Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.205
Filtrar
1.
Sci Rep ; 14(1): 7539, 2024 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-38553472

RESUMO

High grade serous carcinoma (HGSC) metastasises primarily intraperitoneally via cancer spheroids. Podocalyxin (PODXL), an anti-adhesive transmembrane protein, has been reported to promote cancer survival against chemotherapy, however its role in HGSC chemoresistance is unclear. This study investigated whether PODXL plays a role in promoting chemoresistance of HGSC spheroids. We first showed that PODXL was expressed variably in HGSC patient tissues (n = 17) as well as in ovarian cancer cell lines (n = 28) that are more likely categorised as HGSC. We next demonstrated that PODXL-knockout (KO) cells proliferated more slowly, formed less compact spheroids and were more fragile than control cells. Furthermore, when treated with carboplatin and examined for post-treatment recovery, PODXL-KO spheroids showed significantly poorer cell viability, lower number of live cells, and less Ki-67 staining than controls. A similar trend was also observed in ascites-derived primary HGSC cells (n = 6)-spheroids expressing lower PODXL formed looser spheroids, were more vulnerable to fragmentation and more sensitive to carboplatin than spheroids with higher PODXL. Our studies thus suggests that PODXL plays an important role in promoting the formation of compact/hardy HGSC spheroids which are more resilient to chemotherapy drugs; these characteristics may contribute to the chemoresistant nature of HGSC.


Assuntos
Carcinoma , Neoplasias Ovarianas , Feminino , Humanos , Carboplatina/farmacologia , Carboplatina/uso terapêutico , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo
2.
Biomed Pharmacother ; 173: 116403, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38490156

RESUMO

Drug resistance (DR) is one of the challenges in treating retinoblastoma (Rb) that warrants novel approaches. With the emerging evidence on the role of small extracellular vesicles (sEVs) as a drug-delivery carrier system, in this study, we derived the drug-resistant (DR) clones of Y79 cells and evaluated the efficacy of sEVs-loaded with carboplatin (sEVs-CPT) to reverse the chemoresistance. Drug-resistant clones of Y79 cells (DR-Y79) were systematically developed through sequential exposure to carboplatin (CPT), showcasing a sixfold increase in inhibitory concentration when compared to parental Y79 cells (IC50: 41.4 µg/mL and 6.2 µg/mL) (P<0.0001). These DR-Y79 cells show higher expression of ABCG2 and higher expression of DR genes than parental Y79 cells (P<0.0001). The sEVs were isolated from the conditioned media of Y79 cells using ultracentrifugation (UC) and characterized. Further, the sEVs were loaded with CPT and achieved higher encapsulation efficiency at one hour, and drug release of sEVs-CPT was highest at ∼80% at pH 5.0. The cytotoxicity of sEVs-CPT on Y79 cells and DR-Y79 was higher when compared to the CPT (IC50: 3.5 µg/mL vs 6.2 µg/mL; 23.1 µg/mL vs 41.2 µg/mL) (p<0.0001). This study demonstrates that sequential exposure to CPT generates DR clones of Y79 cells, which could serve as an appropriate model to evaluate the efficacy of drugs. The sEVs-CPT were highly effective in enhancing cytotoxicity in DR-Y79 cells, and appear to hold promise as a novel complimentary drug delivery system.


Assuntos
Vesículas Extracelulares , Neoplasias da Retina , Humanos , Carboplatina/farmacologia , Preparações Farmacêuticas , Linhagem Celular Tumoral
3.
J Inorg Biochem ; 254: 112505, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38377623

RESUMO

Platinum-based anticancer drugs, while potent, are associated with numerous and severe side effects. Hyperthermia therapy is an effective adjuvant in anticancer treatment, however, clinically used platinum drugs have not been optimised for combination with hyperthermia. The derivatisation of existing anticancer drugs with appropriately chosen thermoresponsive moieties results in drugs being activated only at the heated site. Perfluorinated chains of varying lengths were installed on carboplatin, a clinically approved drug, leading to the successful synthesis of a series of mono- and di- substituted platinum(IV) carboplatin prodrugs. Some of these complexes display relevant thermosensitivity on ovarian cancer cell lines, i.e., being inactive at 37 °C while having comparable activity to carboplatin under mild hyperthermia (42 °C). Nuclear magnetic resonance spectroscopy and mass spectrometry indicated that carboplatin is likely the active platinum(II) anticancer agent upon reduction and cyclic voltammetry revealed that the length of the fluorinated alkyl chain has a strong influence on the rate of carboplatin formation, regulating the subsequent cytotoxicity.


Assuntos
Antineoplásicos , Neoplasias Ovarianas , Pró-Fármacos , Feminino , Humanos , Carboplatina/farmacologia , Carboplatina/química , Pró-Fármacos/química , Antineoplásicos/química , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Platina/química , Cisplatino/química
4.
Int J Mol Sci ; 25(4)2024 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-38396859

RESUMO

Kinetically inert platinum(IV) complexes are a chemical strategy to overcome the impediments of standard platinum(II) antineoplastic drugs like cisplatin, oxaliplatin and carboplatin. In this study, we reported the syntheses and structural characterisation of three platinum(IV) complexes that incorporate 5-benzyloxyindole-3-acetic acid, a bioactive ligand that integrates an indole pharmacophore. The purity and chemical structures of the resultant complexes, P-5B3A, 5-5B3A and 56-5B3A were confirmed via spectroscopic means. The complexes were evaluated for anticancer activity against multiple human cell lines. All complexes proved to be considerably more active than cisplatin, oxaliplatin and carboplatin in most cell lines tested. Remarkably, 56-5B3A demonstrated the greatest anticancer activity, displaying GI50 values between 1.2 and 150 nM. Enhanced production of reactive oxygen species paired with the decline in mitochondrial activity as well as inhibition of histone deacetylase were also demonstrated by the complexes in HT29 colon cells.


Assuntos
Antineoplásicos , Ácido Hidroxi-Indolacético/análogos & derivados , Pró-Fármacos , Humanos , Cisplatino/farmacologia , Platina/química , Oxaliplatina/farmacologia , Carboplatina/farmacologia , Carboplatina/química , Pró-Fármacos/química , Linhagem Celular Tumoral , Antineoplásicos/química
5.
Oncotarget ; 15: 144-158, 2024 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-38386805

RESUMO

Sacituzumab govitecan (SG) is an antibody-drug conjugate composed of an anti-Trop-2-directed antibody conjugated with the topoisomerase I inhibitory drug, SN-38, via a proprietary hydrolysable linker. SG has received United States Food and Drug Administration (FDA) approval to treat metastatic triple-negative breast cancer (TNBC), unresectable locally advanced or metastatic hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancer, and accelerated approval for metastatic urothelial cancer. We investigated the utility of combining SG with platinum-based chemotherapeutics in TNBC, urinary bladder carcinoma (UBC), and small-cell lung carcinoma (SCLC). SG plus carboplatin or cisplatin produced additive growth-inhibitory effects in vitro that trended towards synergy. Immunoblot analysis of cell lysates suggests perturbation of the cell-cycle and a shift towards pro-apoptotic signaling evidenced by an increased Bax to Bcl-2 ratio and down-regulation of two anti-apoptotic proteins, Mcl-1 and survivin. Significant antitumor effects were observed with SG plus carboplatin in mice bearing TNBC or SCLC tumors compared to all controls (P < 0.0062 and P < 0.0017, respectively) and with SG plus cisplatin in UBC and SCLC tumor-bearing animals (P < 0.0362 and P < 0.0001, respectively). These combinations were well tolerated by the animals. Combining SG with platinum-based chemotherapeutics demonstrates the benefit in these indications and warrants further clinical investigation.


Assuntos
Anticorpos Monoclonais Humanizados , Camptotecina/análogos & derivados , Carcinoma , Imunoconjugados , Neoplasias Pulmonares , Carcinoma de Pequenas Células do Pulmão , Neoplasias de Mama Triplo Negativas , Neoplasias da Bexiga Urinária , Humanos , Estados Unidos , Animais , Camundongos , Bexiga Urinária , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Platina , Cisplatino/farmacologia , Carboplatina/farmacologia , Imunoconjugados/farmacologia , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Pulmão
6.
Oncologist ; 29(1): 47-56, 2024 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-37390616

RESUMO

BACKGROUND: Combination chemotherapy and immunotherapy regimens have significantly improved survival for patients with previously untreated advanced non-small cell lung cancer (NSCLC). Improvements in overall survival (OS) in two separate pembrolizumab trials have demonstrated survival improvements over chemotherapy alone, regardless of PD-L1 status. The optimal chemotherapy backbone for combination with immunotherapy is unknown. We hypothesized nab-paclitaxel may be a well-suited platinum partner to use in combination with checkpoint inhibitor therapy for both adenocarcinoma and squamous histology and conducted a phase I/II trial to assess the efficacy of this regimen in advanced NSCLC. METHODS: Adult patients with previously untreated, stage IIIB/IV NSCLC (any histology) with an Eastern Cooperative Oncology Group performance status of 0-1, any PD-L1 expression, and no EGFR mutations or ALK translocations, received carboplatin area under the curve (AUC) 6 day 1, nab-paclitaxel 100 mg/m2 days 1, 8, 15, and pembrolizumab 200 mg day 1 q21 days for 4 cycles followed by maintenance pembrolizumab q3w. Co-primary endpoints were progression-free survival (PFS) and overall response rate (ORR). RESULTS: Forty-six evaluable patients enrolled, 14 in phase I and 32 in phase II, from June 2015 to July 2018 with a median duration of follow-up of 35.4 months. Median time from enrollment to data lock was 42 months. In the ITT population, the ORR was 35%, median PFS was 5.6 months (95% CI, 4.6-8.2), and median OS was 15.4 months (CI, 12.4-28.1). There were no statistical differences in PFS or OS by PD-L1 status. The 2- and 3-year landmark OS rates were 33% and 24%, respectively. CONCLUSION: Carboplatin, nab-paclitaxel, and pembrolizumab are a safe and effective regimen for patients with both squamous and nonsquamous NSCLC. Although this study did not meet the prespecified endpoints, the median and landmark OS results are consistent with durable benefit of this regimen as seen in phase III trials for first-line treatment of advanced NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Carcinoma de Células Escamosas , Neoplasias Pulmonares , Adulto , Humanos , Carcinoma Pulmonar de Células não Pequenas/patologia , Carboplatina/farmacologia , Carboplatina/uso terapêutico , Antígeno B7-H1 , Neoplasias Pulmonares/patologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Paclitaxel , Carcinoma de Células Escamosas/tratamento farmacológico
7.
Chem Biol Interact ; 387: 110794, 2024 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-37951334

RESUMO

Ferroptosis is an iron-dependent cell death and affects efficacies of multiple antitumor regimens, showing a great potential in cancer therapy. Protein kinase D2 (PKD2) plays a crucial role in regulating necrosis and apoptosis. However, the relationship of PKD2 and ferroptosis is still elusive. In this study, we mainly analyzed the roles of PKD2 on ferroptosis and chemotherapy in lung adenocarcinoma (LUAD). We found PKD2 was highly expressed in LUAD and silencing PKD2 could promote erastin-induced reactive oxygen species (ROS), malondialdehyde (MDA) accumulation, intracellular iron content and LUAD cells death. Mechanistically, augmenting PKD2 could prevent autophagic degradation of ferritin, which could be impaired by bafilomycin A1. We further found that PKD2 overexpression would promote LC3B-II, p62/SQSTM1 accumulation and block autophagosome-lysosome fusion in a TFEB-independent manner, which could be impaired by bafilomycin A1. Bafilomycin A1 stimulation could weaken ferroptosis promotion by PKD2 abrogation. Silencing ferritin heavy chain-1 (FTH1) could reverse the resistance to ferroptosis by PKD2 overexpression. Additionally, in vitro and vivo experiments validated PKD2 promoted proliferation, migration and invasion of LUAD cells. PKD2 knockdown or pharmacological inhibition by CRT0066101 could enhance efficacy of carboplatin in LUAD via ferroptosis and apoptosis. Collectively, our study revealed that abrogation of PKD2 could aggravate ferritinophagy-mediated ferroptosis by promoting autophagosome-lysosome fusion and enhance efficacy of carboplatin in LUAD. Targeting PKD2 to induce ferroptosis may be a promising strategy for LUAD therapy.


Assuntos
Adenocarcinoma de Pulmão , Carboplatina , Ferroptose , Neoplasias Pulmonares , Humanos , Adenocarcinoma de Pulmão/tratamento farmacológico , Autofagossomos/metabolismo , Autofagia , Carboplatina/farmacologia , Carboplatina/uso terapêutico , Ferro/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Lisossomos/metabolismo , Proteína Quinase D2 , Proteínas Serina-Treonina Quinases/metabolismo
8.
J Pathol ; 262(2): 198-211, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37941520

RESUMO

Carboplatin (CPT) and paclitaxel (PCT) are the optimal non-surgical treatment of epithelial ovarian cancer (EOC). Although their growth-restricting influence on EOC cells is well known, their impact on normal peritoneal cells, including mesothelium (PMCs) and fibroblasts (PFBs), is poorly understood. Here, we investigated whether, and if so, by what mechanism, CPT and PCT induce senescence of omental PMCs and PFBs. In addition, we tested whether PMC and PFB exposure to the drugs promotes the development of a pro-cancerogenic phenotype. The results showed that CPT and PCT induce G2/M growth arrest-associated senescence of normal peritoneal cells and that the strongest induction occurs when the drugs act together. PMCs senesce telomere-independently with an elevated p16 level and via activation of AKT and STAT3. In PFBs, telomeres shorten along with an induction of p21 and p53, and their senescence proceeds via the activation of ERK1/2. Oxidative stress in CPT + PCT-treated PMCs and PFBs is extensive and contributes causatively to their premature senescence. Both PMCs and PFBs exposed to CPT + PCT fuel the proliferation, migration, and invasion of established (A2780, OVCAR-3, SKOV-3) and primary EOCs, and this activity is linked with an overproduction of multiple cytokines altering the cancer cell transcriptome and controlled by p38 MAPK, NF-κB, STAT3, Notch1, and JAK1. Collectively, our findings indicate that CPT and PCT lead to iatrogenic senescence of normal peritoneal cells, which paradoxically and opposing therapeutic needs alters their phenotype towards pro-cancerogenic. It cannot be excluded that these adverse outcomes of chemotherapy may contribute to EOC relapse in the case of incomplete tumor eradication and residual disease initiation. © 2023 The Pathological Society of Great Britain and Ireland.


Assuntos
Neoplasias Ovarianas , Paclitaxel , Humanos , Feminino , Carboplatina/farmacologia , Paclitaxel/farmacologia , Linhagem Celular Tumoral , Apoptose , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Senescência Celular , Recidiva Local de Neoplasia/patologia , Epitélio/patologia , Carcinoma Epitelial do Ovário/patologia , Fibroblastos/patologia
9.
Pharmacol Res Perspect ; 11(6): e01152, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38100640

RESUMO

Cannabinoids have been shown to induce anti-tumor activity in a variety of carcinoma cells such as breast, prostate, and brain. The aim of the present study is to investigate the anti-tumor activity of cannabinoids, CBD (cannbidiol), and CBG (cannabigerol) in ovarian carcinoma cells sensitive and resistant to chemotherapeutic drugs. Sensitive A2780 cells and resistant A2780/CP70 carcinoma cells and non-carcinoma cells were exposed to varying concentrations of CBD, CBG, carboplatin or CB1 and CB2 receptor antagonists, AM251 and AM630, respectively, alone or in combination, at different exposure times and cytotoxicity was measured by MTT assay. The mechanism of action of CBD and CB in inducing cytotoxicity was investigated involving a variety of apoptotic and cell cycle assays. Treatment with CBD and CBG selectively, dose and time dependently reduced cell viability and induced apoptosis. The effect of CBD was stronger than CBG in all cell lines tested. Both CBD and CBG induced stronger cytotoxicity than afforded by carboplatin in resistant cells. The cytotoxicity induced by CBD was not CB1 or CB2 receptor dependent in both carcinoma cells, however, CBG-induced cytotoxicity may involve CB1 receptor activity in cisplatin-resistant carcinoma cells. A synergistic effect was observed when cannabinoids at sublethal doses were combined with carboplatin in both carcinoma cells. The apoptotic event may involve loss of mitochondrial membrane potential, Annexin V, caspase 3/7, ROS activities, and cell cycle arrest. Further studies are required to investigate whether these results are translatable in the clinic. Combination therapies with conventional cancer treatments using cannabinoids are suggested.


Assuntos
Canabinoides , Carcinoma , Neoplasias Ovarianas , Masculino , Humanos , Feminino , Linhagem Celular Tumoral , Carboplatina/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Canabinoides/farmacologia , Carcinoma/tratamento farmacológico
10.
Asian Pac J Cancer Prev ; 24(11): 3985-3991, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-38019259

RESUMO

OBJECTIVE: Cancer poses a significant challenge in modern medicine, standing as the primary cause of death in many countries, second only to cardiovascular diseases. Among the various treatments available, carboplatin, a chemotherapy drug, is employed for specific cancer types, including brain carcinoma. The main objective of this investigation is to enhance the therapeutic efficacy of carboplatin by utilizing niosomal nanocarriers. METHODS: We synthesized nanoniosomal carboplatin using the reverse-phase evaporation technique and conducted an assessment of its particle size, zeta potential, and drug-release properties. Subsequently, we evaluated the cytotoxicity of nanoniosomal carboplatin using the C6 rat glioma cell line. RESULTS: Our research revealed that these niosomal nanoparticles possessed a particle size of 290.5±5.5 nm and a zeta potential of -21.7±7.4 mV. The amount of encapsulated drug and drug loading level were found to be 60.2±2.3% and 2.5±1.1%, respectively. Importantly, the cytotoxic impact of these nanoniosomes on the C6 rat glioma cell line exhibited a significant increase compared to the free drug (P<0.05). CONCLUSION: Based on our discoveries, it is evident that carboplatin niosomal nanocarriers hold potential as an innovative approach to chemotherapy for brain cancer therapy.


Assuntos
Neoplasias Encefálicas , Glioma , Nanopartículas , Animais , Ratos , Carboplatina/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Linhagem Celular
11.
Genes (Basel) ; 14(11)2023 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-38003016

RESUMO

The precise mechanism of resistance to anti-cancer drugs such as platinum drugs is not fully revealed. To reveal the mechanism of drug resistance, the molecular networks of anti-cancer drugs such as cisplatin, carboplatin, oxaliplatin, and arsenic trioxide were analyzed in several types of cancers. Since diffuse-type stomach adenocarcinoma, which has epithelial-mesenchymal transition (EMT)-like characteristics, is more malignant than intestinal-type stomach adenocarcinoma, the gene expression and molecular networks in diffuse- and intestinal-type stomach adenocarcinomas were analyzed. Analysis of carboplatin revealed the causal network in diffuse large B-cell lymphoma. The upstream regulators of the molecular networks of cisplatin-treated lung adenocarcinoma included the anti-cancer drug trichostatin A (TSA), a histone deacetylase inhibitor. The upstream regulator analysis of cisplatin revealed an increase in FAS, BTG2, SESN1, and CDKN1A, and the involvement of the tumor microenvironment pathway. The molecular networks were predicted to interact with several microRNAs, which may contribute to the identification of new drug targets for drug-resistant cancer. Analysis of oxaliplatin, a platinum drug, revealed that the SPINK1 pancreatic cancer pathway is inactivated in ischemic cardiomyopathy. The study showed the importance of the molecular networks of anti-cancer drugs and tumor microenvironment in the treatment of cancer resistant to anti-cancer drugs.


Assuntos
Adenocarcinoma , Antineoplásicos , Proteínas Imediatamente Precoces , MicroRNAs , Humanos , Cisplatino , Carboplatina/farmacologia , Platina/farmacologia , Platina/uso terapêutico , Oxaliplatina/uso terapêutico , MicroRNAs/genética , MicroRNAs/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Microambiente Tumoral , Inibidor da Tripsina Pancreática de Kazal , Proteínas Supressoras de Tumor
12.
Gynecol Oncol ; 178: 69-79, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37806229

RESUMO

OBJECTIVE: Assess if MEK inhibitor blockade of RAS-ERK pathway adaptive response in high grade serous ovarian cancers (HGSOC) improves platinum sensitivity. METHODS: Three HGSOC cell lines and three patient derived organoid (PDOs) samples from ascites of platinum resistant HGSOC patients were collected. Cell lines and PDOs were exposed to carboplatin and MEK inhibitors cobimetinib or trametinib. Cytotoxic effects of MEK inhibitors alone or combined with carboplatin were established. Western blots demonstrated RAS-ERK pathway blockage after MEK inhibitor treatment. RNA sequencing assessed gene expression after MEK inhibitor treatment. Cell line NF1 gene knockdown was performed with corresponding chemosensitivity levels. RESULTS: High carboplatin IC50 levels indicated platinum resistance in cell lines and PDOs. Cobimetinib induced cytotoxicity in cell lines and PDOs, while trametinib was less effective. Western blot confirmed MEK-ERK pathway blockage at minimal concentrations of MEK inhibitors in cell lines and PDOs. Phosphorylated-ERK levels of untreated cells indicated higher levels of RAS-ERK pathway activation in OVSAHO and OVCAR7 compared to OVCAR3. OVSAHO harbors a NF1 mutation and had highest levels of RAS-ERK activation. Cotreatment with carboplatin and MEK inhibitors showed varying synergistic cytotoxic effects at different combinations. Synergistic effect was most prominent in the OVSAHO carboplatin and cobimetinib combination. RNA sequencing identified downregulation of c-MYC and FOXM1 gene expression after MEK inhibitor treatment. NF1 gene knockdown showed an acquired increased IC50 compared to parental cells. CONCLUSION: MEK inhibitors block RAS-ERK pathways in platinum resistant HGSOC cells and PDOs. MEK inhibitors with carboplatin have select synergistic effects which may indicate a strategy to improve platinum sensitivity.


Assuntos
Antineoplásicos , Neoplasias Ovarianas , Humanos , Feminino , Sistema de Sinalização das MAP Quinases/fisiologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Carboplatina/farmacologia , Carboplatina/uso terapêutico , Apoptose , Linhagem Celular Tumoral , Antineoplásicos/uso terapêutico , Inibidores de Proteínas Quinases/farmacologia , Carcinoma Epitelial do Ovário/tratamento farmacológico , Quinases de Proteína Quinase Ativadas por Mitógeno
13.
Nat Commun ; 14(1): 5346, 2023 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-37660083

RESUMO

Chimeric antigen receptor (CAR) T cells have transformed the treatment landscape for hematological malignancies. However, CAR T cells are less efficient against solid tumors, largely due to poor infiltration resulting from the immunosuppressive nature of the tumor microenvironment (TME). Here, we assessed the efficacy of Lewis Y antigen (LeY)-specific CAR T cells in patient-derived xenograft (PDX) models of prostate cancer. In vitro, LeY CAR T cells directly killed organoids derived from androgen receptor (AR)-positive or AR-null PDXs. In vivo, although LeY CAR T cells alone did not reduce tumor growth, a single prior dose of carboplatin reduced tumor burden. Carboplatin had a pro-inflammatory effect on the TME that facilitated early and durable CAR T cell infiltration, including an altered cancer-associated fibroblast phenotype, enhanced extracellular matrix degradation and re-oriented M1 macrophage differentiation. In a PDX less sensitive to carboplatin, CAR T cell infiltration was dampened; however, a reduction in tumor burden was still observed with increased T cell activation. These findings indicate that carboplatin improves the efficacy of CAR T cell treatment, with the extent of the response dependent on changes induced within the TME.


Assuntos
Fibroblastos Associados a Câncer , Neoplasias da Próstata , Masculino , Animais , Humanos , Carboplatina/farmacologia , Carboplatina/uso terapêutico , Microambiente Tumoral , Linfócitos T , Neoplasias da Próstata/tratamento farmacológico , Modelos Animais de Doenças
14.
Cell Mol Life Sci ; 80(10): 294, 2023 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-37718345

RESUMO

Resistance to platinum- and taxane-based chemotherapy represents a major obstacle to long-term survival in ovarian cancer (OC) patients. Here, we studied the interplay between acquired carboplatin (CBP) resistance using two OC cell models, MES-OV CBP and SK-OV-3 CBP, and non-P-glycoprotein-mediated cross-resistance to paclitaxel (TAX) observed only in MES-OV CBP cells. Decreased platination, mesenchymal-like phenotype, and increased expression of α- and γ-tubulin were observed in both drug-resistant variants compared with parental cells. Both variants revealed increased protein expression of class III ß-tubulin (TUBB3) but differences in TUBB3 branching and nuclear morphology. Transient silencing of TUBB3 sensitized MES-OV CBP cells to TAX, and surprisingly also to CBP. This phenomenon was not observed in the SK-OV-3 CBP variant, probably due to the compensation by other ß-tubulin isotypes. Reduced TUBB3 levels in MES-OV CBP cells affected DNA repair protein trafficking and increased whole-cell platination level. Furthermore, TUBB3 depletion augmented therapeutic efficiency in additional OC cells, showing vice versa drug-resistant pattern, lacking ß-tubulin isotype compensation visible at the level of total ß-tubulin (TUBB) in vitro and ex vivo. In summary, the level of TUBB in OC should be considered together with TUBB3 in therapy response prediction.


Assuntos
Neoplasias Ovarianas , Tubulina (Proteína) , Humanos , Feminino , Carboplatina/farmacologia , Carboplatina/uso terapêutico , Regulação para Cima , Tubulina (Proteína)/genética , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Ativação Transcricional
15.
Farm. hosp ; 47(5): 183-189, Septiembre - Octubre 2023. tab, ilus
Artigo em Inglês, Espanhol | IBECS | ID: ibc-225605

RESUMO

Objetivo: en 2016 se publicaron las guías de la MASCC/ESMO que incorporaron los esquemas de antraciclinas como quimioterapia altamente emetógena (QAE) proponiendo la triple terapia antiemética, así como para los esquemas de carboplatino. Los objetivos fueron analizar el nivel de concordancia entre las guías y la profilaxis antiemética utilizada en el hospital de día de hematooncología, evaluar su efectividad y determinar el ahorro de la inclusión de netupitant/palonosetrón (NEPA) oral con dexametasona intravenosa (NEPAd) respecto a fosaprepitant con ondansetrón y dexametasona (FOD intravenosa). Método estudio observacional prospectivo registrando variables demográficas, esquema de quimioterapia recibido, localización tumoral, riesgo emetógeno del paciente, pauta antiemética prescrita, concordancia con guía MASCC/ESMO y su efectividad, utilización de medicación de rescate y registro de visitas a urgencias o ingresos por emesis.Se llevó a cabo un estudio farmacoeconómico de minimización de costes. Resultados se incluyeron 61 pacientes, 70% mujeres, mediana edad 60,5.Los esquemas de platino fueron más frecuentes en el periodo 1, siendo el 87,5% respecto al 67,6% en el periodo 2. Los esquemas con antraciclinas fueron del 21,6 y 10% respectivamente en cada periodo. Un 21,1% de las pautas antieméticas no coincidían con las recomendaciones MASCC/ESMO, siendo en su totalidad en el periodo 1. La puntuación de los cuestionarios de efectividad fue de protección total en el 90,9% en las náuseas agudas, del 100% en los vómitos agudos y en las náuseas retardadas, y del 72,7% en los vómitos retardados. La frecuencia de uso de medicación de rescate fue del 18,7% en el periodo 1 y no fue necesaria en el periodo 2.No se detectaron visitas a urgencias ni ingresos en ninguno de los periodos. El uso de NEPAd comportó una reducción del 28% de los costes con respecto al empleo de FOD. Conclusiones: ... (AU)


Objective: Latest MASCC/ESMO guidelines of the recommendations for the prophylaxis of acute and delayed emesis induced by moderately emetogenic chemotherapy was published in 2016 incorporating anthracycline schemes as highly emetogenic chemotherapy (HEC), proposing triple antiemetic therapy to control nausea and vomiting. Likewise, they recommend triple therapy for carboplatin. The objectives of this study were to analyze the degree of concordance between guidelines and antiemetic prophylaxis used in the Chemotherapy Outpatient Unit in patients undergoing treatment with HEC and carboplatin, to evaluate its effectiveness and to determine the savings due to the use of netupitant/palonosetron (NEPA) oral (or) with intravenous (iv) dexamethasone (NEPAd) compared to iv Fosaprepitant with ondansetron and dexamethasone (FOD iv).MethodsProspective observational study recording demographic variables, chemotherapy protocol, tumor location, patient emetogenic risk, antiemetic regimen prescribed, concordance with the MASCC/ESMO guideline, and effectiveness, evaluated by MASCC survey, use of rescue medication and visits to the Emergency Department or hospitalization due to emesis.A cost minimization pharmacoeconomic study was carried out. Results 61 patients were included; 70% women; median age 60.5. Platinum schemes were more frequent in period 1, being 87.5% compared to 67.6% in period 2. Anthracycline schemes were 21.6% and 10% respectively in each period.A 21.1% of the antiemetic regimens did not coincide with the MASCC/ESMO recommendations, being entirely in period 1. The score of the effectiveness questionnaires was total protection in 90.9% in acute nausea, from 100% in acute vomiting and delayed nausea, and 72.7% in delayed vomiting.The frequency of use of rescue medication was 18.7% in period 1 and was not necessary in period 2.No visits to the emergency room or admissions were detected in any of the periods. Conclusions: ...(AU)


Assuntos
Humanos , Masculino , Feminino , Idoso , Antieméticos/administração & dosagem , Antieméticos/farmacologia , Antieméticos/uso terapêutico , Carboplatina/farmacologia , Antraciclinas/farmacologia , Guias de Prática Clínica como Assunto , Estudos Prospectivos , Custos e Análise de Custo
16.
Nature ; 620(7973): 409-416, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37532934

RESUMO

Netrin-1 is upregulated in cancers as a protumoural mechanism1. Here we describe netrin-1 upregulation in a majority of human endometrial carcinomas (ECs) and demonstrate that netrin-1 blockade, using an anti-netrin-1 antibody (NP137), is effective in reduction of tumour progression in an EC mouse model. We next examined the efficacy of NP137, as a first-in-class single agent, in a Phase I trial comprising 14 patients with advanced EC. As best response we observed 8 stable disease (8 out of 14, 57.1%) and 1 objective response as RECIST v.1.1 (partial response, 1 out of 14 (7.1%), 51.16% reduction in target lesions at 6 weeks and up to 54.65% reduction during the following 6 months). To evaluate the NP137 mechanism of action, mouse tumour gene profiling was performed, and we observed, in addition to cell death induction, that NP137 inhibited epithelial-to-mesenchymal transition (EMT). By performing bulk RNA sequencing (RNA-seq), spatial transcriptomics and single-cell RNA-seq on paired pre- and on-treatment biopsies from patients with EC from the NP137 trial, we noted a net reduction in tumour EMT. This was associated with changes in immune infiltrate and increased interactions between cancer cells and the tumour microenvironment. Given the importance of EMT in resistance to current standards of care2, we show in the EC mouse model that a combination of NP137 with carboplatin-paclitaxel outperformed carboplatin-paclitaxel alone. Our results identify netrin-1 blockade as a clinical strategy triggering both tumour debulking and EMT inhibition, thus potentially alleviating resistance to standard treatments.


Assuntos
Neoplasias do Endométrio , Transição Epitelial-Mesenquimal , Netrina-1 , Animais , Feminino , Humanos , Camundongos , Biópsia , Carboplatina/administração & dosagem , Carboplatina/farmacologia , Carboplatina/uso terapêutico , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/imunologia , Neoplasias do Endométrio/patologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Perfilação da Expressão Gênica , Netrina-1/antagonistas & inibidores , Paclitaxel/administração & dosagem , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , RNA-Seq , Análise da Expressão Gênica de Célula Única , Microambiente Tumoral/efeitos dos fármacos
17.
Biomed Pharmacother ; 166: 115349, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37634476

RESUMO

BACKGROUND: DNA methylation, histone modifications, and miRNAs affect ovarian cancer (OC) progression and therapy response. PURPOSE: Identification of epigenetically downregulated miRNAs in drug-resistant OC cell lines with a possible role in drug resistance and/or drug-induced mesenchymal-like phenotype. METHODS: MiRNA profiling was performed on parental and carboplatin-resistant OC cells, MES-OV and MES-OV CBP. RT-qPCR validation, epigenetic modulation and other CBP-resistant OC cell lines were used to select miRNAs of interest. The integration of miRNA-predicted target genes and differentially expressed genes (DEGs), pathway and functional analysis were used for forecasting their biological role. Data mining was performed to determine their possible prognostic and predictive values. RESULTS: MiRNA profiling revealed 48 downregulated miRNAs in OC cells whose drug sensitivity and metastatic potential were impacted by epigenetic modulators. Of the fourteen selected, nine were validated as changed, and seven of these restored their expression upon treatment with epigenetic inhibitors. Only three had similar expression patterns in other OC cell lines. MiRNA-mRNA integrative analysis resulted in 56 target DEGs. Pathway analysis revealed that these genes are involved in cell adhesion, migration, and invasion. The functional analysis confirmed the role of miR-103a-3p, miR-17-5p and miR-107 in cell invasion, while data mining showed their prognostic and predictive values. Only miR-103a-3p was epigenetically regulated at the constitutive level. CONCLUSION: High throughput miRNA and cDNA profiling coupled with pathway analysis and data mining delivered evidence for miRNAs which can be epigenetically regulated in drug-resistant, mesenchymal-like OC cells as possible markers to combat therapy-induced short overall survival and tumor metastatic potential.


Assuntos
Neoplasias Ovarianas , Feminino , Humanos , Carboplatina/farmacologia , Prognóstico , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Fenótipo
18.
Eur Rev Med Pharmacol Sci ; 27(13): 6343-6350, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37458651

RESUMO

OBJECTIVE: This study aimed to determine the role of oxidative stress (OS) in carboplatin-induced gonadotoxicity and whether Nigella Sativa oil (NSO), an herbal antioxidant, has a protective effect on ovarian apoptosis, OS, and the anti-Müllerian hormone (AMH) level in a rat model. MATERIALS AND METHODS: The study included 24 adult female rats that were divided into 4 treatment groups. Group A saline + saline (sham group); group B: NSO + saline; group C: saline + carboplatin; group D: NSO + carboplatin. Saline, NSO, and carboplatin were administered intraperitoneally 24 and/or 48 h before sacrification as 4 mL/kg, 4 mL/kg, and 80 mg/kg, respectively. Apoptosis, OS parameters, and AMH were measured. RESULTS: Oxidant levels and apoptosis were higher, whereas AMH and the antioxidants were lower in group C than in group A. Apoptosis, OS parameters, and AMH levels were negatively affected by chemotherapy (CTx) in group C whilst improvement in those parameters was observed in group D following NSO pretreatment. The levels of apoptosis and malondialdehyde (MDA), an OS parameter, in group D were lower than in group C as they declined from 34.3% to 8.65% (p = 0.002) and from 199.4 nmol/g tissue to 136.4 nmol/g tissue (p = 0.002), respectively. However, the slight increase in AMH level from 2.7 ng/mL to 3.5 ng/mL due to the NSO effect was not significant between groups C and D. CONCLUSIONS: The present findings show that carboplatin has adverse effects on AMH, ovarian tissue apoptosis, and OS parameters. NSO pretreatment might protect ovarian tissue and decrease CTx-induced ovarian injury by decreasing OS and apoptosis, but the protective effect of NSO on AMH is limited.


Assuntos
Antineoplásicos , Nigella sativa , Ratos , Feminino , Animais , Ratos Wistar , Hormônio Antimülleriano/farmacologia , Carboplatina/farmacologia , Estresse Oxidativo , Óleos de Plantas/farmacologia , Óleos de Plantas/uso terapêutico , Antioxidantes/farmacologia , Antineoplásicos/toxicidade
19.
Biochim Biophys Acta Gen Subj ; 1867(10): 130439, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37516256

RESUMO

Programmed death-ligand-1 (PD-L1) is an immune suppressor that inhibits T cell based immunity. Anti-PD-L1/PD-1 immunotherapy benefits those patients receiving platinum-based combinational chemotherapy. However, the underlying mechanism is still largely unknown. In this study, we found that carboplatin could induce PD-L1 expression in NSCLC H292, A549 and H1299 cells in a dose-dependent manner. mRNA sequencing and the subsequent validation assays found that carboplatin significantly induced PVR expression, which is considered as an immuno-adhesion molecule. Mechanistically, PVR knockdown significantly abrogated carboplatin-induced PD-L1 expression. Functionally, knockdown of PVR significantly reversed the CD3+ T cells proliferation inhibition caused by carboplatin increased PD-L1. Moreover, the carboplatin-induced PVR and subsequent up-regulation of PD-L1 might be mediated via the EGFR, PI3K/AKT, and ERK signaling pathways. Immunohistochemical staining results showed that the PD-L1 expression was positively associated with PVR expression in clinical NSCLC samples. Our study reveals a novel regulatory mechanism of PD-L1 expression, provides evidence that carboplatin inhibits tumor immune response by up-regulating PD-L1 expression and explains the rationale for combining platinum-based chemotherapy with PD-L1/PD-1 inhibitors.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Carboplatina/farmacologia , Carboplatina/uso terapêutico , Neoplasias Pulmonares/patologia , Fosfatidilinositol 3-Quinases
20.
J Ovarian Res ; 16(1): 147, 2023 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-37501162

RESUMO

BACKGROUND: A recent study reported the role of long non-coding RNA (lncRNA) RBAT1 in promoting the development of retinoblastoma and bladder cancer. However, its function in other cancers is unclear. We then studied the role of RBAT1 in endometrial carcinoma (EC). METHODS: The expression of RBAT1 and miR-27b in EC and paired non-tumor samples from advanced EC patients, as well as in plasma samples of EC patients and healthy controls were detected by RT-qPCR. The direct interaction between RBAT1 and miR-27b, and the subcellular location of RBAT1 were determined by RNA-RNA pulldown assay and subcellular fractionation assay, respectively. RESULTS: EC tissues showed increased expression levels of RBAT1 and decreased expression levels of miR-27b compared to that in non-tumor tissues. Moreover, EC patients showed higher plasma expression levels of RBAT1 and lower plasma expression levels of miR-27b compared to that in the controls. Drug-resistant (DR) patients showed higher expression levels of RBAT1 and lower expression levels of miR-27b in both EC tissues and plasma samples. RBAT1 was detected in both nuclear and cytoplasm and it directly interacted with miR-27b. RBAT1 and miR-27b did not affect the expression of each other. Upregulation of RBAT1 promoted the expression of multidrug-resistant-related protein (P-gp, MRP1, and BCRP). Overexpression of RBAT1 and inhibition of miR-27b promoted cell viability and impeded cell apoptosis and cell cycle arrest at G0-G1 phase, while knockdown of RBAT1 and overexpression of miR-27b inhibited cell viability and induced cell apoptosis and cell cycle arrest at G0-G1 phase. Moreover, miR-27b could abolish RBAT1-induced effects on cell viability, apoptosis and cell cycle. CONCLUSION: RBAT1 may reduce the chemosensitivity of EC cells to carboplatin/paclitaxel by sponging miR-27b in EC.


Assuntos
Neoplasias do Endométrio , MicroRNAs , RNA Longo não Codificante , Feminino , Humanos , Apoptose/genética , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Carboplatina/farmacologia , Carboplatina/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas de Neoplasias/genética , Paclitaxel/farmacologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...