Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.317
Filtrer
1.
PLoS One ; 19(8): e0306900, 2024.
Article de Anglais | MEDLINE | ID: mdl-39110673

RÉSUMÉ

BACKGROUND: Non-metastatic castration-resistant prostate cancer (nmCRPC) is an asymptomatic condition with the potential to progress to metastasis. Novel hormonal agents (NHAs) are currently considered the gold standard treatment for nmCRPC, offering significant survival benefits. However, further evidence is needed to determine whether there are differences in the performance of these drugs among Asian populations. METHODS: This retrospective analysis of nmCRPC patients aims to compare the efficacy and safety of three NHAs-apalutamide, darolutamide, and enzalutamide. Data were collected from two prominent prostate care centers in Taichung, Taiwan. Patient characteristics, treatment details, PSA responses, and adverse events were analyzed. Statistical comparisons were performed, and the study received Institutional Review Board approval. RESULTS: Total of 64 patients were recruited in this study, including 29 darolutamide, 26 apalutamide, and 9 enzalutamide patients. Baseline characteristics varied between the three patient groups, but the treatment response still revealed similar results. The apalutamide group experienced more adverse events, notably skin rash. Discontinuation rates due to adverse events differed among the groups, and patients receiving darolutamide were less likely to discontinue treatment. CONCLUSION: This real-world study provides insights into NHA utilization in nmCRPC within the Taiwanese population. Adverse event profiles varied, emphasizing the need for individualized treatment decisions. The study underscores the importance of regional considerations and contributes valuable data for optimizing treatment outcomes in nmCRPC.


Sujet(s)
Benzamides , Nitriles , 3-Phényl-2-thiohydantoïne , Tumeurs prostatiques résistantes à la castration , Thiohydantoïnes , Humains , Mâle , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Sujet âgé , 3-Phényl-2-thiohydantoïne/usage thérapeutique , Taïwan , Benzamides/usage thérapeutique , Nitriles/usage thérapeutique , Études rétrospectives , Adulte d'âge moyen , Thiohydantoïnes/usage thérapeutique , Thiohydantoïnes/effets indésirables , Sujet âgé de 80 ans ou plus , Pyrazoles/usage thérapeutique , Pyrazoles/effets indésirables , Antinéoplasiques hormonaux/usage thérapeutique , Résultat thérapeutique , Antigène spécifique de la prostate/sang
2.
Nat Commun ; 15(1): 6672, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39107274

RÉSUMÉ

Castration-resistant prostate cancer (CRPC) is a frequently occurring disease with adverse clinical outcomes and limited therapeutic options. Here, we identify methionine adenosyltransferase 2a (MAT2A) as a critical driver of the androgen-indifferent state in ERG fusion-positive CRPC. MAT2A is upregulated in CRPC and cooperates with ERG in promoting cell plasticity, stemness and tumorigenesis. RNA, ATAC and ChIP-sequencing coupled with histone post-translational modification analysis by mass spectrometry show that MAT2A broadly impacts the transcriptional and epigenetic landscape. MAT2A enhances H3K4me2 at multiple genomic sites, promoting the expression of pro-tumorigenic non-canonical AR target genes. Genetic and pharmacological inhibition of MAT2A reverses the transcriptional and epigenetic remodeling in CRPC models and improves the response to AR and EZH2 inhibitors. These data reveal a role of MAT2A in epigenetic reprogramming and provide a proof of concept for testing MAT2A inhibitors in CRPC patients to improve clinical responses and prevent treatment resistance.


Sujet(s)
Épigenèse génétique , Régulation de l'expression des gènes tumoraux , Methionine adenosyltransferase , Tumeurs prostatiques résistantes à la castration , Régulateur transcriptionnel ERG , Mâle , Humains , Régulateur transcriptionnel ERG/génétique , Régulateur transcriptionnel ERG/métabolisme , Methionine adenosyltransferase/génétique , Methionine adenosyltransferase/métabolisme , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Épigenèse génétique/effets des médicaments et des substances chimiques , Animaux , Androgènes/métabolisme , Épigénome , Souris , Histone/métabolisme , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Protéine-2 homologue de l'activateur de Zeste/génétique , Protéine-2 homologue de l'activateur de Zeste/antagonistes et inhibiteurs
3.
JAMA Netw Open ; 7(8): e2429783, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39190308

RÉSUMÉ

Importance: Novel androgen receptor inhibitors (ARIs; darolutamide, enzalutamide, and apalutamide) are standard-of-care treatments for nonmetastatic castration-resistant prostate cancer (nmCRPC). However, there are sparse data comparing their clinical use and tolerability. Objective: To compare clinical use and outcomes for darolutamide, enzalutamide, and apalutamide in patients with nmCRPC. Design, Setting, and Participants: This retrospective cohort study reviewed electronic medical records from the Precision Point Specialty network of US urology practices. Eligible patients had nmCRPC and no prior novel hormonal therapy and initiated novel ARI treatment between August 1, 2019, and March 31, 2022. Data were analyzed from February 1, 2019, to December 31, 2022. Exposures: Patients were prescribed darolutamide, enzalutamide, or apalutamide as their first novel ARI for nmCRPC. Main Outcomes and Measures: The main outcome was a composite of 2 end points, treatment discontinuation and progression to metastatic CRPC (mCRPC), whichever occurred first. Both end points were also assessed separately. Results: All 870 patients meeting eligibility criteria were included (362 receiving darolutamide [41.6%]; 382, enzalutamide [43.9%]; 126, apalutamide [14.5%]); mean (SD) age was 78.8 (8.7) years. Self-reported race was Black or African American in 187 patients (21.5%), White in 585 (67.2%), and other or unknown in 98 (11.3%). The darolutamide cohort had lower proportions of patients with a composite end point event (134 [37.0%] vs 201 [52.6%] for enzalutamide and 66 [52.4%] for apalutamide), discontinuation (110 [30.4%] for darolutamide vs 156 [40.8%] for enzalutamide and 58 [46.0%] for apalutamide), and progression to mCRPC (64 [17.7%] for darolutamide vs 108 [28.3%] for enzalutamide and 35 [27.8%] for apalutamide) during the study period. After adjusting for baseline covariates, patients receiving darolutamide had a lower risk of a composite end point event compared with enzalutamide (risk reduction, 33.8%; hazard ratio [HR], 0.66 [95% CI, 0.53-0.84]) and apalutamide (risk reduction, 35.1%; HR, 0.65 [95% CI, 0.48-0.88]). Similarly, patients receiving darolutamide had a lower risk of discontinuation compared with enzalutamide (risk reduction, 27.4%; HR, 0.73 [95% CI, 0.56-0.94]) and apalutamide (risk reduction, 39.1%; HR, 0.61 [95% CI, 0.44-0.85]) and a lower risk of progression to mCRPC compared with enzalutamide (risk reduction, 40.6%; HR, 0.59 [95% CI, 0.43-0.82]) and apalutamide (risk reduction, 35.3%; HR, 0.65 [95% CI, 0.42-0.99]). There was no difference between enzalutamide and apalutamide treatment across outcomes. Conclusions and Relevance: In this large cohort study of patients with nmCRPC treated with novel ARIs, results suggest better tolerability for darolutamide compared with enzalutamide and apalutamide, which may be associated with a clinical effectiveness advantage. Comparative clinical studies are needed to guide treatment decisions in the absence of head-to-head clinical trials.


Sujet(s)
Antagonistes du récepteur des androgènes , Benzamides , Nitriles , 3-Phényl-2-thiohydantoïne , Tumeurs prostatiques résistantes à la castration , Thiohydantoïnes , Humains , Mâle , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Sujet âgé , Études rétrospectives , Antagonistes du récepteur des androgènes/usage thérapeutique , 3-Phényl-2-thiohydantoïne/usage thérapeutique , Benzamides/usage thérapeutique , Nitriles/usage thérapeutique , Thiohydantoïnes/usage thérapeutique , Pyrazoles/usage thérapeutique , Sujet âgé de 80 ans ou plus , Adulte d'âge moyen
4.
Sci Rep ; 14(1): 19895, 2024 08 27.
Article de Anglais | MEDLINE | ID: mdl-39191899

RÉSUMÉ

Although overall survival data are still premature, the PROpel study found radiological progression-free survival (PFS) benefits of abiraterone and olaparib in patients with metastatic castration-resistant prostate cancer (mCRPC). However, for patients who have not been genetically tested or lack BRCA1/2 mutations (BRCAm), this combination therapy has been questioned as a first-line conventional treatment for mCRPC, mainly due to significant health economics and side effects. In our retrospective study, we found that treatment with low-dose abiraterone plus olaparib as a late-line treatment for mCRPC could lead to prostate-specific antigen (PSA) and symptom PFS in selective cases even without BRCAm. The median PSA-PFS was 8 months (IQR: 6.5-11.5), with a median follow-up duration of 39.0 months (IQR: 27.5-64.5). Gene tests were conducted in all patients, identifying non-BRCA mutations through ctDNA testing (24%), tumor tissue testing (12%), or both (64%). Adverse events occurred in 72% of patients, with 16% experiencing Grade ≥ 3 events. Common adverse events included anemia (64%), decreased appetite (48%), and fatigue (25%). Our findings support low-dose abiraterone plus olaparib as a potential option for mCRPC patients without BRCAm, offering manageable safety and efficacy profiles.


Sujet(s)
Androstènes , Protocoles de polychimiothérapie antinéoplasique , Protéine BRCA1 , Protéine BRCA2 , Phtalazines , Pipérazines , Tumeurs prostatiques résistantes à la castration , Humains , Phtalazines/administration et posologie , Phtalazines/usage thérapeutique , Mâle , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Pipérazines/administration et posologie , Pipérazines/usage thérapeutique , Pipérazines/effets indésirables , Études rétrospectives , Sujet âgé , Adulte d'âge moyen , Protéine BRCA2/génétique , Androstènes/administration et posologie , Androstènes/usage thérapeutique , Projets pilotes , Protéine BRCA1/génétique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Mutation , Antigène spécifique de la prostate/sang , Sujet âgé de 80 ans ou plus , Survie sans progression
5.
Cells ; 13(16)2024 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-39195285

RÉSUMÉ

Neuroendocrine differentiation (NED) represents a possible androgen receptor pathway inhibitors (ARPI) resistance mechanism in metastatic castration resistance prostate cancer (mCRPC). As mCRPC with NED has been excluded from clinical trials evaluating ARPI efficacy, this study investigates the prognostic impact of NED in mCRPC patients treated with ARPIs. Methods: We retrospectively analyzed 327 mCRPC patient data treated with Enzalutamide or Abiraterone in the first and second or successive lines of treatment. NED was assessed using prostate biopsy samples through immunohistochemical staining. Results: NED was confirmed in 32/327 (9.8%) mCRPC patients. In the overall population, mCRPC with NED showed worse PFS (4.38 vs. 11.48 months HR 2.505 [1.71-3.68] p < 0.05), disease control rate (DCR), and PSA response. In the first line setting, mCRPC with NED demonstrated worse PFS (8.5 vs. 14.9 months HR 2.13 [1.18-3.88], p < 0.05). Similarly, in the second or successive lines, mCRPC with NED showed worse PFS (4.0 vs. 7.5 months HR 2.43 [1.45-4.05] p < 0.05), DCR, PSA response and OS (12.53 vs. 18.03 months HR 1.86 [1.12-3.10] p < 0.05). The adverse impact of NED on PFS was consistence across all subgroups; we also noted a trend of worse PFS in patients with high vs. low NED. Conclusions: In our study, mCRPC with NED treated with Enzalutamide or Abiraterone showed worse clinical outcomes. NED assessment should be considered to optimize treatment decisions in the mCRPC setting.


Sujet(s)
Androstènes , Benzamides , Nitriles , 3-Phényl-2-thiohydantoïne , Tumeurs prostatiques résistantes à la castration , Mâle , Humains , 3-Phényl-2-thiohydantoïne/usage thérapeutique , 3-Phényl-2-thiohydantoïne/pharmacologie , Nitriles/usage thérapeutique , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Études rétrospectives , Androstènes/usage thérapeutique , Androstènes/pharmacologie , Sujet âgé , Adulte d'âge moyen , Différenciation cellulaire/effets des médicaments et des substances chimiques , Métastase tumorale , Sujet âgé de 80 ans ou plus , Résultat thérapeutique , Cellules neuroendocrines/anatomopathologie , Cellules neuroendocrines/métabolisme , Cellules neuroendocrines/effets des médicaments et des substances chimiques
6.
Nat Commun ; 15(1): 6779, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39117665

RÉSUMÉ

Enhancer of zeste homolog 2 (EZH2) is a histone methyltransferase and emerging therapeutic target that is overexpressed in most castration-resistant prostate cancers and implicated as a driver of disease progression and resistance to hormonal therapies. Here we define the lineage-specific action and differential activity of EZH2 in both prostate adenocarcinoma and neuroendocrine prostate cancer (NEPC) subtypes of advanced prostate cancer to better understand the role of EZH2 in modulating differentiation, lineage plasticity, and to identify mediators of response and resistance to EZH2 inhibitor therapy. Mechanistically, EZH2 modulates bivalent genes that results in upregulation of NEPC-associated transcriptional drivers (e.g., ASCL1) and neuronal gene programs in NEPC, and leads to forward differentiation after targeting EZH2 in NEPC. Subtype-specific downstream effects of EZH2 inhibition on cell cycle genes support the potential rationale for co-targeting cyclin/CDK to overcome resistance to EZH2 inhibition.


Sujet(s)
Protéine-2 homologue de l'activateur de Zeste , Régulation de l'expression des gènes tumoraux , Tumeurs de la prostate , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Protéine-2 homologue de l'activateur de Zeste/génétique , Mâle , Humains , Lignée cellulaire tumorale , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Animaux , Adénocarcinome/génétique , Adénocarcinome/métabolisme , Adénocarcinome/anatomopathologie , Résistance aux médicaments antinéoplasiques/génétique , Différenciation cellulaire , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Souris , Lignage cellulaire
7.
Tidsskr Nor Laegeforen ; 144(9)2024 Aug 20.
Article de Anglais, Norvégien | MEDLINE | ID: mdl-39166985

RÉSUMÉ

Background: Treatment of castration-resistant metastatic prostate cancer with [¹77Lu]PSMA radioligand. Case presentation: A man in his seventies with metastatic prostate cancer received castration therapy for four years, developing castration-resistant disease. PET/CT with [68Ga]PSMA-11 showed high uptake in metastatic lymph nodes. The patient received 7.4 GBq [¹77Lu]PSMA-I&T (Curium, Finland) as five treatments at five-week intervals. Five weeks after the first treatment, p-PSA dropped from 154 to 53 µg/L. Five weeks after the fifth treatment, p-PSA was 1.8 µg/L. [68Ga]PSMA-11 PET/CT showed significant reduction in the size of metastases, with the largest decreasing in diameter from 10 to 4 mm. Seven months after the fifth treatment, p-PSA increased to 14.3 µg/L, and [68Ga]PSMA-11 PET/ CT revealed additional skeletal metastases, while the lymph node metastases remained unchanged. Thus, the treatment had a good but temporary effect on the metastases. Interpretation: Treatment with [¹77Lu]PSMA radioligand resulted in a temporary regression of the metastases.


Sujet(s)
Radio-isotopes du gallium , Lutétium , Tomographie par émission de positons couplée à la tomodensitométrie , Antigène spécifique de la prostate , Tumeurs prostatiques résistantes à la castration , Radiopharmaceutiques , Humains , Mâle , Tumeurs prostatiques résistantes à la castration/radiothérapie , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Radiopharmaceutiques/usage thérapeutique , Radiopharmaceutiques/pharmacocinétique , Antigène spécifique de la prostate/sang , Lutétium/usage thérapeutique , Sujet âgé , Métastase lymphatique , Dipeptides/usage thérapeutique , Dipeptides/pharmacocinétique , Oligopeptides , Isotopes du gallium
8.
Eur J Cancer ; 209: 114183, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39111209

RÉSUMÉ

BACKGROUND: MAGNITUDE (NCT03748641) demonstrated favourable outcomes with niraparib plus abiraterone acetate plus prednisone (+AAP) versus placebo+AAP in patients with BRCA1/2-altered metastatic castration-resistant prostate cancer (mCRPC). Imbalances in prognostic variables were reported between arms, which impacts estimation of both the clinical benefit and cost­effectiveness of niraparib+AAP for healthcare systems. A pre-specified multivariable analysis (MVA) demonstrated improved overall survival (OS) with niraparib+AAP. Here, we used an inverse probability of treatment weighting (IPTW) model to adjust for covariate imbalances and assess time-to-event outcomes. METHODS: IPTW analysis of time-to-event outcomes was conducted using data from patients with BRCA1/2-altered mCRPC (N = 225) in MAGNITUDE. Patients received niraparib+AAP or placebo+AAP. OS, radiographic progression-free survival, time to symptomatic progression, time to initiation of cytotoxic chemotherapy and time to prostate-specific antigen progression were assessed. Weighted Kaplan-Meier curves were generated for each endpoint, and adjusted hazard ratios (HR) were obtained from a weighted Cox model. RESULTS: Improvements in survival outcomes were estimated for niraparib+AAP versus placebo+AAP: unadjusted median OS was 30.4 months versus 28.6 months, respectively (HR: 0.79; 95 % confidence interval [CI]: 0.55, 1.12; p = 0.183). Following IPTW, median OS increased to 34.1 months with niraparib+AAP versus a decrease to 27.4 with placebo (HR: 0.65; 95 % CI: 0.46, 0.93; p = 0.017). Similar improvements were observed for other time-to-event endpoints. CONCLUSIONS: IPTW adjustment provided a more precise estimate of the clinical benefit of niraparib+AAP versus placebo+AAP in patients with BRCA1/2-altered mCRPC. Results were consistent with the pre-specified MVA, and further demonstrated the value of adjusting for baseline imbalances, particularly in smaller studies. TRIAL REGISTRATION: NCT03748641 (MAGNITUDE).


Sujet(s)
Acétate d'abiratérone , Protocoles de polychimiothérapie antinéoplasique , Indazoles , Pipéridines , Prednisone , Tumeurs prostatiques résistantes à la castration , Humains , Mâle , Indazoles/usage thérapeutique , Indazoles/administration et posologie , Prednisone/administration et posologie , Prednisone/usage thérapeutique , Pipéridines/usage thérapeutique , Pipéridines/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Acétate d'abiratérone/administration et posologie , Acétate d'abiratérone/usage thérapeutique , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/mortalité , Sujet âgé , Adulte d'âge moyen , Méthode en double aveugle , Survie sans progression , Sujet âgé de 80 ans ou plus
9.
Proc Natl Acad Sci U S A ; 121(33): e2402903121, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39102549

RÉSUMÉ

Immune checkpoint therapy has limited efficacy for patients with bone-metastatic castration-resistant prostate cancer (bmCRPC). To improve immunotherapy for bmCRPC, we aimed to identify the mechanism of bmCRPC-induced changes in the immune microenvironment. Among bmCRPC patients, higher levels of a 32-gene M2-like macrophage signature in bone metastasis samples correlated with shorter overall survival. Immunohistochemistry showed that CD206-positive (CD206+) macrophages were enriched in bmCRPC bone biopsy specimens compared with primary tumors or lymph node metastases. In preclinical osteogenic prostate cancer (Pca) xenograft models, CD206+ macrophages were recruited to areas with tumor-induced bone. RNA sequencing (RNAseq) analysis showed higher expression of an M2-like gene signature, with activated canonical and noncanonical Wnt pathways, in tumor-associated macrophages isolated from osteogenic tumors (bone-TAMs) than in TAMs isolated from nonosteogenic tumors (ctrl-TAMs). Mechanistic studies showed that endothelial cells (ECs) that had undergone EC-to-osteoblast (EC-to-OSB) transition, the precursors of tumor-induced OSBs, produced paracrine factors, including Wnts, CXCL14, and lysyl oxidase, which induced M2 polarization and recruited M2-like TAMs to the bone-tumor microenvironment (bone-TME). Bone-TAMs suppressed CD8+ T cells' proliferation and cytolytic activity, and these effects were partially reversed by treating bone-TAMs with Wnt inhibitors. Genetic or pharmacological inhibition of Pca-induced EC-to-OSB transition reduced the levels of M2-like macrophages in osteogenic tumors. Our study demonstrates that Pca-induced EC-to-OSB transition drives immunosuppression in the bone-TME, suggesting that therapies that reduce Pca-induced bone formation may improve immunotherapeutic outcomes for bmCRPC.


Sujet(s)
Tumeurs osseuses , Cellules endothéliales , Macrophages , Ostéoblastes , Microenvironnement tumoral , Voie de signalisation Wnt , Mâle , Microenvironnement tumoral/immunologie , Humains , Tumeurs osseuses/immunologie , Tumeurs osseuses/secondaire , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/métabolisme , Animaux , Souris , Macrophages/métabolisme , Macrophages/immunologie , Cellules endothéliales/métabolisme , Cellules endothéliales/immunologie , Ostéoblastes/métabolisme , Ostéoblastes/immunologie , Tumeurs prostatiques résistantes à la castration/immunologie , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/métabolisme , Lignée cellulaire tumorale , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/immunologie , Tumeurs de la prostate/métabolisme , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/immunologie
10.
Cancer Control ; 31: 10732748241274190, 2024.
Article de Anglais | MEDLINE | ID: mdl-39150340

RÉSUMÉ

The treatment of metastatic castration-sensitive prostate cancer (mCSPC) has seen remarkable breakthroughs over the last few years. Diagnostic and therapeutic advances have given rise to debates about risk stratification and optimal first-line treatment selection, as well as to concerns about potential overtreatment in a disease state with a highly heterogeneous clinical behavior. Here, we use case reports from our practice to review the clinical trials exploring intensified triplet regimens combining androgen deprivation therapy with second-generation androgen receptor signaling inhibitors and docetaxel, and we offer our recommendations on how to best select candidates for these novel combinations. Furthermore, the growing adoption of PET imaging with increasingly sensitive and prostate tissue-specific tracers replacing conventional staging technologies has led to the identification of a subset of low-volume mCSPC with nodal metastases which would otherwise not be considered abnormal by RECIST criteria. We describe our PSA-adapted approach to treatment in this unique population with non-measurable low-volume mCSPC which has not been specifically investigated in any phase III clinical trials. We also discuss ongoing clinical trials evaluating treatment de-escalation strategies. Finally, we review how local treatment modalities directed at the prostate or distant sites of disease in oligometastatic CSPC may benefit patients, and how we incorporate metastasis-directed therapy in the management of mCSPC.


Sujet(s)
Métastase tumorale , Humains , Mâle , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Antagonistes des androgènes/usage thérapeutique , Docetaxel/usage thérapeutique , Docetaxel/administration et posologie
11.
JAMA Netw Open ; 7(8): e2428444, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39150709

RÉSUMÉ

Importance: Abiraterone acetate and enzalutamide are recommended as preferred treatments for metastatic castration-resistant prostate cancer (mCRPC), but differences in their relative efficacy are unclear due to a lack of head-to-head clinical trials. Clear guidance is needed for making informed mCRPC therapeutic choices. Objective: To compare clinical outcomes in patients with mCRPC treated with abiraterone acetate or enzalutamide. Design, Setting, and Participants: This retrospective, multicenter cohort study included patients with mCRPC in the US Department of Veterans Affairs health care system who initiated treatment with abiraterone acetate or enzalutamide between January 1, 2014, and October 30, 2022. Exposures: Abiraterone acetate or enzalutamide. Main Outcomes and Measures: The study used inverse probability of treatment weighting to balance baseline characteristics between patients initiating abiraterone acetate or enzalutamide and evaluated restricted mean survival time (RMST) differences in overall survival (OS), prostate cancer-specific survival (PCS), time to next treatment switching or death (TTS), and time to prostate-specific antigen (PSA) response (TTR) at different time points after treatment initiation. Results: The study included 5779 patients (median age, 74.42 years [IQR, 68.94-82.14 years]). Median follow-up was between 38 and 60 months. Patients initiating enzalutamide on average had longer OS than those initiating abiraterone acetate, with RMSTs of 24.29 months (95% CI, 23.58-24.99 months) and 23.38 months (95% CI, 22.85-23.92 months), respectively, and a difference in RMST of 0.90 months (95% CI, 0.02-1.79 months) at 4 years. Similarly, TTS and TTR were improved in patients initiating enzalutamide, with an RMST at 4 years of 1.95 months (95% CI, 0.92-2.99 months) longer for TTS and 3.57 months (95% CI, 1.76-5.38 months) shorter for TTR. For PCS, the RMST at 2 years was 0.48 months (95% CI, 0.01-0.95 months) longer. An examination of subgroups identified that enzalutamide initiation was associated with longer RMST in OS among patients without prior docetaxel treatment (1.14 months; 95% CI, 0.19-2.10 months) and in those with PSA doubling time of 3 months or longer (2.23 months; 95% CI, 0.81-3.66 months) but not among patients with prior docetaxel (-0.25 months; 95% CI, -2.59 to 2.09 months) or with PSA doubling time of less than 3 months (0.05 months; 95% CI, -1.05 to 1.15 months). Conclusions and Relevance: In this cohort study of patients with mCRPC, initiation of enzalutamide was associated with small but statistically significant improvements in OS, PCS, TTS, and TTR compared with initiation of abiraterone acetate. The improvements were more prominent in short-term outcomes, including TTS and TTR, and in patient subgroups without prior docetaxel or with PSA doubling time longer than 3 months.


Sujet(s)
Benzamides , Nitriles , 3-Phényl-2-thiohydantoïne , Tumeurs prostatiques résistantes à la castration , Mâle , Humains , 3-Phényl-2-thiohydantoïne/usage thérapeutique , Nitriles/usage thérapeutique , Benzamides/usage thérapeutique , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/mortalité , Sujet âgé , Études rétrospectives , Androstènes/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Sujet âgé de 80 ans ou plus , Adulte d'âge moyen , États-Unis , Acétate d'abiratérone/usage thérapeutique , Résultat thérapeutique , Métastase tumorale
12.
Sci Rep ; 14(1): 18824, 2024 08 13.
Article de Anglais | MEDLINE | ID: mdl-39138333

RÉSUMÉ

To address the challenge of finding new combination therapies against castration-sensitive prostate cancer, we introduce Vini, a computational tool that predicts the efficacy of drug combinations at the intracellular level by integrating data from the KEGG, DrugBank, Pubchem, Protein Data Bank, Uniprot, NCI-60 and COSMIC databases. Vini is a computational tool that predicts the efficacy of drugs and their combinations at the intracellular level. It addresses the problem comprehensively by considering all known target genes, proteins and small molecules and their mutual interactions involved in the onset and development of cancer. The results obtained point to new, previously unexplored combination therapies that could theoretically be promising candidates for the treatment of castration-sensitive prostate cancer and could prevent the inevitable progression of the cancer to the incurable castration-resistant stage. Furthermore, after analyzing the obtained triple combinations of drugs and their targets, the most common targets became clear: ALK, BCL-2, mTOR, DNA and androgen axis. These results may help to define future therapies against castration-sensitive prostate cancer. The use of the Vini computer model to explore therapeutic combinations represents an innovative approach in the search for effective treatments for castration-sensitive prostate cancer, which, if clinically validated, could potentially lead to new breakthrough therapies.


Sujet(s)
Tumeurs prostatiques résistantes à la castration , Mâle , Humains , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Étude de validation de principe , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Biologie informatique/méthodes
13.
Cancer Med ; 13(12): e7334, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-39143030

RÉSUMÉ

INTRODUCTION: Metastatic castration-resistant prostate cancer (mCRPC) is a heterogeneous disease with prognoses varying from months to years at time of castration-resistant diagnosis. Optimal first-line therapy for those with different prognoses is unknown. METHODS: We conducted a retrospective cohort study of men in a national healthcare delivery system receiving first-line therapy for mCRPC (abiraterone, enzalutamide, docetaxel, or ketoconazole) from 2010 to 2017, with follow-up through 2019. Using commonly drawn prognostic labs at start of mCRPC therapy (hemoglobin, albumin, and alkaline phosphatase), we categorized men into favorable, intermediate, or poor prognostic groups depending on whether they had none, one to two, or all three laboratory values worse than designated laboratory cutoffs. We used Kaplan-Meier methods to examine prostate specific antigen (PSA) progression-free and overall survival (OS) according to prognostic group and first-line therapy, and multivariable cox regression to determine variables associated with survival outcomes. RESULTS: Among 4135 patients, median PSA progression-free survival (PFS) was 6.9 months (95% confidence interval [CI] 6.6-7.3), and median OS 18.8 months (95% CI 18.0-19.6), ranging from 5.7 months (95% CI 4.8-7.0) in the poor prognosis group to 31.3 months (95% CI 29.7-32.9) in the favorable group. OS was similar regardless of initial treatment received for favorable and intermediate groups, but worse for those in the poor prognostic group who received ketoconazole (adjusted hazard ratio 2.07, 95% CI 1.2-3.6). PSA PFS was worse for those who received ketoconazole compared to abiraterone across all prognostic groups (favorable HR 1.76, 95% CI 1.34-2.31; intermediate HR 1.78, 95% CI 1.41-2.25; poor HR 8.01, 95% CI 2.93-21.9). CONCLUSION: Commonly drawn labs at mCRPC treatment start may aid in predicting survival and response to therapies, potentially informing discussions with care teams. First-line treatment selection impacts disease progression for all men with mCRPC regardless of prognostic group, but impacted OS only for men with poor prognosis at treatment start.


Sujet(s)
Androstènes , Docetaxel , Kétoconazole , 3-Phényl-2-thiohydantoïne , Tumeurs prostatiques résistantes à la castration , Mâle , Humains , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/mortalité , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/sang , Sujet âgé , Études rétrospectives , Kétoconazole/usage thérapeutique , Pronostic , Adulte d'âge moyen , 3-Phényl-2-thiohydantoïne/usage thérapeutique , 3-Phényl-2-thiohydantoïne/analogues et dérivés , Docetaxel/usage thérapeutique , Docetaxel/administration et posologie , Androstènes/usage thérapeutique , Antigène spécifique de la prostate/sang , Benzamides/usage thérapeutique , Nitriles/usage thérapeutique , Sujet âgé de 80 ans ou plus , Survie sans progression , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Estimation de Kaplan-Meier
14.
In Vivo ; 38(5): 2328-2334, 2024.
Article de Anglais | MEDLINE | ID: mdl-39187338

RÉSUMÉ

BACKGROUND/AIM: In patients with metastatic castration-sensitive prostate cancer (mCSPC), upfront treatment intensification with the addition of new hormonal agents and/or docetaxel to androgen deprivation therapy (ADT) is recommended. However, this modality is potentially excessive in a subset of these patients. This study aimed to identify patients who may be eligible to omit upfront treatment intensification. PATIENTS AND METHODS: Patients with mCSPC who underwent ADT were enrolled. The association between undetectable prostate-specific antigen (PSA) (<0.2 ng/ml) after ADT initiation and overall or castration-resistance-free survival was evaluated. RESULTS: Ninety-seven out of the 242 enrolled patients had low-risk and/or low-volume cancer and were further analyzed. Of these, 45 (46.4%) patients achieved undetectable PSA. The median follow-up period after ADT initiation was 70 months. The median overall survival among patients with undetectable PSA was quite long, reaching 226 months and significantly longer than that among patients with detectable PSA [71 months, hazard ratio (HR)=0.27, 95% confidence interval (CI)=0.15-0.49, p<0.001]. Time to development of castration-resistance was also long and significantly longer in the undetectable PSA group than that in the detectable PSA group (median: 124 vs. 17 months, HR=0.20, 95% CI=0.12-0.34, p<0.001). CONCLUSION: Patients with low-risk and/or low-volume mCSPC showed long-term survival when undetectable PSA was achieved during conventional ADT. In these patients, skipping upfront treatment intensification does not seem to negatively impact survival.


Sujet(s)
Antagonistes des androgènes , Antigène spécifique de la prostate , Tumeurs prostatiques résistantes à la castration , Humains , Mâle , Sujet âgé , Antigène spécifique de la prostate/sang , Adulte d'âge moyen , Antagonistes des androgènes/usage thérapeutique , Antagonistes des androgènes/administration et posologie , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/mortalité , Résultat thérapeutique , Métastase tumorale , Sujet âgé de 80 ans ou plus , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/mortalité , Docetaxel/administration et posologie , Docetaxel/usage thérapeutique
15.
Int J Mol Sci ; 25(16)2024 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-39201655

RÉSUMÉ

Biochemical recurrence is a process that progresses to castration-resistant prostate cancer (CRPC) and prediction of biochemical recurrence is useful in determining early therapeutic intervention and disease treatment. Prostate cancer is surrounded by adipose tissue, which secretes adipokines, affecting cancer progression. This study aimed to investigate the correlation between blood adipokines and CRPC biochemical recurrence. We retrospectively analyzed the clinical data, including preoperative serum adipokine levels, of 99 patients with pT3a pN0 prostate cancer who underwent proctectomy between 2011 and 2019. The primary outcome was biochemical recurrence (prostate-specific antigen: PSA > 0.2). We identified 65 non-recurrences and 34 biochemical recurrences (one progressed to CRPC). The initial PSA level was significantly higher (p = 0.006), but serum adiponectin (p = 0.328) and leptin (p = 0.647) levels and their ratio (p = 0.323) were not significantly different in the biochemical recurrence group compared with the non-recurrence group. In contrast, significantly more biochemical recurrences were observed in the group with adiponectin < 6 µg/mL and Leptin < 4 ng/mL (p = 0.046), initial PSA > 15 ng/mL, clinical Gleason pattern ≥ 4, and positive resection margin. A significant difference was also observed in the multivariate analysis (hazard ratio: 4.04, 95% confidence interval: 1.21-13.5, p = 0.0232). Thus, low preoperative serum adiponectin and high leptin levels were significantly associated with biochemical recurrence in adipose tissue-invasive prostate cancer, suggesting that they may be useful predictors of biochemical recurrence. Further studies with larger cases are needed to increase the validity of this study.


Sujet(s)
Adiponectine , Tissu adipeux , Leptine , Récidive tumorale locale , Antigène spécifique de la prostate , Tumeurs de la prostate , Humains , Mâle , Adiponectine/sang , Leptine/sang , Sujet âgé , Récidive tumorale locale/sang , Récidive tumorale locale/anatomopathologie , Adulte d'âge moyen , Tissu adipeux/métabolisme , Tissu adipeux/anatomopathologie , Antigène spécifique de la prostate/sang , Études rétrospectives , Tumeurs de la prostate/sang , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/chirurgie , Marqueurs biologiques tumoraux/sang , Pronostic , Tumeurs prostatiques résistantes à la castration/sang , Tumeurs prostatiques résistantes à la castration/anatomopathologie
16.
Gene ; 930: 148818, 2024 Dec 20.
Article de Anglais | MEDLINE | ID: mdl-39098513

RÉSUMÉ

Prostate cancer (PCa) incidence and cancer-related deaths are both high in the male population. Once castration-resistant prostate cancer (CRPC) has developed, PCa can be difficult to manage. Circular RNAs (circRNAs) play essential roles in the regulation of carcinogenesis and cancer progression. In CRPC, however, the potential molecular mechanisms and biological functions of circRNAs are yet to be defined. In this study, we conducted RNA sequencing on four hormone-sensitive prostate cancer (HSPC) tumor tissue samples and three CRPC samples. We recognized hsa_circ_0001610, a novel circRNA that was highly expressed in the cells and tissue of CRPC. We used quantitative real-time PCR (qRT-PCR) to evaluate hsa_circ_0001610 expression. We conducted in vivo and in vitro experiments and found that hsa_circ_0001610 overexpression caused PCa cells to proliferate and migrate and caused enzalutamide resistance. In contrast, the opposite results were found for hsa_circ_0001610 knockdown. We used Western blot, dual-luciferase reporter assays, RNA immunoprecipitation (RIP), qRT-PCR, and rescue experiments to reveal the underlying mechanisms of hsa_circ_0001610. Mechanistically, hsa_circ_0001610 acted as a molecular sponge for miR-1324 and thus reversed its inhibitory effect on its target gene PTK6. As a result, the PTK6 expression was enhanced, which accelerated PCa progression. The findings of this study confirmed that hsa_circ_0001610 drives the progression of PCa through the hsa_circ_0001610/miR-1324/PTK6 axis. Thus, hsa_circ_0001610 is potentially an effective therapeutic target and specific biomarker for advanced PCa.


Sujet(s)
Évolution de la maladie , Régulation de l'expression des gènes tumoraux , microARN , ARN circulaire , Régulation positive , Humains , Mâle , ARN circulaire/génétique , ARN circulaire/métabolisme , microARN/génétique , microARN/métabolisme , Lignée cellulaire tumorale , Régulation positive/génétique , Protein-tyrosine kinases/génétique , Protein-tyrosine kinases/métabolisme , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Prolifération cellulaire/génétique , Animaux , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Souris , Mouvement cellulaire/génétique , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Résistance aux médicaments antinéoplasiques/génétique
17.
Cell Death Dis ; 15(8): 558, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090086

RÉSUMÉ

Prostate cancer (PCa) is the second leading cause of cancer-related death in American men. PCa that relapses after hormonal therapies, referred to as castration resistant PCa (CRPC), often presents with metastases (mCRPC) that are the major cause of mortality. The few available therapies for mCRPC patients include taxanes docetaxel (DTX) and cabazitaxel (CBZ). However, development of resistance limits their clinical use. Mechanistically, resistance arises through upregulation of multidrug resistance (MDR) proteins such as MDR1/ABCB1, making ABCB1 an attractive therapeutic target. Yet, ABCB1 inhibitors failed to be clinically useful due to low specificity and toxicity issues. To study taxanes resistance, we produced CBZ resistant C4-2B cells (RC4-2B) and documented resistance to both CBZ and DTX in cell culture and in 3D prostaspheres settings. RNAseq identified increased expression of ABCB1 in RC4-2B, that was confirmed by immunoblotting and immunofluorescent analysis. ABCB1-specific inhibitor elacridar reversed CBZ and DTX resistance in RC4-2B cells, confirming ABCB1-mediated resistance mechanism. In a cell-based screen using a curated library of cytotoxic drugs, we found that DNA damaging compounds Camptothecin (CPT) and Cytarabine (Ara-C) overcame resistance as seen by similar cytotoxicity in parental C4-2B and resistant RC4-2B. Further, these compounds were cytotoxic to multiple PC cells resistant to taxanes with high ABCB1 expression and, therefore, can be used to conquer the acquired resistance to taxanes in PCa. Finally, inhibition of cyclin-dependent kinases 4/6 (CDK4/6) with small molecule inhibitors (CDK4/6i) potentiated cytotoxic effect of CPT or Ara-C in both parental and resistant cells. Overall, our findings indicate that DNA damaging agents CPT and Ara-C alone or in combination with CDK4/6i can be suggested as a new treatment regimen in CRPC patients, including those that are resistant to taxanes.


Sujet(s)
Sous-famille B de transporteurs à cassette liant l'ATP , Docetaxel , Multirésistance aux médicaments , Résistance aux médicaments antinéoplasiques , Tumeurs prostatiques résistantes à la castration , Taxoïdes , Humains , Mâle , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Sous-famille B de transporteurs à cassette liant l'ATP/métabolisme , Sous-famille B de transporteurs à cassette liant l'ATP/génétique , Lignée cellulaire tumorale , Docetaxel/pharmacologie , Multirésistance aux médicaments/effets des médicaments et des substances chimiques , Taxoïdes/pharmacologie , Taxoïdes/usage thérapeutique , Antinéoplasiques/pharmacologie
18.
Radiology ; 312(2): e233460, 2024 08.
Article de Anglais | MEDLINE | ID: mdl-39162634

RÉSUMÉ

Background Lutetium 177 [177Lu]Lu-PSMA-617 (177Lu-PSMA-617) is a prostate-specific membrane antigen (PSMA)-targeted radioligand therapy for metastatic castration-resistant prostate cancer (mCRPC). Quantitative PSMA PET/CT analysis could provide information on 177Lu-PSMA-617 treatment benefits. Purpose To explore the association between quantitative baseline gallium 68 [68Ga]Ga-PSMA-11 (68Ga-PSMA-11) PET/CT parameters and treatment response and outcomes in the VISION trial. Materials and Methods This was an exploratory secondary analysis of the VISION trial. Eligible participants were randomized (June 2018 to October 2019) in a 2:1 ratio to 177Lu-PSMA-617 therapy (7.4 GBq every 6 weeks for up to six cycles) plus standard of care (SOC) or to SOC only. Baseline 68Ga-PSMA-11 PET parameters, including the mean and maximum standardized uptake value (SUVmean and SUVmax), PSMA-positive tumor volume, and tumor load, were extracted from five anatomic regions and the whole body. Associations of quantitative PET parameters with radiographic progression-free survival (rPFS), overall survival (OS), objective response rate, and prostate-specific antigen response were investigated using univariable and multivariable analyses (with treatment as the only other covariate). Outcomes were assessed in subgroups based on SUVmean quartiles. Results Quantitative PET parameters were well balanced between study arms for the 826 participants included. The median whole-body tumor SUVmean was 7.6 (IQR, 5.8-9.9). Whole-body tumor SUVmean was the best predictor of 177Lu-PSMA-617 efficacy, with a hazard ratio (HR) range of 0.86-1.43 for all outcomes (all P < .001). A 1-unit whole-body tumor SUVmean increase was associated with a 12% and 10% decrease in risk of an rPFS event and death, respectively. 177Lu-PSMA-617 plus SOC prolonged rPFS and OS in all SUVmean quartiles versus SOC only, with no identifiable optimum among participants receiving 177Lu-PSMA-617. Higher baseline PSMA-positive tumor volume and tumor load were associated with worse rPFS (HR range, 1.44-1.53 [P < .05] and 1.02-1.03 [P < .001], respectively) and OS (HR range, 1.36-2.12 [P < .006] and 1.04 [P < .001], respectively). Conclusion Baseline 68Ga-PSMA-11 PET/CT whole-body tumor SUVmean was the best predictor of 177Lu-PSMA-617 efficacy in participants in the VISION trial. Improvements in rPFS and OS with 177Lu-PSMA-617 plus SOC were greater among participants with higher whole-body tumor SUVmean, with evidence for benefit at all SUVmean levels. ClinicalTrials.gov identifier: NCT03511664 Published under a CC BY 4.0 license. Supplemental material is available for this article.


Sujet(s)
Dipeptides , Isotopes du gallium , Radio-isotopes du gallium , Composés hétéromonocycliques , Lutétium , Tomographie par émission de positons couplée à la tomodensitométrie , Tumeurs prostatiques résistantes à la castration , Radiopharmaceutiques , Humains , Mâle , Tumeurs prostatiques résistantes à la castration/imagerie diagnostique , Tumeurs prostatiques résistantes à la castration/radiothérapie , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tomographie par émission de positons couplée à la tomodensitométrie/méthodes , Lutétium/usage thérapeutique , Composés hétéromonocycliques/usage thérapeutique , Sujet âgé , Dipeptides/usage thérapeutique , Radiopharmaceutiques/usage thérapeutique , Adulte d'âge moyen , Résultat thérapeutique , Radio-isotopes/usage thérapeutique , Acide édétique/analogues et dérivés , Acide édétique/usage thérapeutique , Antigène spécifique de la prostate
19.
Med Oncol ; 41(9): 226, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39136842

RÉSUMÉ

Prostate cancer is one of the most common cancers and leading cause of death due to cancer across the globe. This persuaded researchers to devise innovative treatment modalities that may prove effective, safe, and demonstrate better outcomes in terms of patient morbidity and survival. The advancement in theranostics such as lutetium-177 (177Lu)-PSMA-617 radioligand therapies can target prostate cancer cells causing negligible or no damage to most of the normal tissues in patients. It has been proven to effectively improve the quality of life and progression-free survival. In this study, stage IV metastatic castration-resistant prostate cancer patients were treated with 177Lu-PSMA-617, and the therapeutic response and safety of 177Lu-PSMA-617 radioligand therapy were evaluated six months after the treatment. Additionally, molecular docking studies were also conducted to find the possible mechanism at the molecular level that causes the effectiveness of 177Lu-PSMA-617 in prostate cancer.


Sujet(s)
Dipeptides , Composés hétéromonocycliques , Lutétium , Tumeurs prostatiques résistantes à la castration , Radio-isotopes , Radiopharmaceutiques , Humains , Mâle , Tumeurs prostatiques résistantes à la castration/radiothérapie , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Lutétium/usage thérapeutique , Composés hétéromonocycliques/usage thérapeutique , Dipeptides/usage thérapeutique , Radio-isotopes/usage thérapeutique , Radiopharmaceutiques/usage thérapeutique , Sujet âgé , Simulation de docking moléculaire , Antigène spécifique de la prostate , Adulte d'âge moyen , Résultat thérapeutique
20.
J Pineal Res ; 76(5): e13003, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39143673

RÉSUMÉ

RNA N6-methyladenosine (m6A) readers mediate cancer progression. However, the functional role and potential mechanisms of the m6A readers in prostate cancer tumorigenicity remain to be elucidated. In this study, we demonstrate that YTHDF3 expression is elevated in castration-resistant prostate cancer (CRPC) and positively correlated to high grade, bone metastasis and poor survival. YTHDF3 expression promoted CRPC cell proliferation, epithelial to mesenchymal transition (EMT) and tumour progression. Mechanistically, YTHDF3 promoted the RNA degradation of SPOP and NXK3.1 but stabilized RNA expressions of TWIST1 and SNAI2 dependent on m6A to facilitate cell proliferation and EMT. Additionally, YTHDF3 expression enhanced AKT activity via degrading SPOP in an m6A-dependent manner. Importantly, we found that melatonin can compete with m6A to occupy the m6A-binding cage of YTHDF3, leading to inhibition of YTHFD3 and its target expressions as well as CRPC tumour growth. Our findings uncover an essential role of YTHDF3 in the progression of CRPC and highlight the role of melatonin in anti-CRPC activity.


Sujet(s)
Évolution de la maladie , Tumeurs prostatiques résistantes à la castration , Protéines de liaison à l'ARN , Mâle , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Humains , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Animaux , Lignée cellulaire tumorale , Adénosine/analogues et dérivés , Adénosine/métabolisme , Prolifération cellulaire/génétique , Souris , Transition épithélio-mésenchymateuse/génétique , Régulation de l'expression des gènes tumoraux , Mélatonine/métabolisme , Souris nude
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE