successful antineoplastic drugs generally mediate their effects indirectly, through the immune system, rather via merely cell-autonomous effects on malignant cells.


Assuntos
Antineoplásicos , Proteínas Proto-Oncogênicas c-bcl-2 , Proteínas Proto-Oncogênicas c-bcl-2/genética , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/uso terapêutico , Células Dendríticas , Linhagem Celular Tumoral
19.
Polymers (Basel) ; 15(17)2023 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-37688132

RESUMO

In this study, graphene quantum dots (GQDs) with a diameter of ~3 nm were successfully synthesized and incorporated into a poly(ethylene terephthalate) (PET) matrix to fabricate PET/GQDs nanocomposites. The impact of GQDs on the crystallization and thermal stability of the PET/GQDs nanocomposites was investigated. It was observed that the addition of only 0.5 wt% GQDs into the nanocomposites resulted in a significant increase in the crystallization temperature (peak temperature) of PET, from 194.3 °C to 206.0 °C during the cooling scan process. This suggested that an optimal concentration of GQDs could function as a nucleating agent and effectively enhance the crystallization temperature of PET. The isothermal crystallization method was employed to analyze the crystallization kinetics of the PET/GQDs nanocomposites, and the data showed that 0.5 wt% GQDs significantly accelerated the crystallization rate. Furthermore, the incorporation of GQDs into the PET matrix imparted photoluminescent properties to the resulting PET/GQDs nanocomposites. The PET crystals with GQDs as crystal nuclei and the crazes caused by defects played a vital role in isolating and suppressing the concentration quenching of GQDs. This effect facilitated the detection of defects in PET.

20.
Medicine (Baltimore) ; 102(34): e35031, 2023 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-37653733

RESUMO

To compare the efficacy, safety, and complications of transurethral 1470 nm diode laser vaporization and plasma kinetic enucleation of prostate (PKEP) in benign prostatic hyperplasia treatment. A retrospective matched-paired comparison of patients treated using transurethral 1470 nm diode laser vaporization (n = 40) or PKEP (n = 40) was conducted. Baseline characteristics, preoperative data, and postoperative outcomes at the 24-month follow-up of the patients were recorded. The present study found no significant preoperative differences between the 2 treatment groups. Compared with PKEP, 1470 nm diode laser vaporization had a significantly shorter operation time and less intraoperative blood loss, but there were no marked differences between the 2 groups in terms of postoperative bladder irrigation time, catheterization time, and hospital stay. Moreover, at the 24-month follow-up postoperatively, there were no marked differences in the International Prostatic Symptomatic Score (IPSS), quality of life (QOL), maximum urinary flow rate (Qmax), and post-void residual urine volume (PVR) between the 2 groups. IPSS, QOL, Qmax, and PVR had improved significantly compared to preoperative assessment at 24-month follow-up in both groups and there was no significant difference in the variation of IPSS, QOL, Qmax and PVR before and after the operation. Furthermore, complications were comparable between the 2 treatment groups. Transurethral 1470 nm diode laser vaporization and PKEP are effective strategies in the treatment of benign prostatic hyperplasia. However, 1470 nm diode laser vaporization offers advantages over PKEP in terms of shortening operation time and reducing intraoperative bleeding. Nonetheless, further research with a larger number of patients and long-term follow-up is necessary to confirm and validate these findings.


Assuntos
Terapia a Laser , Hiperplasia Prostática , Masculino , Humanos , Estudos Retrospectivos , Próstata/cirurgia , Hiperplasia Prostática/cirurgia , Qualidade de Vida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA