Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.411
Filtrer
1.
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
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.
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
4.
Theranostics ; 14(9): 3623-3633, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948055

RÉSUMÉ

Introduction: Prostate Specific Membrane Antigen Positron Emission Tomography (PSMA-PET) is routinely used for the staging of patients with prostate cancer, but data on response assessment are sparse and primarily stem from metastatic castration-resistant prostate cancer (mCRPC) patients treated with PSMA radioligand therapy. Still, follow-up PSMA-PET is employed in earlier disease stages in case of clinical suspicion of disease persistence, recurrence or progression to decide if localized or systemic treatment is indicated. Therefore, the prognostic value of PSMA-PET derived tumor volumes in earlier disease stages (i.e., hormone-sensitive prostate cancer (HSPC) and non-[177Lu]Lu-PSMA-617 (LuPSMA) therapy castration resistant prostate cancer (CRPC)) are evaluated in this manuscript. Methods: A total number of 73 patients (6 primary staging, 42 HSPC, 25 CRPC) underwent two (i.e., baseline and follow-up, median interval: 379 days) whole-body [68Ga]Ga-PSMA-11 PET/CT scans between Nov 2014 and Dec 2018. Analysis was restricted to non-LuPSMA therapy patients. PSMA-PETs were retrospectively analyzed and primary tumor, lymph node-, visceral-, and bone metastases were segmented. Body weight-adjusted organ-specific and total tumor volumes (PSMAvol: sum of PET volumes of all lesions) were measured for baseline and follow-up. PSMAvol response was calculated as the absolute difference of whole-body tumor volumes. High metastatic burden (>5 metastases), RECIP 1.0 and PSMA-PET Progression Criteria (PPP) were determined. Survival data were sourced from the cancer registry. Results: The average number of tumor lesions per patient on the initial PET examination was 10.3 (SD 28.4). At baseline, PSMAvol was strongly associated with OS (HR 3.92, p <0.001; n = 73). Likewise, response in PSMAvol was significantly associated with OS (HR 10.48, p < 0.005; n = 73). PPP achieved significance as well (HR 2.19, p <0.05, n = 73). Patients with hormone sensitive disease and poor PSMAvol response (upper quartile of PSMAvol change) in follow-up had shorter outcome (p < 0.05; n = 42). PSMAvol in bones was the most relevant parameter for OS prognostication at baseline and for response assessment (HR 31.11 p < 0.001; HR 32.27, p < 0.001; n = 73). Conclusion: PPP and response in PSMAvol were significantly associated with OS in the present heterogeneous cohort. Bone tumor volume was the relevant miTNM region for OS prognostication. Future prospective evaluation of the performance of organ specific PSMAvol in more homogeneous cohorts seems warranted.


Sujet(s)
Tomographie par émission de positons couplée à la tomodensitométrie , Tumeurs prostatiques résistantes à la castration , Humains , Mâle , Sujet âgé , Tomographie par émission de positons couplée à la tomodensitométrie/méthodes , Tumeurs prostatiques résistantes à la castration/imagerie diagnostique , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Adulte d'âge moyen , Études de suivi , Radio-isotopes du gallium , Études rétrospectives , Sujet âgé de 80 ans ou plus , Tumeurs de la prostate/imagerie diagnostique , Tumeurs de la prostate/anatomopathologie , Glutamate carboxypeptidase II/métabolisme , Radiopharmaceutiques , Antigènes de surface/métabolisme , Isotopes du gallium , Pronostic , Lutétium/usage thérapeutique , Tomographie par émission de positons/méthodes , Charge tumorale , Composés hétéromonocycliques/usage thérapeutique , Dipeptides/usage thérapeutique
6.
Nagoya J Med Sci ; 86(2): 169-180, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38962407

RÉSUMÉ

Androgen receptor signaling inhibitors combined with androgen deprivation therapy have become the standard of care for metastatic castration-sensitive prostate cancer (mCSPC), regardless of tumor volume or risk. However, survival of approximately one-third of these patients has not improved, necessitating further treatment escalation. On the other hand, for patients with oligometastatic mCSPC, there is an emerging role for local radiation therapy. Although data remain scarce, it is expected that treatment of both primary tumor as well as metastasis-directed therapy may improve survival outcomes. In these patients, systemic therapy may be de-escalated to intermittent therapy. However, precise risk stratification is necessary for risk-based treatment escalation or de-escalation. In addition to risk stratification based on clinical parameters, research has been conducted to incorporate genomic and/or transcriptomic data into risk stratification. In future, an integrated risk model is expected to precisely stratify patients and guide treatment strategies. Here, we first review the transition of the standard treatment for mCSPC over the last decade and further discuss the newest concept of escalating or de-escalating treatment using a multi-modal approach based on the currently available literature.


Sujet(s)
Métastase tumorale , Humains , Mâle , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/thérapie , Antagonistes des androgènes/usage thérapeutique , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/thérapie , Antagonistes du récepteur des androgènes/usage thérapeutique
7.
Sci Rep ; 14(1): 15307, 2024 07 03.
Article de Anglais | MEDLINE | ID: mdl-38961131

RÉSUMÉ

A multicenter study of nonmetastatic castration-resistant prostate cancer (nmCRPC) was conducted to identify the optimal cut-off value of prostate-specific antigen (PSA) doubling time (PSADT) that correlated with the prognosis in Japanese nmCRPC. Of the 515 patients diagnosed and treated for nmCRPC at 25 participating Japanese Urological Oncology Group centers, 450 patients with complete clinical information were included. The prognostic values of clinical factors were evaluated with respect to prostate specific antigen progression-free (PFS), cancer-specific survival (CSS), and overall survival (OS). The optimal cutoff value of PSADT was identified using survival tree analysis by Python. The Median PSA and PSADT at diagnosis of nmCRPC were 3.3 ng/ml, and 5.2 months, respectively. Patients treated with novel hormonal therapy (NHT) showed significantly longer PFS (HR: hazard ratio 0.38, p < 0.0001) and PFS2 (HR 0.45, p < 0.0001) than those treated with vintage nonsteroidal antiandrogen agent (Vintage). The survival tree identified 4.65 months as the most prognostic PSADT cutoff point. Among the clinical and pathological factors PSADT of < 4.65 months remained an independent prognostic factor for OS (HR 2.96, p = 0.0003) and CSS (HR 3.66, p < 0.0001). Current data represented optimal cut-off of PSADT 4.65 months for a Japanese nmCRPC.


Sujet(s)
Antigène spécifique de la prostate , Tumeurs prostatiques résistantes à la castration , Humains , Mâle , Antigène spécifique de la prostate/sang , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/sang , Tumeurs prostatiques résistantes à la castration/mortalité , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Sujet âgé , Adulte d'âge moyen , Japon/épidémiologie , Pronostic , Sujet âgé de 80 ans ou plus , Peuples d'Asie de l'Est
8.
Int J Mol Sci ; 25(14)2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39063105

RÉSUMÉ

Androgen deprivation therapy (ADT) is the primary treatment for advanced prostate cancer (PCa). However, prolonged ADT inevitably results in therapy resistance with the emergence of the castration-resistant PCa phenotype (CRPC). Hence, there is an urgent need to explore new treatment options capable of delaying PCa progression. Hispidin (HPD) is a natural polyketide primarily derived from plants and fungi. HPD has been shown to have a diverse pharmacological profile, exhibiting anti-inflammatory, antiviral, cardiovascular and neuro-protective activities. However, there is currently no research regarding its properties in the context of PCa treatment. This research article seeks to evaluate the anti-cancer effect of HPD and determine the underlying molecular basis in both androgen-sensitive PCa and CRPC cells. Cell growth, migration, and invasion assays were performed via the MTS method, a wound healing assay and the transwell method. To investigate if HPD affected the expression of proteins, Western blot analysis was conducted. Furthermore, apoptosis was assessed by Annexin V-FITC/PI staining and Western blot analyses. HPD exhibited a favorable pharmaceutical profile to inhibit cell growth; disrupt the cell cycle; attenuate wound healing, migration and invasion; and induce apoptosis in PCa cells in vitro. The mechanistic results demonstrated that HPD reduced AR, MMP-2 and MMP-9 expression and activated the caspase-related pathway, leading to programmed cell death in PCa cells. We showed the anti-cancer effect of HPD on PCa cells and confirmed its feasibility as a novel therapeutic agent. This study provides significant insights into the delineation of the molecular mechanism of HPD in PCa cells and the development of an effective and safe therapy using HPD to eliminate PCa progression.


Sujet(s)
Apoptose , Mouvement cellulaire , Prolifération cellulaire , Tumeurs de la prostate , Humains , Mâle , Apoptose/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Cycle cellulaire/effets des médicaments et des substances chimiques , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Polycétides/pharmacologie , Polycétides/composition chimique
9.
FASEB J ; 38(13): e23663, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38958986

RÉSUMÉ

This study delves into the unexplored realm of castration-resistant prostate cancer (CRPC) by investigating the role of TRIM28 and its intricate molecular mechanisms using high-throughput single-cell transcriptome sequencing and advanced bioinformatics analysis. Our comprehensive examination unveiled dynamic TRIM28 expression changes, particularly in immune cells such as macrophages and CD8+ T cells within CRPC. Correlation analyses with TCGA data highlighted the connection between TRIM28 and immune checkpoint expression and emphasized its pivotal influence on the quantity and functionality of immune cells. Using TRIM28 knockout mouse models, we identified differentially expressed genes and enriched pathways, unraveling the potential regulatory involvement of TRIM28 in the cGAS-STING pathway. In vitro, experiments further illuminated that TRIM28 knockout in prostate cancer cells induced a notable anti-tumor immune effect by inhibiting M2 macrophage polarization and enhancing CD8+ T cell activity. This impactful discovery was validated in an in situ transplant tumor model, where TRIM28 knockout exhibited a deceleration in tumor growth, reduced proportions of M2 macrophages, and enhanced infiltration of CD8+ T cells. In summary, this study elucidates the hitherto unknown anti-tumor immune role of TRIM28 in CRPC and unravels its potential regulatory mechanism via the cGAS-STING signaling pathway. These findings provide novel insights into the immune landscape of CRPC, offering promising directions for developing innovative therapeutic strategies.


Sujet(s)
Lymphocytes T CD8+ , Protéines membranaires , Tumeurs prostatiques résistantes à la castration , Protéine-28 à motif tripartite , Animaux , Humains , Mâle , Souris , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Macrophages/métabolisme , Macrophages/immunologie , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Souris de lignée C57BL , Souris knockout , Nucleotidyltransferases/métabolisme , Nucleotidyltransferases/génétique , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/immunologie , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Transduction du signal , Protéine-28 à motif tripartite/métabolisme , Protéine-28 à motif tripartite/génétique
10.
BMC Cancer ; 24(1): 817, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38978000

RÉSUMÉ

BACKGROUND: Inflammation plays a pivotal role in the progression of prostate cancer (PCa). Several immune-inflammatory indices, including neutrophil to lymphocyte ratio (NLR), derived neutrophil to lymphocyte ratio (dNLR), lymphocyte to monocyte ratio (LMR) and platelet to lymphocyte ratio (PLR), lung immune prognostic index (LIPI), systemic inflammation response index (SIRI) and systemic immune inflammation index (SII), have demonstrated their prognostic values in several solid malignancies. However, Comparisons of superiority with these seven indices' predictive efficacy within metastatic hormone-sensitive PCa (mHSPC) and metastatic castration-resistant PCa (mCRPC) remain uncertain. METHODS: We retrospectively included 407 patients diagnosed with mHSPC and 158 patients with mCRPC at West China Hospital from 2005 to 2022. The seven immune-inflammatory indices were computed based on hematological data of mHSPC at initial diagnosis and mCRPC at progression to CRPC. Prognostic value for castration-resistant prostate cancer-free survival (CFS), overall survival (OS), prostate-specific antigen progression-free survival (PSA-PFS) and prostate-specific antigen (PSA) response was assessed using Kaplan-Meier curves, Cox regression models, and chi-square tests. The predictive performance of each immune-inflammatory index was assessed using the area under the curve (AUC) in time-dependent receiver operating characteristic curve (ROC) analysis and C-index calculation. RESULTS: All seven immune-inflammatory indices were significantly associated with CFS and OS in the mHSPC cohort, as well as with PSA response, PSA-PFS, and OS in the mCRPC cohort. In the mHSPC cohort, LIPI consistently exhibited higher AUC values compared to NLR, dNLR, LMR, PLR, SII, and SIRI for predicting CFS and OS. This indicates that LIPI had a superior discriminative ability compared to the other indices (C-index of LIPI: 0.643 and 0.686 for CFS and OS, respectively). Notably, the predictive advantage of LIPI over other indices in the mHSPC stage diminished in the mCRPC stage. CONCLUSIONS: This study firstly confirmed the prognostic value of SII, SIRI and LIPI in mHSPC and mCRPC, and revealed that LIPI had a higher predictive power than NLR, dNLR, LMR, PLR, SII and SIRI in mHSPC. These non-invasive indices can enable clinicians to quickly assess the prognosis of patients.


Sujet(s)
Granulocytes neutrophiles , Tumeurs prostatiques résistantes à la castration , Humains , Mâle , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/sang , Tumeurs prostatiques résistantes à la castration/immunologie , Tumeurs prostatiques résistantes à la castration/mortalité , Sujet âgé , Études rétrospectives , Adulte d'âge moyen , Pronostic , Granulocytes neutrophiles/immunologie , Inflammation , Lymphocytes/immunologie , Antigène spécifique de la prostate/sang , Courbe ROC , Sujet âgé de 80 ans ou plus , Plaquettes/anatomopathologie , Plaquettes/immunologie
11.
Int J Biol Sci ; 20(9): 3638-3655, 2024.
Article de Anglais | MEDLINE | ID: mdl-38993562

RÉSUMÉ

Castration-resistant prostate cancer (CRPC) is the leading cause of prostate cancer (PCa)-related death in males, which occurs after the failure of androgen deprivation therapy (ADT). PIWI-interacting RNAs (piRNAs) are crucial regulators in many human cancers, but their expression patterns and roles in CRPC remain unknown. In this study, we performed small RNA sequencing to explore CRPC-associated piRNAs using 10 benign prostate tissues, and 9 paired hormone-sensitive PCa (HSPCa) and CRPC tissues from the same patients. PiRNA-4447944 (piR-4447944) was discovered to be highly expressed in CRPC group compared with HSPCa and benign groups. Functional analyses revealed that piR-4447944 overexpression endowed PCa cells with castration resistance ability in vitro and in vivo, whereas knockdown of piR-4447944 using anti-sense RNA suppressed the proliferation, migration and invasion of CRPC cells. Additionally, enforced piR-4447944 expression promoted in vitro migration and invasion of PCa cells, and reduced cell apoptosis. Mechanistically, piR-4447944 bound to PIWIL2 to form a piR-4447944/PIWIL2 complex and inhibited tumor suppressor NEFH through direct interaction at the post-transcriptional level. Collectively, our study indicates that piR-4447944 is essential for prostate tumor-propagating cells and mediates androgen-independent growth of PCa, which extends current understanding of piRNAs in cancer biology and provides a potential approach for CRPC treatment.


Sujet(s)
Protéines Argonaute , Prolifération cellulaire , Tumeurs prostatiques résistantes à la castration , Petit ARN interférent , Mâle , Humains , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Petit ARN interférent/métabolisme , Protéines Argonaute/métabolisme , Protéines Argonaute/génétique , Animaux , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Souris , Apoptose , Mouvement cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Souris nude , ARN interagissant avec Piwi
12.
Sci Rep ; 14(1): 15095, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956125

RÉSUMÉ

Nanogels offer hope for precise drug delivery, while addressing drug delivery hurdles is vital for effective prostate cancer (PCa) management. We developed an injectable elastin nanogels (ENG) for efficient drug delivery system to overcome castration-resistant prostate cancer (CRPC) by delivering Decursin, a small molecule inhibitor that blocks Wnt/ßcatenin pathways for PCa. The ENG exhibited favourable characteristics such as biocompatibility, flexibility, and low toxicity. In this study, size, shape, surface charge, chemical composition, thermal stability, and other properties of ENG were used to confirm the successful synthesis and incorporation of Decursin (DEC) into elastin nanogels (ENG) for prostate cancer therapy. In vitro studies demonstrated sustained release of DEC from the ENG over 120 h, with a pH-dependent release pattern. DU145 cell line induces moderate cytotoxicity of DEC-ENG indicates that nanomedicine has an impact on cell viability and helps strike a balance between therapeutics efficacy and safety while the EPR effect enables targeted drug delivery to prostate tumor sites compared to free DEC. Morphological analysis further supported the effectiveness of DEC-ENG in inducing cell death. Overall, these findings highlight the promising role of ENG-encapsulated decursin as a targeted drug delivery system for CRPC.


Sujet(s)
Élastine , Nanogels , Tumeurs prostatiques résistantes à la castration , Mâle , Élastine/composition chimique , Humains , Lignée cellulaire tumorale , Nanogels/composition chimique , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/métabolisme , Systèmes de délivrance de médicaments , Survie cellulaire/effets des médicaments et des substances chimiques , Libération de médicament , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/administration et posologie , Benzopyranes , Butyrates
13.
EBioMedicine ; 105: 105212, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38954976

RÉSUMÉ

BACKGROUND: The E1A-associated protein p300 (p300) has emerged as a promising target for cancer therapy due to its crucial role in promoting oncogenic signaling pathways in various cancers, including prostate cancer. This need is particularly significant in prostate cancer. While androgen deprivation therapy (ADT) has demonstrated promising efficacy in prostate cancer, its long-term use can eventually lead to the development of castration-resistant prostate cancer (CRPC) and neuroendocrine prostate cancer (NEPC). Notably, p300 has been identified as an important co-activator of the androgen receptor (AR), highlighting its significance in prostate cancer progression. Moreover, recent studies have revealed the involvement of p300 in AR-independent oncogenes associated with NEPC. Therefore, the blockade of p300 may emerge as an effective therapeutic strategy to address the challenges posed by both CRPC and NEPC. METHODS: We employed AI-assisted design to develop a peptide-based PROTAC (proteolysis-targeting chimera) drug that targets p300, effectively degrading p300 in vitro and in vivo utilizing nano-selenium as a peptide drug delivery system. FINDINGS: Our p300-targeting peptide PROTAC drug demonstrated effective p300 degradation and cancer cell-killing capabilities in both CRPC, AR-negative, and NEPC cells. This study demonstrated the efficacy of a p300-targeting drug in NEPC cells. In both AR-positive and AR-negative mouse models, the p300 PROTAC drug showed potent p300 degradation and tumor suppression. INTERPRETATION: The design of peptide PROTAC drug targeting p300 is feasible and represents an efficient therapeutic strategy for CRPC, AR-negative prostate cancer, and NEPC. FUNDING: The funding details can be found in the Acknowledgements section.


Sujet(s)
Protéine p300-E1A , Peptides , Tumeurs de la prostate , Protéolyse , Tests d'activité antitumorale sur modèle de xénogreffe , Mâle , Humains , Protéolyse/effets des médicaments et des substances chimiques , Animaux , Souris , Lignée cellulaire tumorale , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Protéine p300-E1A/métabolisme , Peptides/pharmacologie , Peptides/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Modèles animaux de maladie humaine , Récepteurs aux androgènes/métabolisme , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Découverte de médicament
14.
Proc Natl Acad Sci U S A ; 121(28): e2322203121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38968122

RÉSUMÉ

Targeting cell surface molecules using radioligand and antibody-based therapies has yielded considerable success across cancers. However, it remains unclear how the expression of putative lineage markers, particularly cell surface molecules, varies in the process of lineage plasticity, wherein tumor cells alter their identity and acquire new oncogenic properties. A notable example of lineage plasticity is the transformation of prostate adenocarcinoma (PRAD) to neuroendocrine prostate cancer (NEPC)-a growing resistance mechanism that results in the loss of responsiveness to androgen blockade and portends dismal patient survival. To understand how lineage markers vary across the evolution of lineage plasticity in prostate cancer, we applied single-cell analyses to 21 human prostate tumor biopsies and two genetically engineered mouse models, together with tissue microarray analysis on 131 tumor samples. Not only did we observe a higher degree of phenotypic heterogeneity in castrate-resistant PRAD and NEPC than previously anticipated but also found that the expression of molecules targeted therapeutically, namely PSMA, STEAP1, STEAP2, TROP2, CEACAM5, and DLL3, varied within a subset of gene-regulatory networks (GRNs). We also noted that NEPC and small cell lung cancer subtypes shared a set of GRNs, indicative of conserved biologic pathways that may be exploited therapeutically across tumor types. While this extreme level of transcriptional heterogeneity, particularly in cell surface marker expression, may mitigate the durability of clinical responses to current and future antigen-directed therapies, its delineation may yield signatures for patient selection in clinical trials, potentially across distinct cancer types.


Sujet(s)
Analyse sur cellule unique , Mâle , Humains , Analyse sur cellule unique/méthodes , Animaux , Souris , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/traitement médicamenteux , Antigènes de surface/métabolisme , Antigènes de surface/génétique , Antigènes néoplasiques/métabolisme , Antigènes néoplasiques/génétique , Antigènes néoplasiques/immunologie , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Adénocarcinome/génétique , Adénocarcinome/anatomopathologie , Adénocarcinome/métabolisme , Adénocarcinome/traitement médicamenteux , Carcinome neuroendocrine/génétique , Carcinome neuroendocrine/anatomopathologie , Carcinome neuroendocrine/métabolisme , Carcinome neuroendocrine/traitement médicamenteux , Régulation de l'expression des gènes tumoraux , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux
15.
Clin Pharmacokinet ; 63(7): 1025-1036, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38963459

RÉSUMÉ

BACKGROUND AND OBJECTIVE: Trough abiraterone concentration (ABI Cmin) of 8.4 ng/mL has been identified as an appropriate efficacy threshold in patients treated for metastatic castration-resistant prostate cancer (mCRPC). The aim of the phase II OPTIMABI study was to evaluate the efficacy of pharmacokinetics (PK)-guided dose escalation of abiraterone acetate (AA) in underexposed patients with mCRPC with early tumour progression. METHODS: This multicentre, non-randomised study consisted of two sequential steps. In step 1, all patients started treatment with 1000 mg of AA once daily. Abiraterone Cmin was measured 22-26 h after the last dose intake each month during the first 12 weeks of treatment. In step 2, underexposed patients (Cmin < 8.4 ng/mL) with tumour progression within the first 6 months of treatment were enrolled and received AA 1000 mg twice daily. The primary endpoint was the rate of non-progression at 12 weeks after the dose doubling. During step 1, adherence to ABI treatment was assessed using the Girerd self-reported questionnaire. A post-hoc analysis of pharmacokinetic (PK) data was conducted using Bayesian estimation of Cmin from samples collected outside the sampling guidelines (22-26 h). RESULTS: In the intention-to-treat analysis (ITT), 81 patients were included in step 1. In all, 21 (26%) patients were underexposed in step 1, and 8 of them (38%) experienced tumour progression within the first 6 months. A total of 71 patients (88%) completed the Girerd self-reported questionnaire. Of the patients, 62% had a score of 0, and 38% had a score of 1 or 2 (minimal compliance failure), without a significant difference in mean ABI Cmin in the two groups. Four patients were enrolled in step 2, and all reached the exposure target (Cmin > 8.4 ng/mL) after doubling the dose, but none met the primary endpoint. In the post-hoc analysis of PK data, 32 patients (39%) were underexposed, and ABI Cmin was independently associated with worse progression-free survival [hazard ratio (HR) 2.50, 95% confidence interval (CI) 1.07-5.81; p = 0.03], in contrast to the ITT analysis. CONCLUSION: The ITT and per-protocol analyses showed no statistical association between ABI underexposure and an increased risk of early tumour progression in patients with mCRPC, while the Bayesian estimator showed an association. However, other strategies than dose escalation at the time of progression need to be evaluated. Treatment adherence appeared to be uniformly good in the present study. Finally, the use of a Bayesian approach to recover samples collected outside the predefined blood collection time window could benefit the conduct of clinical trials based on drug monitoring. OPTIMABI trial is registered as National Clinical Trial number NCT03458247, with the EudraCT number 2017-000560-15).


Sujet(s)
Tumeurs prostatiques résistantes à la castration , Humains , Mâle , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/sang , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Sujet âgé , Adulte d'âge moyen , Sujet âgé de 80 ans ou plus , Évolution de la maladie , Relation dose-effet des médicaments , Androstènes/administration et posologie , Androstènes/pharmacocinétique , Androstènes/usage thérapeutique , Acétate d'abiratérone/administration et posologie , Acétate d'abiratérone/pharmacocinétique , Acétate d'abiratérone/usage thérapeutique , Antinéoplasiques/pharmacocinétique , Antinéoplasiques/administration et posologie , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/sang , Métastase tumorale
16.
Adv Cancer Res ; 161: 403-429, 2024.
Article de Anglais | MEDLINE | ID: mdl-39032955

RÉSUMÉ

Worldwide, prostate cancer (PCa) remains a leading cause of death in men. Histologically, the majority of PCa cases are classified as adenocarcinomas, which are mainly composed of androgen receptor-positive luminal cells. PCa is initially driven by the androgen receptor axis, where androgen-mediated activation of the receptor is one of the primary culprits for disease progression. Therefore, in advanced stage PCa, patients are generally treated with androgen deprivation therapies alone or in combination with androgen receptor pathway inhibitors. However, after an initial decrease, the cancer recurs for majority patients. At this stage, cancer is known as castration-resistant prostate cancer (CRPC). Majority of CRPC tumors still depend on androgen receptor axis for its progression to metastasis. However, in around 20-30% of cases, CRPC progresses via an androgen receptor-independent pathway and is often presented as neuroendocrine cancer (NE). This NE phenotype is highly aggressive with poor overall survival as compared to CRPC adenocarcinoma. NE cancers are resistant to standard taxane chemotherapies, which are often used to treat metastatic disease. Pathologically and morphologically, NE cancers are highly diverse and often co-exist with adenocarcinoma. Due to the lack of proper biomarkers, it is often difficult to make an early diagnosis of this lethal disease. Moreover, increased tumor heterogeneity and admixtures of adeno and NE subtypes in the same tumor make early detection of NE tumors very difficult. With the advancement of our knowledge and sequencing technology, we are now able to better understand the molecular mediators of this transformation pathway. This current study will give an update on how various molecular regulators are involved in these lineage transformation processes and what challenges we are still facing to detect and treat this cancer.


Sujet(s)
Tumeurs neuroendocrines , Humains , Mâle , Tumeurs neuroendocrines/anatomopathologie , Tumeurs neuroendocrines/métabolisme , Tumeurs neuroendocrines/génétique , Récepteurs aux androgènes/métabolisme , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Animaux , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/génétique , Régulation de l'expression des gènes tumoraux
17.
J Immunother Cancer ; 12(7)2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39060021

RÉSUMÉ

BACKGROUND: Androgen deprivation therapy (ADT) is a front-line treatment for prostate cancer. In some men, their tumors can become refractory leading to the development of castration-resistant prostate cancer (CRPC). This causes tumors to regrow and metastasize, despite ongoing treatment, and impacts negatively on patient survival. ADT is known to stimulate the accumulation of immunosuppressive cells like protumoral tumor-associated macrophages (TAMs), myeloid-derived suppressor cells and regulatory T cells in prostate tumors, as well as hypofunctional T cells. Protumoral TAMs have been shown to accumulate around tumor blood vessels during chemotherapy and radiotherapy in other forms of cancer, where they drive tumor relapse. Our aim was to see whether such perivascular (PV) TAMs also accumulate in ADT-treated prostate tumors prior to CRPC, and, if so, whether selectively inducing them to express a potent immunostimulant, interferon beta (IFNß), would stimulate antitumor immunity and delay CRPC. METHODS: We used multiplex immunofluorescence to assess the effects of ADT on the distribution and activation status of TAMs, CD8+T cells, CD4+T cells and NK cells in mouse and/or human prostate tumors. We then used antibody-coated, lipid nanoparticles (LNPs) to selectively target a STING agonist, 2'3'-cGAMP (cGAMP), to PV TAMs in mouse prostate tumors during ADT. RESULTS: TAMs accumulated at high density around blood vessels in response to ADT and expressed markers of a protumoral phenotype including folate receptor-beta (FR-ß), MRC1 (CD206), CD169 and VISTA. Additionally, higher numbers of inactive (PD-1-) CD8+T cells and reduced numbers of active (CD69+) NK cells were present in these PV tumor areas. LNPs coated with an antibody to FR-ß selectively delivered cGAMP to PV TAMs in ADT-treated tumors, where they activated STING and upregulated the expression of IFNß. This resulted in a marked increase in the density of active CD8+T cells (along with CD4+T cells and NK cells) in PV tumor areas, and significantly delayed the onset of CRPC. Antibody depletion of CD8+T cells during LNP administration demonstrated the essential role of these cells in delay in CRPC induced by LNPs. CONCLUSION: Together, our data indicate that targeting a STING agonist to PV TAMs could be used to extend the treatment window for ADT in prostate cancer.


Sujet(s)
Macrophages , Protéines membranaires , Mâle , Animaux , Souris , Protéines membranaires/métabolisme , Protéines membranaires/agonistes , Macrophages/métabolisme , Macrophages/immunologie , Macrophages/effets des médicaments et des substances chimiques , Humains , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Antagonistes des androgènes/usage thérapeutique , Antagonistes des androgènes/pharmacologie , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Macrophages associés aux tumeurs/immunologie , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Résistance aux médicaments antinéoplasiques
18.
J Manag Care Spec Pharm ; 30(7): 684-697, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38950154

RÉSUMÉ

BACKGROUND: The advent of next-generation imaging will likely reduce nonmetastatic prostate cancer (PC) prevalence and increase identification of metastatic prostate cancer cases, resulting in two predominant advanced stages in the metastatic setting. There is a need to characterize changes in health care resource utilization (HRU) and costs when metastatic castration-sensitive PC (mCSPC) progresses to metastatic castration-resistant PC (mCRPC) to identify value drivers from current and new treatments. OBJECTIVE: To describe treatment patterns, HRU, and total health care costs among patients with mCSPC, before and after progression to mCRPC. METHODS: Clinical data from the Flatiron Metastatic PC Core Registry (January 1, 2013, to December 1, 2021) and linked claims from Komodo Health (January 1, 2014, to December 1, 2021) were used to identify patients with progression from mCSPC to mCRPC (date of progression was the index date) and subsequently initiated first-line mCRPC therapy on/after January 1, 2017. Treatment patterns and all-cause/PC-related HRU and health care costs were described per-patient-per-month (PPPM), separately for no more than 12 months pre-index (mCSPC disease state) and post-index (mCRPC disease state). Costs (payer's perspective) included those for services/procedures from medical claims and costs from pharmacy claims. Continuous HRU and costs were compared between the mCSPC and mCRPC disease states using Wilcoxon signed rank tests. RESULTS: Among 296 patients with mCSPC progressing to mCRPC (median age 69.0 years, 60.5% White, 15.9% Black), use of systemic therapies with androgen deprivation therapy increased dramatically from 35.1% in the mCSPC disease state to 92.9% in the mCRPC disease state, and use of androgen deprivation therapy monotherapy decreased from 25.7% to 2.4%, respectively. Although 39.2% received none of these therapies in the mCSPC disease state, this proportion decreased to 4.7% after transition to mCRPC. The mean number of days with PC-related outpatient visits increased from 1.57 to 2.16 PPPM in the mCSPC and mCRPC disease states (P < 0.001). From the mCSPC to mCRPC disease states, mean all-cause total health care costs PPPM increased from $4,424 (medical costs: $2,846) to $9,717 (medical costs: $4,654), and mean PC-related total health care costs PPPM increased from $2,859 (medical costs: $1,626) to $8,012 (medical costs: $3,285; all P < 0.001). CONCLUSIONS: In this real-world study of patients with disease progression from mCSPC to mCRPC in US clinical practice, nearly 2-in-3 patients did not receive treatment with additional systemic therapies before progression to castration resistance. Post-progression, mean PC-related total costs increased nearly 3-fold, with a more than 2-fold increase in PC-related medical costs. Use of additional systemic therapies may delay the time and cost associated with disease progression to castration resistance.


Sujet(s)
Coûts indirects de la maladie , Évolution de la maladie , Coûts des soins de santé , Tumeurs prostatiques résistantes à la castration , Humains , Mâle , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/économie , Tumeurs prostatiques résistantes à la castration/anatomopathologie , États-Unis , Sujet âgé , Coûts des soins de santé/statistiques et données numériques , Adulte d'âge moyen , Études rétrospectives , Sujet âgé de 80 ans ou plus , Métastase tumorale , Enregistrements
19.
Hinyokika Kiyo ; 70(6): 173-177, 2024 Jun.
Article de Japonais | MEDLINE | ID: mdl-38967030

RÉSUMÉ

A 74-year-old man visited the urology clinic with the chief complaint of urinary retention in December 2014. Serum level of initial prostate specific antigen (PSA) was 50 ng/ml and he was diagnosed with Gleason Score 4+4 prostate adenocarcinoma with regional lymphadenopathy (cT3aN1M0). PSA level had declined after the treatment with combined androgen blockade. In November 2018, he was diagnosed with castration resistant prostate cancer (CRPC) as local progression was detected by computed tomography (CT) while PSA level did not increase. Since local symptoms worsened, resulting in repeated hematuria after the treatment with enzalutamide, palliative radiation therapy to the prostate (45 Gy) was performed. Five months later, follow-up CT showed multiple metastasis in bilateral lung and left testicle. Serum level of neuron-specific enolase (NSE) was 24.4 ng/ml without an elevated in serum PSA level. He received rebiopsy of the prostate, but no malignant findings were observed. Consequently, bilateral orchiectomy was performed for diagnosis of left testicular tumor. Pathological examination revealed metastasis of neuroendocrine prostate cancer (NEPC). Chemotherapy using cisplatin and irinotecan was administered after orchiectomy. Complete response of lung lesions was achieved and serum level of NSE decreased within normal range. No recurrence has been confirmed for 4 years after the completion of chemotherapy.


Sujet(s)
Tumeurs de la prostate , Mâle , Humains , Sujet âgé , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/thérapie , Tumeurs de la prostate/traitement médicamenteux , Association thérapeutique , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/thérapie , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Facteurs temps , Tumeurs du testicule/anatomopathologie , Tumeurs du testicule/thérapie , Orchidectomie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du poumon/secondaire , Tumeurs du poumon/thérapie
20.
Chem Biol Drug Des ; 104(1): e14583, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38991995

RÉSUMÉ

In this work, a series of curcumin derivatives (1a-1h, 2a-2g, and 3a-3c) were synthesized for the suppression of castration-resistant prostate cancer cells. All synthesized compounds were characterized by 1H NMR, 13C NMR, HRMS, and melting point. The in vitro cytotoxicity study shows that compounds 1a, 1e, 1f, 1h, 2g, 3a, and 3c display similar or enhanced cytotoxicity against 22Rv1 and C4-2 cells as compared to ASC-J9, other synthesized compounds display reduced cytotoxicity against 22Rv1 and C4-2 cells as compared to ASC-J9. Molecular docking simulation was performed to study the binding affinity and probable binding modes of the synthesized compounds with androgen receptor. The results show that all synthesized compounds exhibit higher cdocker interaction energies as compared to ASC-J9. Compounds 1h, 2g, and 3c not only show strong cytotoxicity against 22Rv1 and C4-2 cells but also exhibit high binding affinity with androgen receptor. In androgen receptor suppression study, compounds 1f and 2g show similar androgen receptor suppression effect as compared to ASC-J9 on C4-2 cells, compound 3c displays significantly enhanced AR suppression effect as compared to ASC-J9, 1f and 2g. Compounds 1a, 1e, 1f, 1h, 2g, 3a and 3c prepared in this work have significant potential for castration-resistant prostate cancer therapy.


Sujet(s)
Curcumine , Simulation de docking moléculaire , Tumeurs prostatiques résistantes à la castration , Récepteurs aux androgènes , Curcumine/pharmacologie , Curcumine/composition chimique , Curcumine/synthèse chimique , Curcumine/métabolisme , Mâle , Humains , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/composition chimique , 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 , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Antagonistes du récepteur des androgènes/pharmacologie , Antagonistes du récepteur des androgènes/composition chimique , Antagonistes du récepteur des androgènes/synthèse chimique , Antagonistes du récepteur des androgènes/métabolisme , Sites de fixation , Liaison aux protéines
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE