Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Front Oncol ; 14: 1356000, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38496758

RESUMO

Penile schwannoma is an uncommonly seen peripheral nerve tumor, of which penile plexiform schwannomas (PS) is extremely rare that has only been reported in several adults. We present a case of penile PS with a similar lesion in inguinal region in a 9-year-old child, which appeared as painless masses and rapidly growing within one year. Penile ultrasonography suggested well-defined lesions with limited vascularity. Both masses presented with low-to-intermediated signal intensity and no definite enhancement in computed tomography. The lesions were completely resected with minimal intraoperative bleeding, and a diagnose of benign PS was confirmed based on H&E staining and positive S-100 expression in immunohistochemistry. There was no evidence of tumor recurrence or metastasis after 6 months of follow-up. Only 6 cases of penile schwannoma in children were recorded, of which 5 were malignant, and none was PS. The malignancy rate of penile schwannoma in children may be overestimated due to delayed diagnose of benign ones. A rapidly growing penile mass with a suspected metastatic lesion in inguinal region could be easily misdiagnosed as malignant. This case report and literature review is expected to assist clinicians in getting a comprehensive understanding of children penile schwannomas and choosing the best management strategy when faced with this rare condition.

2.
Antioxidants (Basel) ; 13(2)2024 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-38397821

RESUMO

Candida albicans, a prominent opportunistic pathogenic fungus in the human population, possesses the capacity to induce life-threatening invasive candidiasis in individuals with compromised immune systems despite the existence of antifungal medications. When faced with macrophages or neutrophils, C. albicans demonstrates its capability to endure oxidative stress through the utilization of antioxidant enzymes. Therefore, the enhancement of oxidative stress in innate immune cells against C. albicans presents a promising therapeutic approach for the treatment of invasive candidiasis. In this study, we conducted a comprehensive analysis of a library of drugs approved by the Food and Drug Administration (FDA). We discovered that halofantrine hydrochloride (HAL) can augment the antifungal properties of oxidative damage agents (plumbagin, menadione, and H2O2) by suppressing the response of C. albicans to reactive oxygen species (ROS). Furthermore, our investigation revealed that the inhibitory mechanism of HAL on the oxidative response is dependent on Cap1. In addition, the antifungal activity of HAL has been observed in the Galleria mellonella infection model. These findings provide evidence that targeting the oxidative stress response of C. albicans and augmenting the fungicidal capacity of oxidative damage agents hold promise as effective antifungal strategies.

3.
BMC Cancer ; 24(1): 57, 2024 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-38200410

RESUMO

BACKGROUND: Anus preservation has been a challenge in the treatment of patients with low rectal adenocarcinoma (within 5 cm from the anal verge) because it is difficult to spare the anus with its functioning sphincter complex under the safe margin of tumour resection. Patients with dMMR/MSI-H can achieve a favourable complete response (CR) rate by using a single immune checkpoint inhibitor. For patients with pMMR/MSS/MSI-L, intensified neoadjuvant three-drug chemotherapy may be the preferred option for anal preservation. In addition, the watch and wait (W&W) strategy has been proven safe and feasible for patients with rectal cancer who achieve a clinical complete response (cCR). Therefore, we initiated this clinical trial to explore the optimal neoadjuvant treatment pattern for patients with low locally advanced rectal cancer (LARC) with different MMR/MSI statuses, aiming to achieve a higher cCR rate with the W&W strategy and ultimately provide more patients with a chance of anus preservation. METHODS: This is a randomised, controlled, open-label, multicentre phase III trial. Patients with clinical stage T2-4 and/or N + tumours located within 5 cm from the anal verge are considered eligible. Based on the results of pathological biopsy, the patients are divided into two groups: dMMR/MSI-H and pMMR/MSS. Patients in the dMMR/MSI-H group will be randomly allocated in a 1:1 ratio to either arm A (monoimmunotherapy) or arm B (short-course radiotherapy followed by monoimmunotherapy). Patients in the pMMR/MSS group will be initially treated with long-term pelvic radiation with concurrent capecitabine combined with irinotecan. Two weeks after the completion of chemoradiotherapy (CRT), the patients will be randomly allocated in a 1:1 ratio to arm C (XELIRI six cycle regime) or arm D (FOLFIRINOX nine cycle regime). The irinotecan dose will be adjusted according to the UGT1A1-genotype. After treatment, a comprehensive assessment will be performed to determine whether a cCR has been achieved. If achieved, the W&W strategy will be adopted; otherwise, total mesorectal excision (TME) will be performed. The primary endpoint is cCR with the maintenance of 12 months at least, determined using digital rectal examination, endoscopy, and rectal MRI or PET/CT as a supplementary method. DISCUSSION: APRAM will explore the best anus preservation model for low LARC, combining the strategies of consolidation chemotherapy, immunotherapy, and short-course radiotherapy, and aims to preserve the anus of more patients using W&W. Our study provides an accurate individual treatment mode based on the MMR/MSI status for patients with low LARC, and more patients will receive the opportunity for anus preservation under our therapeutic strategy, which would transform into long-term benefits. TRIAL REGISTRATION: Clinicaltrials.gov NCT05669092 (Registered 28th Nov 2022).


Assuntos
Adenocarcinoma , Neoplasias Encefálicas , Neoplasias Colorretais , Síndromes Neoplásicas Hereditárias , Neoplasias Pancreáticas , Neoplasias Retais , Humanos , Canal Anal , Protocolos de Quimioterapia Combinada Antineoplásica , Irinotecano , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Neoplasias Retais/tratamento farmacológico , Neoplasias Retais/genética , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Ensaios Clínicos Controlados Aleatórios como Assunto , Estudos Multicêntricos como Assunto , Ensaios Clínicos Fase III como Assunto
4.
PeerJ ; 11: e16631, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38144182

RESUMO

Background: The heterogeneity of colorectal cancer (CRC) is the main cause of the disparity of drug sensitivity and the variability of prognosis. Pyroptosis is closely associated with the development and prognosis of various tumors, including CRC. Dividing CRC into distinct subgroups based on pyroptosis is a worthwhile topic for improving the precision treatment and prognosis prediction of CRC. Methods: We classified patients into two clusters using the consensus clustering based on the pyroptosis-related genes (PRGs). Next, the prognostic signature was developed with LASSO regression analysis using the screened genes from differentially expressed genes (DEGs) by univariate and multivariate Cox analyses. According to the pyroptosis-related score (PR score) calculated with the signature, patients belonged to two groups with distinct prognosis. Moreover, we assessed the immune profile to explore the relationship between the signature and immunological characteristics. Two single cell sequencing databases were adopted for further exploration of tumor immune microenvironment (TME). In addition, we applied our own cohort and Drugbank to explore the correlation of the signature and clinical therapies. We also studied the expression of key genes by immunohistochemistry. Results: The signature performed well in predicting the prognosis of CRC as the high area under curve (AUC) value demonstrated. Patients with a higher PR score had poorer prognosis and higher expression of immune checkpoints but more abundant infiltration of immune cells. Combining with the indicator of therapeutic analysis, they might benefit more from immune checkpoint blockade (ICB) and neo-adjuvant chemoradiotherapy (nCRT). Conclusion: In conclusion, our study is based on genomics and transcriptomics to investigate the role of PRGs in CRC. We have established a prognostic signature and integrated single-cell data to study the relationship between the signature with the TME in CRC. Its clinical application in reliable prediction of prognosis and personalized treatment was validated by public and own sequencing cohort. It provided a new insight for the personalized treatment of CRC.


Assuntos
Neoplasias Colorretais , Piroptose , Humanos , Prognóstico , Piroptose/genética , Área Sob a Curva , Quimiorradioterapia Adjuvante , Neoplasias Colorretais/genética , Microambiente Tumoral/genética
5.
Radiat Oncol ; 18(1): 175, 2023 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-37891611

RESUMO

BACKGROUND: Accurate prediction of response to neoadjuvant chemoradiotherapy (nCRT) is very important for treatment plan decision in locally advanced rectal cancer (LARC). The aim of this study was to investigate whether self-attention mechanism based multi-sequence fusion strategy applied to multiparametric magnetic resonance imaging (MRI) based deep learning or hand-crafted radiomics model construction can improve prediction of response to nCRT in LARC. METHODS: This retrospective analysis enrolled 422 consecutive patients with LARC who received nCRT before surgery at two hospitals. All patients underwent multiparametric MRI scans with three imaging sequences. Tumor regression grade (TRG) was used to assess the response of nCRT based on the resected specimen. Patients were separated into 2 groups: poor responders (TRG 2, 3) versus good responders (TRG 0, 1). A self-attention mechanism, namely channel attention, was applied to fuse the three sequence information for deep learning and radiomics models construction. For comparison, other two models without channel attention were also constructed. All models were developed in the same hospital and validated in the other hospital. RESULTS: The deep learning model with channel attention mechanism achieved area under the curves (AUCs) of 0.898 in the internal validation cohort and 0.873 in the external validation cohort, which was the best performed model in all cohorts. More importantly, both the deep learning and radiomics model that applied channel attention mechanism performed better than those without channel attention mechanism. CONCLUSIONS: The self-attention mechanism based multi-sequence fusion strategy can improve prediction of response to nCRT in LARC.


Assuntos
Imageamento por Ressonância Magnética Multiparamétrica , Neoplasias Retais , Humanos , Neoplasias Retais/diagnóstico por imagem , Neoplasias Retais/terapia , Neoplasias Retais/patologia , Estudos Retrospectivos , Terapia Neoadjuvante/métodos , Resultado do Tratamento , Quimiorradioterapia/métodos
6.
Cancer Biol Med ; 20(4)2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-37144561

RESUMO

OBJECTIVE: The identification of biomarkers for predicting chemoradiotherapy efficacy is essential to optimize personalized treatment. This study determined the effects of genetic variations in genes involved in apoptosis, pyroptosis, and ferroptosis on the prognosis of patients with locally advanced rectal cancer receiving postoperative chemoradiotherapy (CRT). METHODS: The Sequenom MassARRAY was used to detect 217 genetic variations in 40 genes from 300 patients with rectal cancer who received postoperative CRT. The associations between genetic variations and overall survival (OS) were evaluated using hazard ratios (HRs) and 95% confidence intervals (CIs) computed using a Cox proportional regression model. Functional experiments were performed to determine the functions of the arachidonate 5-lipoxygenase (ALOX5) gene and the ALOX5 rs702365 variant. RESULTS: We detected 16 genetic polymorphisms in CASP3, CASP7, TRAILR2, GSDME, CASP4, HO-1, ALOX5, GPX4, and NRF2 that were significantly associated with OS in the additive model (P < 0.05). There was a substantial cumulative effect of three genetic polymorphisms (CASP4 rs571407, ALOX5 rs2242332, and HO-1 rs17883419) on OS. Genetic variations in the CASP4 and ALOX5 gene haplotypes were associated with a higher OS. We demonstrated, for the first time, that rs702365 [G] > [C] represses ALOX5 transcription and corollary experiments suggested that ALOX5 may promote colon cancer cell growth by mediating an inflammatory response. CONCLUSIONS: Polymorphisms in genes regulating cell death may play essential roles in the prognosis of patients with rectal cancer who are treated with postoperative CRT and may serve as potential genetic biomarkers for individualized treatment.


Assuntos
Polimorfismo Genético , Neoplasias Retais , Humanos , Prognóstico , Quimiorradioterapia , Morte Celular , Biomarcadores , Neoplasias Retais/genética , Neoplasias Retais/terapia
7.
Biomaterials ; 295: 121994, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36775789

RESUMO

Prompting higher-order death receptor (DR) clustering by increasing the valency of DR agonist is efficient to induce apoptosis of tumor cells. As an attractive DR agonist with superior biosafety, the trimeric tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) exerts limited antitumor effect in patients, which is predominantly attributed to its low DR clustering ability and short serum half-life. Previous antibody scaffolds-based engineering strategies to increase the valency and/or prolong the serum half-life of TRAIL improve apoptosis induction, however, often produce large proteins with poor tumor penetration. Covalent protein ligation mediated by small molecular superglues such as SpyTag/SpyCatcher might be a novel strategy to assemble higher-order TRAIL variants. Upon fusion to TRAIL promotor, SpyTag/SpyCatcher molecular superglue preferentially ligated two trimeric TRAIL to produce a hexameric TRAIL variant, HexaTR, exhibiting a significantly increased apoptosis induction. In addition, an albumin-binding HexaTR, ABD-HexaTR, with a prolonged serum half-life by binding to endogenous albumin was also produced using the same strategy. Compared to the trimeric TRAIL, the hexameric HexaTR and ABD-HexaTR showed 20-50 times greater in vivo antitumor effect, resulting in eradication of several types of large (150-300 mm3) tumor xenografts. Combination with bortezomib carried by liposome further improved the antitumor effects of the hexavalent HexaTR and ABD-HexaTR in refractory cancer. Our results indicate that the superglue-mediated higher-order assembly is promising to improve the DR clustering and proapoptotic signaling of TRAIL, showing great advantages in constructing the next generation of DR agonists for cancer therapy.


Assuntos
Apoptose , Ligante Indutor de Apoptose Relacionado a TNF , Humanos , Linhagem Celular Tumoral , Ligantes , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Albuminas/farmacologia
8.
ChemSusChem ; 15(24): e202201584, 2022 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-36195829

RESUMO

Small-molecule induction can lead to the oriented migration of metal elements, which affords functional materials with synergistic components. In this study, phosphating nickel foam (NF)-supported octahedral WO3 with phosphine affords P-WO3 /NF electrocatalyst. Ni is found to form Ni-P bonds that migrate from NF to WO3 under the induction of P, resulting in the complex oxides W1.3 Ni0.24 O4 and Ni2 P2 O7 in the particle interior and nickel phosphide on the octahedral grain surface. The catalytic activity of P-WO3 /NF in the urea oxidation reaction (UOR) is improved by synergistic action of the components in the synthesized hybrid particles. A current density of 10 mA cm-2 can be reached at a potential of 1.305 V, the double layer capacitance of the catalyst is significantly increased, and the electron transfer impedance in catalytic UOR is reduced. This work demonstrates that small-molecule induction is suitable for constructing co-catalysts with complex components in a simple protocol, which provides a new route for the design of highly efficient urea oxidation electrocatalysts.


Assuntos
Níquel , Óxidos , Oxirredução , Transporte de Elétrons , Ureia
9.
Drug Deliv ; 29(1): 1698-1711, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35635308

RESUMO

Multidrug resistance (MDR), which is common in colorectal cancer (CRC), induces high mortality in patients. Due to its robust and selective apoptosis induction in some CRC cells with MDR, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is attractive as a novel tool for CRC therapy. However, TRAIL is limited by its poor tumor-homing ability and inefficient apoptosis induction in CRC cells expressing low levels of death receptor (DR). Here, the tumor-homing RGR peptide (CRGRRST) was fused to TRAIL to produce RGR-TRAIL. Compared with TRAIL, RGR-TRAIL showed greater cell binding and cytotoxicity in CRC cells. In addition, RGR-TRAIL exerted significantly enhanced tumor uptake and growth suppression in mice bearing CRC tumor xenografts. Notably, RGR-TRAIL eradicated all tumor xenografts of DR-overexpressing COLO205 cells. However, TRAIL only showed mild tumor growth suppression under the same conditions, indicating that RGR fusion significantly increased the antitumor effect of TRAIL in DR-overexpressing CRC cells by improving tumor homing. Nevertheless, RGR fusion did not significantly enhance the antitumor effect of TRAIL in HT29 cells expressing low levels of DR. We found that DR expression in HT29 cells was enhanced by epidermal growth factor receptor (EGFR)-targeted photodynamic therapy (PDT). Moreover, both the in vitro and in vivo antitumor effects of RGR-TRAIL were significantly improved by combination with PDT. HT29 tumor xenografts (∼20%) were even eradicated by combination therapy. These results indicate that it is valuable to further evaluate the combination therapy of RGR-TRAIL and tumor-targeted PDT for clinical therapy of CRC with MDR.


Assuntos
Neoplasias Colorretais , Fotoquimioterapia , Animais , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Xenoenxertos , Humanos , Camundongos , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia
10.
J Anim Sci ; 100(4)2022 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-35275597

RESUMO

Keel bone damage (KBD) is more prevalent in alternative laying hen housing systems than in conventional cages, and its incidence differs from strain to strain. However, the information of KBD in Lindian chickens, a native Chinese strain, is limited. To investigate the effect of KBD on fearfulness and physiological indicators of stress in Lindian chickens and commercial laying hens, a total of two hundred 25-wk-old chickens (100 Hy-line Brown and 100 Lindian chickens) were studied for 7 wk. The birds were housed in furnished cages with 10 birds per cage for each strain. At 32-wk of age, the birds in each strain were divided into normal (NK), deviated (DK), and fractured (FK) hens according to the keel bone status. Ten birds in each keel bone status per strain were subsequently selected to collect blood for the determination of stress and fear-related indicators, including corticosterone, serotonin, interleukin-1ß, and interleukin-6, and measure fear responses, including novel object test (NOT), human approach test (HAT), and tonic immobility (TI) test. The results showed that egg production was lower and the incidence of keel bone fractures was higher in Lindian chickens than in Hy-line Brown hens (P < 0.05). Lindian chickens showed a significantly increased whole blood serotonin content, NOT-latency, HAT-score, and TI induction times (P < 0.05) and decreased serum interleukin-6 content and TI-duration (P < 0.05) compared with Hy-line Brown hens. Additionally, FK hens had significantly elevated whole blood corticosterone, serum interleukin-1ß and interleukin-6 levels, TI-duration, and NOT-latency (P < 0.05), and a reduced whole blood serotonin content (P < 0.05) compared with NK and DK hens. Our results indicated that KBD affected stress and fear responses, and this impact was mainly reflected by FK hens compared with NK and DK hens. We suggest that keel bone fractures are the main factor impairing hen welfare. Besides, the incidence of keel bone fractures and stress and fear responses of Lindian chickens are more severe than Hy-line Brown laying hens, indicating that the strain type can affect the health and welfare of laying hens.


Keel bone damage (KBD) impairs production performance, welfare, and health in laying hens. This study aimed to compare the incidence of KBD and investigate the effects of KBD on stress and fear in two strains of laying hens. The results showed that commercial Hy-line Brown laying hens had high egg production and low incidence of KBD compared with Lindian chickens, a Chinese native breed. Besides, Lindian chickens had higher blood serotonin content and fear responses to human approach test and novel object test than Hy-line Brown laying hens. In addition, laying hens with keel bone fractures had elevated concentrations of blood corticosterone, interleukin-1ß, and interleukin-6, and had a longer duration of tonic immobility and latency to approach a novel object, as well as reduced blood serotonin content compared with laying hens with normal and deviated keel bone. Overall, keel bone fractures caused stress and fear responses, impairing hen welfare; and behavioral and physiological responses in relation to stress and fear differed between strains of hens.


Assuntos
Galinhas , Fraturas Ósseas , Criação de Animais Domésticos/métodos , Bem-Estar do Animal , Animais , Galinhas/fisiologia , Corticosterona , Medo , Feminino , Fraturas Ósseas/epidemiologia , Fraturas Ósseas/etiologia , Fraturas Ósseas/veterinária , Abrigo para Animais , Interleucina-1beta , Interleucina-6 , Serotonina
11.
Ann Surg Treat Res ; 102(3): 153-158, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35317354

RESUMO

Purpose: This study was performed to investigate the association of Glasgow prognostic score (GPS), combined positive score (CPS), and clinicopathological characteristics of locally advanced rectal cancer. Methods: Between February 2012 and February 2018, 103 patients with locally advanced rectal cancer treated by neoadjuvant chemoradiotherapy and total mesorectal excision (TME) were retrospectively evaluated. Results: According to the classification of the GPS, 85 (82.5%), 13 (12.6%), and 5 patients (4.9%) were classified as a score of 0, 1, and 2, respectively. Patients were classified into the GPS-low group (GPS of 0, n = 85) and GPS-high group (GPS of 1 or 2, n = 18) with an area under the curve of 0.582 for overall survival (OS). The mean programmed death-ligand 1 (PD-L1) CPS of the whole group was 2.24 (range, 0-70). The PD-L1 CPS of the GPS-high group was higher than the GPS-low group (P < 0.001). Multivariate analysis by Cox proportional hazards model indicated that GPS was associated with OS and disease-free survival (DFS). Furthermore, PD-L1 CPS was associated with DFS (hazard ratio, 1.050; 95% confidence interval, 1.017-1.083; P = 0.003). Conclusion: Elevated GPS was related to the PD-L1 CPS. GPS and PD-L1 CPS were associated with the prognosis of locally advanced rectal cancer treated with neoadjuvant chemoradiotherapy followed by TME.

12.
Discov Oncol ; 13(1): 10, 2022 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-35201501

RESUMO

PURPOSE: To evaluate the role of programmed death-ligand 1 (PD-L1) and mammalian target of rapamycin (mTOR) signaling pathway in locally advanced rectal cancer (LARC). METHODS: Between February 2012 and February 2018, 103 patients with LARC treated by neoadjuvant chemoradiotherapy (neoCRT) and total mesorectal excision (TME) were included. PD-L1, mTOR and p-mTOR of pair-matched pre-neoCRT biopsies and post-neoCRT surgical tissue were evaluated by immunohistochemistry. RESULTS: The mean combined positive score (CPS), tumor proportion score (TPS) and immune cell score (IC) of pre-neoCRT were 2.24 (0-70), 1.87 (0-70) and 0.67 (0-10), respectively. The mean CPS, TPS and IC of post-neoCRT were 2.19 (0-80), 1.38 (0-80) and 1.60 (0-20), respectively. Significant difference was observed in terms of IC between pre-neoCRT and post-neoCRT (p = 0.010). The 5-year disease-free survival (DFS) rate of the whole group was 62.4%. Multivariate analysis by Cox model indicated that pre-neoCRT TPS [hazard ratio (HR) 1.052, 95% confidence interval (CI) 1.020-1.086, p = 0.001] and post-neoCRT CPS (HR 0.733, 95% CI 0.555-0.967, p = 0.028) were associated with DFS. In the 89 patients without pathological complete response, p-mTOR and IC were upregulated after neoCRT. CONCLUSIONS: For patients with LARC treated by neoCRT and TME, p-mTOR and IC were upregulated after neoCRT. Pre-neoCRT TPS and post-neoCRT CPS were independent prognostic predictors of DFS.

13.
J Control Release ; 340: 243-258, 2021 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-34752799

RESUMO

Immunotherapies based on immune checkpoint-blocking antibodies have been considered the most attractive cancer treatments in recent years. However, the systemic administration of immune checkpoint-blocking antibodies is limited by low response rates and high risk of inducing immune-related adverse events (irAEs), which might be overcome by the tumor-targeted delivery of these antibodies. To achieve tumor-targeted delivery, immune checkpoint-blocking antibodies are usually modified with tumor-homing ligands through difficult genetic fusion or chemical conjugation. As most immune checkpoint-blocking antibodies are immunoglobin G (IgG) antibodies, we hypothesize that these IgG antibodies might be noncovalently modified with a tumor-homing ligand fused to an IgG-binding domain (IgBD). To test this hypothesis, the tumor-homing ZPDGFRß affibody, which targets platelet-derived growth factor receptor ß (PDGFRß), was fused to the Fab-selective IgBD in a trimeric format. After mixing ZPDGFRß fused to the IgBD with immune checkpoint-blocking IgG against programmed death-ligand 1 (αPD-L1), a novel homogenous complex was formed, indicating that αPD-L1 had been successfully modified with ZPDGFRß fused to the IgBD. ZPDGFRß-modified αPD-L1 bound to both PDGFRß and PD-L1, thus leading to greater tumor uptake and antitumor effects in mice bearing PDGFRß+PD-L1+ tumor grafts. In addition, due to the broad spectrum of IgBD for IgG, immune checkpoint-blocking IgG antibodies against cytotoxic T-lymphocyte-associated protein 4 (αCTLA-4) and signal regulatory protein alpha (αSIRPα) were also modified with ZPDGFRß fused to the IgBD. These results demonstrated that a tumor-homing ligand fused to the IgBD might be developed as a versatile platform for the modification of immune checkpoint-blocking IgG antibodies to achieve tumor-targeted delivery.


Assuntos
Sistemas de Liberação de Medicamentos , Inibidores de Checkpoint Imunológico , Imunoglobulina G/imunologia , Neoplasias , Animais , Imunoterapia , Camundongos , Neoplasias/tratamento farmacológico , Receptor beta de Fator de Crescimento Derivado de Plaquetas
14.
JAMA Netw Open ; 4(11): e2136116, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34846525

RESUMO

Importance: Several studies have explored the efficacy and toxic effects of concurrent 5-fluorouracil (5-FU)- or capecitabine-based chemoradiotherapy (CRT) with or without oxaliplatin in the neoadjuvant setting. Addition of oxaliplatin to 5-FU or capecitabine elicited similar outcomes but with significantly increased toxic effects; however, there is a need for randomized clinical trials comparing 2 CRT regimens for patients receiving CRT in the adjuvant setting. Objective: To explore the efficacy and toxic effects of oxaliplatin combined with postoperative concurrent capecitabine and radiotherapy (RT) for pathological stage II and III rectal cancer. Design, Setting, and Participants: This multicenter randomized clinical trial enrolled patients from 7 centers in China between April 1, 2008, and December 30, 2015. Patients with pathologically confirmed stage II and III rectal cancer were randomized (1:1) to receive concurrent CRT with capecitabine or capecitabine plus oxaliplatin. Analysis was conducted from December 31, 2019, to March 15, 2020. Interventions: RT comprised 45 to 50 Gy in 25 fractions of 1.8 to 2.0 Gy over 5 weeks. In the capecitabine with RT group, concurrent chemotherapy included 2 cycles of capecitabine (1600 mg/m2) on days 1 to 14 and 22 to 35. The capecitabine and oxaliplatin with RT group received identical postoperative RT to that in the capecitabine with RT group combined with capecitabine (1300 mg/m2) on days 1 to 14 and 22 to 35 and a 2-hour infusion of oxaliplatin (60 mg/m2) on weeks 1, 2, 4, and 5. Patients in both groups received adjuvant chemotherapy (capecitabine or fluorouracil and oxaliplatin) after CRT. Main Outcomes and Measures: The primary end point was 3-year disease-free survival (DFS). Results: A total of 589 patients (median [IQR] age, 55 [47-52] years; 375 [63.7%] men and 214 [36.3%] women) were enrolled, including 294 patients randomized to the capecitabine with RT group and 295 patients randomized to the capecitabine and oxaliplatin with RT group. Median (IQR) follow-up was 68 (45-96) months. Most patients had stage III disease (574 patients [75.9%]). Three-year DFS was 76.3% for the capecitabine with RT group and 74.1% for the capecitabine and oxaliplatin with RT group, and 5-year DFS was 72.0% for the capecitabine with RT group and 71.1% for the capecitabine and oxaliplatin with RT group (hazard ratio [HR], 1.07; 95% CI, 0.79-1.44; P = .68). There was no significant difference between groups in overall survival (HR, 0.93; 95% CI, 0.64-1.34; P = .70) or local recurrence (HR, 0.61; 95% CI, 0.31-1.22; P = .16). More grade 3 and 4 acute toxic effects were observed in the capecitabine and oxaliplatin with RT group than in the capecitabine with RT group (114 patients [38.6%] vs 84 patients [28.6%]; P = .01). Conclusions and Relevance: This randomized clinical trial found that addition of oxaliplatin to capecitabine-based postoperative CRT did not improve the efficacy of treatment but increased the risk of severe acute toxic effects. This finding highlights the basic role of postoperative capecitabine with RT for patients with locally advanced rectal cancer. Trial Registration: ClinicalTrials.gov Identifier: NCT00714077.


Assuntos
Capecitabina/uso terapêutico , Quimiorradioterapia/métodos , Fluoruracila/uso terapêutico , Oxaliplatina/uso terapêutico , Neoplasias Retais/tratamento farmacológico , Neoplasias Retais/radioterapia , Neoplasias Retais/cirurgia , Antimetabólitos Antineoplásicos/uso terapêutico , Antineoplásicos/uso terapêutico , China , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Terapia Neoadjuvante/métodos , Cuidados Pós-Operatórios/métodos , Resultado do Tratamento
15.
Theranostics ; 11(9): 4281-4297, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33754061

RESUMO

Chemotherapeutic multidrug resistance (MDR) is the major hindrance for clinical therapy of colorectal cancer (CRC). Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) with selective cytotoxicity might overcome MDR of CRC cells. Unfortunately, cross-resistance to TRAIL has been detected in many CRC cells, suggesting the need to combine TRAIL with sensitizers to combat refractory CRC. Our purpose is to explore the potential of combination therapy of TRAIL and tumor-cell targeted photodynamic therapy (PDT) in combating CRC with both chemotherapeutic MDR and TRAIL resistance. Methods: Tumor cell-targeted PDT was performed using a Ze-IR700 photosensitizer with high affinity for epidermal growth factor receptor (EGFR). The impact of PDT on the gene expression of CRC cells was revealed by RNA sequencing. The synergistic antitumor effect of long-acting TRAIL and PDT was evaluated in mice bearing tumor grafts of CRC cells with both chemotherapeutic MDR and TRAIL resistance. Results: Chemotherapeutic MDR and TRAIL resistance are common in CRC cells. Pretreatment of CRC cells with tumor cell-targeted PDT significantly (10-60 times) increased the sensitivity of these CRC cells to TRAIL by upregulating death receptors. Combination therapy, but not monotherapy, of long-acting TRAIL and PDT greatly induced apoptosis of CRC cells, thus efficiently eradicated large (~150 mm3) CRC tumor xenografts in mice. Conclusions: Tumor cell-targeted PDT extensively sensitizes CRC cells to TRAIL. Combination therapy of long-acting TRAIL and PDT is promising to combat CRC with both chemotherapeutic MDR and TRAIL resistance, which might be developed as a novel strategy for precision therapy of refractory CRC.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Células HCT116 , Células HT29 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia
16.
Appl Microbiol Biotechnol ; 105(4): 1447-1460, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33528691

RESUMO

Due to their potent immune stimulation, tumor necrosis factor alpha (TNFα) variants with tumor-homing activity are attractive as novel antitumor drugs. The promising antitumor effect of NGR-TNFα in clinical trials triggered extensive interest in developing novel tumor-homing TNFα variants in recent years. Owing to its promising antitumor effect, NGR-TNFα is usually used as a control for newly developed tumor-homing TNFα variants. In our previous works, we produced a pericyte-targeting Z-TNFα at high levels using the Escherichia coli (E. coli) M15-pQE30 system. To further compare Z-TNFα and NGR-TNFα, we attempted to express NGR-TNFα using the same system. Surprisingly, native NGR-TNFα was expressed at a low (~ 0.2 mg/L) level in E. coli M15 containing the pQE30 plasmid. However, a single nucleotide mutation of C to G, resulting in a substitution of leucine (L) with valine (V) at the start of TNFα, increased the expression of NGR-TNFα by ~ 100 times through improving transcription. In addition, the amino acid substitution showed a little impact on the receptor binding, in vitro cytotoxicity, and in vivo antitumor effect of NGR-TNFα. As fusing NGR to the N-terminus of TNFα with a valine substitution did not reduce the protein yield, the TNFα gene with a C > G mutation might be used to prepare novel tumor-homing TNFα when the native TNFα-based variant is expressed at an extremely low level in E. coli. Notably, in addition to the mutated valine, the impact of N-terminal additional amino acids provided by pQE30 vector on the function of TNFα variant must be carefully evaluated. KEY POINTS : • A single nucleotide mutation increased the expression of NGR-TNFα by two orders. • Nucleotide mutation-induced amino acid substitution did not reduce NGR-TNFα activity.


Assuntos
Escherichia coli , Fator de Necrose Tumoral alfa , Linhagem Celular Tumoral , Escherichia coli/genética , Galanina/análogos & derivados , Mutação , Nucleotídeos , Oligopeptídeos/genética , Substância P/análogos & derivados , Transcrição Gênica , Fator de Necrose Tumoral alfa/genética
17.
Mol Med Rep ; 23(2)2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33300054

RESUMO

Colorectal cancer (CRC) is one of the most common types of malignancy and the third most commonly diagnosed form of cancer worldwide, ranking as the fourth leading cause of cancer­associated mortality. MicroRNA (miR)­576­5p has been reported to be highly expressed in patients with CRC; however, its biological role remains unclear. The present study aimed therefore to investigate the biological role and underlying mechanism of miR­576­5p in CRC cell line SW480. The viability of SW480 cells following transfection with miR­576­5p mimic or inhibitor was analyzed using MTT assay. Wound healing and Transwell assays were performed to determine the cell migratory and invasive abilities, respectively. A dual luciferase reporter assay was used to verify the predicted binding site between miR­576­5p and Wnt5a. Reverse transcription­quantitative PCR and western blotting were used to analyze the expression levels of miR­576­5p, E­cadherin, N­cadherin, vimentin, Snail1, Wnt5a, ß­catenin, c­myc, cyclin D1 and p/t­c­Jun. Using bioinformatics analysis, high expression of miR­576­5p was found not only in tumor tissues, compared with the normal tissue, but also in CRC cells, compared with NCM460 cells. Furthermore, the inhibition of miR­576­5p expression significantly decreased the cell viability and the migratory and invasive abilities of SW480 cells, and suppressed the epithelial­to­mesenchymal transition (EMT). In addition, miR­576­5p could interact with Wnt5a and regulate the expression level of Wnt5a in order to influence the activity of Wnt/ß­catenin signaling. The results from rescue experiments further demonstrated that the effect of miR­576­5p overexpression on cell metastasis and EMT was reversed by Wnt5a overexpression or treatment with XAV­939, which is an inhibitor of the Wnt/ß­catenin signaling pathway. In conclusion, the findings from the present study suggested that inhibition of miR­576­5p may suppress SW480 cell metastasis and EMT by targeting Wnt5a and regulating the Wnt5a­mediated Wnt/ß­catenin signaling pathway, providing a potential therapeutic target for the treatment of CRC.


Assuntos
Neoplasias Colorretais/metabolismo , Transição Epitelial-Mesenquimal , MicroRNAs/metabolismo , RNA Neoplásico/metabolismo , Via de Sinalização Wnt , Proteína Wnt-5a/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Células HCT116 , Humanos , RNA Neoplásico/genética , Proteína Wnt-5a/genética
18.
BMC Cancer ; 20(1): 862, 2020 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-32894078

RESUMO

BACKGROUND: The aim of this study is to assess biplane transrectal ultrasonography (TRUS) plus ultrasonic elastosonography (UE) and contrast-enhanced ultrasonography (CEUS) in T staging of rectal cancer. METHODS: Between March 2016 and January 2019, 66 rectal cancer patients who completed biplane TRUS plus UE and CEUS for preoperative workup and were treated by primary total mesorectal excision (TME) were retrospectively analyzed. RESULTS: The accuracy of TRUS plus UE and CEUS in all T staging of rectal cancer was 69.7%. The highest accuracy was achieved in the T3 stage (87.5%), while it was 71.4 and 50.0% in the T1 and T2 stage, respectively. The mean sizes of uT1-T2 lesions and uT3-T4 lesions were 30.0 ± 10.6 mm (range, 10.0-55.0) and 40.2 ± 11.2 mm (range, 14.0-57.0), respectively (p < 0.001). According to the receiver operating characteristic (ROC) curve to predict pT stages (pT1,2 vs. pT3), the optimal cut-off value of lesions in greatest dimension was 28.5 mm by TRUS with areas under the curve (AUC) of 0.769, and the optimal cut-off values of peak systolic velocity (PSV) and resistive index (RI) were 18.8 cm/sec and 0.645, respectively. The AUCs of PSV and RI were 0.588 and 0.555, respectively. CONCLUSIONS: Diagnostic accuracy of TRUS plus UE and CEUS in T staging of rectal cancer does not reach the excellent published study results, especially for patients with early rectal cancer. Tumor sizes, PSV and RI are useful additions for TRUS in T staging of rectal cancer.


Assuntos
Endossonografia , Neoplasias Retais/diagnóstico , Reto/diagnóstico por imagem , Ultrassom Focalizado Transretal de Alta Intensidade , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Cintilografia/métodos , Neoplasias Retais/diagnóstico por imagem , Neoplasias Retais/patologia , Reto/patologia , Estudos Retrospectivos
19.
Biomacromolecules ; 21(10): 4017-4029, 2020 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-32804484

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an attractive antitumor drug candidate for precision cancer therapy due to its superior selective cytotoxicity in a variety of tumor cells. However, the clinical application of TRAIL in cancer therapy has been limited by its poor tumor-homing capacities and short half-life. Herein, we designed a tridomain TRAIL variant, Z-ABD-TRAIL, by sequentially fusing the platelet-derived growth factor receptor beta (PDGFRß)-specific affibody ZPDGFRß and an albumin-binding domain (ABD) to the N-terminus of TRAIL. The fusion protein Z-ABD-TRAIL was produced as a soluble protein with high yield in Escherichia coli (E. coli). The ZPDGFRß domain provided Z-ABD-TRAIL with PDGFRß-binding properties and thus promoted its tumor homing via the engagement of PDGFRß-expressing pericytes on tumor microvessels. ABD-mediated binding of Z-ABD-TRAIL to albumin in the blood endowed TRAIL with long-lasting (>72 h for Z-ABD-TRAIL vs <0.5 h for TRAIL) abilities to kill tumor cells. Although the in vitro cytotoxicity of Z-ABD-TRAIL in tumor cells was similar to that of the parent TRAIL, the in vivo tumor uptake, apoptosis-inducing ability, and antitumor effect of Z-ABD-TRAIL were much greater than those of TRAIL, indicating that ZPDGFRß-mediated tumor homing and ABD-introduced albumin binding significantly improved the pharmacodynamics of TRAIL. In addition, repeated injection of high-dose Z-ABD-TRAIL showed no obvious acute toxicity in mice. These results demonstrate that the newly designed tridomain Z-ABD-TRAIL is a promising agent for precision cancer therapy.


Assuntos
Escherichia coli , Ligante Indutor de Apoptose Relacionado a TNF , Albuminas , Animais , Apoptose , Linhagem Celular Tumoral , Preparações de Ação Retardada , Camundongos , Ligante Indutor de Apoptose Relacionado a TNF/genética
20.
Apoptosis ; 25(1-2): 105-119, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31894448

RESUMO

Liver fibrosis usually progresses to liver cirrhosis and even hepatocellular carcinoma. Since activated hepatic stellate cells (aHSCs) are responsible for liver fibrosis, reducing the quantity of aHSCs was considered the essential strategy for clinical antihepatofibrotic therapy. Due to the overexpression of TRAIL receptor 2 (DR5) in aHSCs, human TNF-related apoptosis-inducing ligand (hTRAIL) that could induce aHSCs apoptosis might be feasible for antihepatofibrotic therapy. However, the in vivo aHSCs-apoptosis-induction of hTRAIL is limited by its poor cell-targeting and a short half-life. In this study, we found that platelet-derived growth factor receptor ß (PDGFRß) was co-expressed with DR5 in aHSCs. And the ZPDGFRß affibody with high affinity for PDGFRß could bind aHSCs and, thus, accumulate in the fibrotic liver. ZPDGFRß was fused to hTRAIL to produce the fusion protein Z-hTRAIL. Compared to hTRAIL, Z-hTRAIL showed greater in vitro cell binding and apoptosis-induction in aHSCs. In addition, Z-hTRAIL induced apoptosis of aHSCs but spared other normal liver cells. In vivo, Z-hTRAIL accumulated preferentially in fibrotic livers and exerted greater effects than hTRAIL in inducing aHSCs apoptosis and reducing extracellular matrix (ECM) deposition. These results demonstrated that the antihepatofibrotic effect of hTRAIL was improved by PDGFRß-targeted delivery. To enhance its pharmacokinetics, Z-hTRAIL was modified with 10 kDa polyethylene glycol (PEG), which significantly (30-40 times) prolonged its half-life. The PEGylated long-acting Z-hTRAIL was more potent than the native Z-hTRAIL in regressing liver fibrosis. These results suggest that the aHSC-targeting and long-acting Z-hTRAIL might serve as a novel tool for antihepatofibrotic therapy.


Assuntos
Células Estreladas do Fígado/metabolismo , Cirrose Hepática/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Feminino , Células Estreladas do Fígado/citologia , Humanos , Cirrose Hepática/genética , Cirrose Hepática/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA