Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters

Publication year range
1.
Int J Cancer ; 155(2): 314-323, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38491867

ABSTRACT

The addition of darolutamide, an androgen receptor signalling inhibitor, to therapy with docetaxel has recently been approved as a strategy to treat metastatic prostate cancer. OATP1B3 is an SLC transporter that is highly expressed in prostate cancer and is responsible for the accumulation of substrates, including docetaxel, into tumours. Given that darolutamide inhibits OATP1B3 in vitro, we sought to characterise the impact of darolutamide on docetaxel pharmacokinetics. We investigated the influence of darolutamide on OATP1B3 transport using in vitro and in vivo models. We assessed the impact of darolutamide on the tumour accumulation of docetaxel in a patient-derived xenograft (PDX) model and on an OATP1B biomarker in patients. Darolutamide inhibited OATP1B3 in vitro at concentrations higher than the reported Cmax. Consistent with these findings, in vivo studies revealed that darolutamide does not influence the pharmacokinetics of Oatp1b substrates, including docetaxel. Docetaxel accumulation in PDX tumours was not decreased in the presence of darolutamide. Metastatic prostate cancer patients had similar levels of OATP1B biomarkers, regardless of treatment with darolutamide. Consistent with a low potential to inhibit OATP1B3-mediated transport in vitro, darolutamide does not significantly impede the transport of Oatp1b substrates in vivo or in patients. Our findings support combined treatment with docetaxel and darolutamide, as no OATP1B3 transporter based drug-drug interaction was identified.


Subject(s)
Docetaxel , Prostatic Neoplasms , Pyrazoles , Solute Carrier Organic Anion Transporter Family Member 1B3 , Xenograft Model Antitumor Assays , Humans , Male , Docetaxel/pharmacology , Docetaxel/pharmacokinetics , Animals , Mice , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Pyrazoles/pharmacology , Pyrazoles/pharmacokinetics , Drug Interactions , Cell Line, Tumor , HEK293 Cells
2.
Prostate ; 84(7): 623-635, 2024 May.
Article in English | MEDLINE | ID: mdl-38450798

ABSTRACT

BACKGROUND: There are relatively few widely used models of prostate cancer compared to other common malignancies. This impedes translational prostate cancer research because the range of models does not reflect the diversity of disease seen in clinical practice. In response to this challenge, research laboratories around the world have been developing new patient-derived models of prostate cancer, including xenografts, organoids, and tumor explants. METHODS: In May 2023, we held a workshop at the Monash University Prato Campus for researchers with expertise in establishing and using a variety of patient-derived models of prostate cancer. This review summarizes our collective ideas on how patient-derived models are currently being used, the common challenges, and future opportunities for maximizing their usefulness in prostate cancer research. RESULTS: An increasing number of patient-derived models for prostate cancer are being developed. Despite their individual limitations and varying success rates, these models are valuable resources for exploring new concepts in prostate cancer biology and for preclinical testing of potential treatments. Here we focus on the need for larger collections of models that represent the changing treatment landscape of prostate cancer, robust readouts for preclinical testing, improved in vitro culture conditions, and integration of the tumor microenvironment. Additional priorities include ensuring model reproducibility, standardization, and replication, and streamlining the exchange of models and data sets among research groups. CONCLUSIONS: There are several opportunities to maximize the impact of patient-derived models on prostate cancer research. We must develop large, diverse and accessible cohorts of models and more sophisticated methods for emulating the intricacy of patient tumors. In this way, we can use the samples that are generously donated by patients to advance the outcomes of patients in the future.


Subject(s)
Prostatic Neoplasms , Male , Humans , Reproducibility of Results , Prostatic Neoplasms/therapy , Prostatic Neoplasms/pathology , Prostate/pathology , Organoids/pathology , Heterografts , Tumor Microenvironment
3.
Br J Cancer ; 130(8): 1377-1387, 2024 May.
Article in English | MEDLINE | ID: mdl-38396173

ABSTRACT

BACKGROUND/OBJECTIVE: To explore the anti-tumour activity of combining AKT inhibition and docetaxel in PTEN protein null and WT prostate tumours. METHODS: Mechanisms associated with docetaxel capivasertib treatment activity in prostate cancer were examined using a panel of in vivo tumour models and cell lines. RESULTS: Combining docetaxel and capivasertib had increased activity in PTEN null and WT prostate tumour models in vivo. In vitro short-term docetaxel treatment caused cell cycle arrest in the majority of cells. However, a sub-population of docetaxel-persister cells did not undergo G2/M arrest but upregulated phosphorylation of PI3K/AKT pathway effectors GSK3ß, p70S6K, 4E-BP1, but to a lesser extent AKT. In vivo acute docetaxel treatment induced p70S6K and 4E-BP1 phosphorylation. Treating PTEN null and WT docetaxel-persister cells with capivasertib reduced PI3K/AKT pathway activation and cell cycle progression. In vitro and in vivo it reduced proliferation and increased apoptosis or DNA damage though effects were more marked in PTEN null cells. Docetaxel-persister cells were partly reliant on GSK3ß as a GSK3ß inhibitor AZD2858 reversed capivasertib-induced apoptosis and DNA damage. CONCLUSION: Capivasertib can enhance anti-tumour effects of docetaxel by targeting residual docetaxel-persister cells, independent of PTEN status, to induce apoptosis and DNA damage in part through GSK3ß.


Subject(s)
Prostatic Neoplasms , Proto-Oncogene Proteins c-akt , Pyrimidines , Pyrroles , Male , Humans , Docetaxel/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/pharmacology , Signal Transduction , Apoptosis , Phosphatidylinositol 3-Kinases/metabolism , Glycogen Synthase Kinase 3 beta , Cell Line, Tumor , G2 Phase Cell Cycle Checkpoints , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , PTEN Phosphohydrolase/metabolism
4.
Am J Pathol ; 192(9): 1321-1335, 2022 09.
Article in English | MEDLINE | ID: mdl-35750257

ABSTRACT

Toll-like receptor 3 (TLR3) is an endosomal receptor expressed in several immune and epithelial cells. Recent studies have highlighted its expression also in solid tumors, including prostate cancer (PCa), and have described its role primarily in the proinflammatory response and induction of apoptosis. It is up-regulated in some castration-resistant prostate cancers. However, the role of TLR3 in prostate cancer progression remains largely unknown. The current study experimentally demonstrated that exogenous TLR3 activation in PCa cell lines leads to a significant induction of secretion of the cytokines IL-6, IL-8, and interferon-ß, depending on the model and chemoresistance status. Transcriptomic analysis of TLR3-overexpressing cells revealed a functional program that is enriched for genes involved in the regulation of cell motility, migration, and tumor invasiveness. Increased motility, migration, and invasion in TLR3-overexpressing cell line were confirmed by several in vitro assays and using an orthotopic prostate xenograft model in vivo. Furthermore, TLR3-ligand induced apoptosis via cleavage of caspase-3/7 and poly (ADP-ribose) polymerase, predominantly in TLR3-overexpressing cells. These results indicate that TLR3 may be involved in prostate cancer progression and metastasis; however, it might also represent an Achilles heel of PCa, which can be exploited for targeted therapy.


Subject(s)
Prostatic Neoplasms , Toll-Like Receptor 3 , Animals , Apoptosis , Cell Line, Tumor , Humans , Male , Poly I-C/pharmacology , Prostate/pathology , Prostatic Neoplasms/pathology , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism
5.
Prostate ; 82(5): 505-516, 2022 04.
Article in English | MEDLINE | ID: mdl-35037287

ABSTRACT

INTRODUCTION: Castration-resistant prostate cancer (CRPC) remains dependent on androgen receptor (AR) signalling, which is largely driven by conversion of adrenal androgen precursors lasting after castration. Abiraterone, an inhibitor of the steroidogenic enzyme CYP17A1, has been demonstrated to reduce adrenal androgen synthesis and prolong CRPC patient survival. To study mechanisms of resistance to castration and abiraterone, we created coculture models using human prostate and adrenal tumours. MATERIALS AND METHODS: Castration-naïve and CRPC clones of VCaP were incubated with steroid substrates or cocultured with human adrenal cells (H295R) and treated with abiraterone or the antiandrogen enzalutamide. Male mice bearing VCaP xenografts with and without concurrent H295R xenografts were castrated and treated with placebo or abiraterone. Response was assessed by tumour growth and PSA release. Plasma and tumour steroid levels were assessed by LC/MS-MS. Quantitative polymerase chain reaction determined steroidogenic enzyme, nuclear receptor and AR target gene expression. RESULTS: In vitro, adrenal androgens induced castration-naïve and CRPC cell growth, while precursors steroids for de novo synthesis did not. In a coculture system, abiraterone blocked H295R-induced growth of VCaP cells. In vivo, H295R promoted castration-resistant VCaP growth. Abiraterone only inhibited VCaP growth or PSA production in the presence of H295R. Plasma steroid levels demonstrated CYP17A1 inhibition by abiraterone, whilst CRPC tumour tissue steroid levels showed no evidence of de novo intratumoural androgen production. Castration-resistant and abiraterone-resistant VCaP tumours had increased levels of AR, AR variants and glucocorticoid receptor (GR) resulting in equal AR target gene expression levels compared to noncastrate tumours. CONCLUSIONS: In our model, ligand-dependent AR-regulated regrowth of CRPC was predominantly supported via adrenal androgen precursor production while there was no evidence for intratumoural androgen synthesis. Abiraterone-resistant tumours relied on AR overexpression, expression of ligand-independent AR variants and GR signalling.


Subject(s)
Androgens , Prostatic Neoplasms, Castration-Resistant , Androgens/metabolism , Androstenes/pharmacology , Androstenes/therapeutic use , Animals , Cell Line, Tumor , Humans , Ligands , Male , Mice , Nitriles/therapeutic use , Orchiectomy , Prostate-Specific Antigen , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Receptors, Glucocorticoid
6.
Eur J Nucl Med Mol Imaging ; 49(11): 3627-3638, 2022 09.
Article in English | MEDLINE | ID: mdl-35556158

ABSTRACT

PURPOSE: Targeting the prostate-specific membrane antigen (PSMA) using lutetium-177-labeled PSMA-specific tracers has become a very promising novel therapy option for prostate cancer (PCa). The efficacy of this therapy might be further improved by replacing the ß-emitting lutetium-177 with the α-emitting actinium-225. Actinium-225 is thought to have a higher therapeutic efficacy due to the high linear energy transfer (LET) of the emitted α-particles, which can increase the amount and complexity of the therapy induced DNA double strand breaks (DSBs). Here we evaluated the relative biological effectiveness of [225Ac]Ac-PSMA-I&T and [177Lu]Lu-PSMA-I&T by assessing in vitro binding characteristics, dosimetry, and therapeutic efficacy. METHODS AND RESULTS: The PSMA-expressing PCa cell line PC3-PIP was used for all in vitro assays. First, binding and displacement assays were performed, which revealed similar binding characteristics between [225Ac]Ac-PSMA-I&T and [177Lu]Lu-PSMA-I&T. Next, the assessment of the number of 53BP1 foci, a marker for the number of DNA double strand breaks (DSBs), showed that cells treated with [225Ac]Ac-PSMA-I&T had slower DSB repair kinetics compared to cells treated with [177Lu]Lu-PSMA-I&T. Additionally, clonogenic survival assays showed that specific targeting with [225Ac]Ac-PSMA-I&T and [177Lu]Lu-PSMA-I&T caused a dose-dependent decrease in survival. Lastly, after dosimetric assessment, the relative biological effectiveness (RBE) of [225Ac]Ac-PSMA-I&T was found to be 4.2 times higher compared to [177Lu]Lu-PSMA-I&T. CONCLUSION: We found that labeling of PSMA-I&T with lutetium-177 or actinium-225 resulted in similar in vitro binding characteristics, indicating that the distinct biological effects observed in this study are not caused by a difference in uptake of the two tracers. The slower repair kinetics of [225Ac]Ac-PSMA-I&T compared to [177Lu]Lu-PSMA-I&T correlates to the assumption that irradiation with actinium-225 causes more complex, more difficult to repair DSBs compared to lutetium-177 irradiation. Furthermore, the higher RBE of [225Ac]Ac-PSMA-I&T compared to [177Lu]Lu-PSMA-I&T underlines the therapeutic potential for the treatment of PCa.


Subject(s)
Lutetium , Prostatic Neoplasms, Castration-Resistant , Actinium , Cell Line, Tumor , DNA , Dipeptides , Heterocyclic Compounds, 1-Ring , Humans , Lutetium/therapeutic use , Male , Prostate-Specific Antigen , Prostatic Neoplasms, Castration-Resistant/drug therapy , Radioisotopes
7.
Int J Mol Sci ; 23(14)2022 Jul 21.
Article in English | MEDLINE | ID: mdl-35887398

ABSTRACT

Prostate specific membrane antigen targeted radionuclide therapy (PSMA-TRT) is a promising novel treatment for prostate cancer (PCa) patients. However, PSMA-TRT cannot be used for curative intent yet, thus additional research on how to improve the therapeutic efficacy is warranted. A potential way of achieving this, is combining TRT with poly ADP-ribosylation inhibitors (PARPi), which has shown promising results for TRT of neuroendocrine tumor cells. Currently, several clinical trials have been initiated for this combination for PCa, however so far, no evidence of synergism is available for PCa. Therefore, we evaluated the combination of PSMA-TRT with three classes of PARPi in preclinical PCa models. In vitro viability and survival assays were performed using PSMA-expressing PCa cell lines PC3-PIP and LNCaP to assess the effect of increasing concentrations of PARPi veliparib, olaparib or talazoparib in combination with PSMA-TRT compared to single PARPi treatment. Next, DNA damage analyses were performed by quantifying the number of DNA breaks by immunofluorescent stainings. Lastly, the potential of the combination treatments was studied in vivo in mice bearing PC3-PIP xenografts. Our results show that combining PSMA-TRT with PARPi did not synergistically affect the in vitro clonogenic survival or cell viability. DNA-damage analysis revealed only a significant increase in DNA breaks when combining PSMA-TRT with veliparib and not in the other combination treatments. Moreover, PSMA-TRT with PARPi treatment did not improve tumor control compared to PSMA-TRT monotherapy. Overall, the data presented do not support the assumption that combining PSMA-TRT with PARPi leads to a synergistic antitumor effect in PCa. These results underline that extensive preclinical research using various PCa models is imperative to validate the applicability of the combination strategy for PCa, as it is for other cancer types.


Subject(s)
Poly(ADP-ribose) Polymerase Inhibitors , Prostatic Neoplasms , Animals , Cell Line, Tumor , Humans , Male , Mice , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Prostate/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Radioisotopes/therapeutic use
8.
Eur J Nucl Med Mol Imaging ; 48(5): 1339-1350, 2021 05.
Article in English | MEDLINE | ID: mdl-33094433

ABSTRACT

PURPOSE: Various radiolabeled prostate-specific membrane antigen (PSMA)-targeting tracers are clinically applied for prostate cancer (PCa) imaging and targeted radionuclide therapy. The PSMA binding affinities, biodistribution, and DNA-damaging capacities of these radiotracers have not yet been compared in detail. A major concern of PSMA-targeting radiotracers is the toxicity in other PSMA-expressing organs, such as the salivary glands, thus demanding careful evaluation of the most optimal and safest radiotracer. In this extensive preclinical study, we evaluated the clinically applied PSMA-targeting small molecule inhibitors DOTA-PSMA-617 (PSMA-617) and DOTAGA-PSMA-I&T (PSMA-I&T) and the PSMA nanobody DOTA-JVZ-007 (JVZ-007) using PSMA-expressing cell lines, a unique set of PCa patient-derived xenografts (PDX) and healthy human tissues. METHODS AND RESULTS: In vitro displacement studies on PSMA-expressing cells and cryosections of a PSMA-positive PDX revealed high and specific binding affinity for all three tracers labeled with lutetium-177 with IC50 values in the nanomolar range. Interestingly, [177Lu]Lu-JVZ-007 could not be displaced by PSMA-617 or PSMA-I&T, suggesting that this tracer targets an alternative binding site. Autoradiography assays on cryosections of human salivary and renal tissues revealed [177Lu]Lu-PSMA-617 to have the lowest binding to these healthy organs compared with [177Lu]Lu-PSMA-I&T. In vivo biodistribution assays confirmed the in vitro results with comparable tumor uptake of [177Lu]Lu-PSMA-617 and [177Lu]Lu-PSMA-I&T at all timepoints, resulting in induction of similar levels of DNA double-strand breaks in the tumors. However, [177Lu]Lu-PSMA-I&T demonstrated approximately 40× higher renal uptake at 4 and 8 h post injection resulting in an unfavorable tumor-to-kidney ratio. CONCLUSION: [177Lu]Lu-PSMA-617 has the most favorable biodistribution in mice as well as more favorable binding characteristics in vitro in PSMA-positive cells and human kidney and salivary gland specimens compared with [177Lu]Lu-PSMA-I&T and [177Lu]Lu-JVZ-007. Based on our preclinical evaluation, [177Lu]Lu-PSMA-617 is the best performing tracer to be taken further into clinical evaluation for PSMA-targeted radiotherapeutic development although with careful evaluation of the tracer binding to PSMA-expressing organs.


Subject(s)
Glutamate Carboxypeptidase II , Prostatic Neoplasms , Animals , Antigens, Surface/metabolism , Cell Line, Tumor , Glutamate Carboxypeptidase II/metabolism , Humans , Lutetium/therapeutic use , Male , Mice , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/radiotherapy , Radioisotopes , Tissue Distribution
9.
Br J Cancer ; 123(12): 1715-1719, 2020 12.
Article in English | MEDLINE | ID: mdl-32989230

ABSTRACT

Androgen receptor (AR) signalling drives neoplastic growth and therapy resistance in prostate cancer. Recent clinical data show that docetaxel combined with androgen deprivation therapy improves outcome in hormone-sensitive disease. We studied whether testosterone and AR signalling interferes with docetaxel treatment efficacy in castration-resistant prostate cancer (CRPC). We found that testosterone supplementation significantly impaired docetaxel tumour accumulation in a CRPC model, resulting in decreased tubulin stabilisation and antitumour activity. Furthermore, testosterone competed with docetaxel for uptake by the drug transporter OATP1B3. Irrespective of docetaxel-induced tubulin stabilisation, AR signalling by testosterone counteracted docetaxel efficacy. AR-pathway activation could also reverse long-term tumour regression by docetaxel treatment in vivo. These results indicate that to optimise docetaxel efficacy, androgen levels and AR signalling need to be suppressed. This study lends evidence for continued maximum suppression of AR signalling by combining targeted therapeutics with docetaxel in CRPC.


Subject(s)
Androgen Antagonists/therapeutic use , Antineoplastic Agents/therapeutic use , Docetaxel/therapeutic use , Drug Resistance, Neoplasm/physiology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/metabolism , Testosterone/pharmacology , Acetylation , Androgen Antagonists/pharmacokinetics , Androgen Receptor Antagonists/therapeutic use , Animals , Antineoplastic Agents/pharmacokinetics , Cell Death , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival , Disease Progression , Docetaxel/pharmacokinetics , Drug Interactions , Humans , In Situ Nick-End Labeling , Male , Mice , Mice, Nude , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Prostate-Specific Antigen/biosynthesis , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/drug effects , Signal Transduction/drug effects , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Testosterone/administration & dosage , Testosterone/antagonists & inhibitors , Testosterone/metabolism , Tubulin/drug effects , Tubulin/metabolism
10.
Int J Mol Sci ; 21(16)2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32824188

ABSTRACT

Reporter genes are used to visualize intracellular biological phenomena, including viral infection. Here we demonstrate bioluminescent imaging of viral infection using the NanoBiT system in combination with intraperitoneal injection of a furimazine analogue, hydrofurimazine. This recently developed substrate has enhanced aqueous solubility allowing delivery of higher doses for in vivo imaging. The small high-affinity peptide tag (HiBiT), which is only 11 amino-acids in length, was engineered into a clinically used oncolytic adenovirus, and the complementary large protein (LgBiT) was constitutively expressed in tumor cells. Infection of the LgBiT expressing cells with the HiBiT oncolytic virus will reconstitute NanoLuc in the cytosol of the cell, providing strong bioluminescence upon treatment with substrate. This new bioluminescent system served as an early stage quantitative viral transduction reporter in vitro and also in vivo in mice, for longitudinal monitoring of oncolytic viral persistence in infected tumor cells. This platform provides novel opportunities for studying the biology of viruses in animal models.


Subject(s)
Furans/pharmacokinetics , Imidazoles/pharmacokinetics , Luminescent Agents/pharmacokinetics , Luminescent Proteins/genetics , Optical Imaging/methods , Pyrazines/pharmacokinetics , Virus Diseases/diagnostic imaging , Adenoviridae/genetics , Animals , Cell Line, Tumor , Furans/administration & dosage , HEK293 Cells , Humans , Imidazoles/administration & dosage , Injections, Intraperitoneal , Luminescent Agents/administration & dosage , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Oligopeptides/genetics , Oligopeptides/metabolism , Oncolytic Viruses/genetics , Pyrazines/administration & dosage , Recombinant Proteins/genetics
11.
J Cell Biochem ; 120(6): 10787-10795, 2019 06.
Article in English | MEDLINE | ID: mdl-30672018

ABSTRACT

Adoptive transfer of T cells expressing chimeric antigen receptors (CARs) is considered to be a novel anticancer therapy. To date, in most cases, single-chain variable fragments (scFvs) of murine origin have been used in CARs. However, this structure has limitations relating to the potential immunogenicity of mouse antigens in humans and the relatively large size of scFvs. For the first time, we used camelid nanobody (VHH) to construct CAR T cells against prostate specific membrane antigen (PSMA). The nanobody against PSMA (NBP) was used to show the feasibility of CAR T cells against prostate cancer cells. T cells were transfected, and then the surface expression of the CAR T cells was confirmed. Then, the functions of VHH-CAR T cell were evaluated upon coculture with prostate cancer cells. At the end, the cytotoxicity potential of NBPII-CAR in T cells was approximated by determining the cell surface expression of CD107a after encountering PSMA. Our data show the specificity of VHH-CAR T cells against PSMA+ cells (LNCaP), not only by increasing the interleukin 2 (IL-2) cytokine (about 400 pg/mL), but also the expression of CD69 by almost 38%. In addition, VHH-CAR T cells were proliferated by nearly 60% when cocultured with LNCaP, as compared with PSMA negative prostate cancer cell (DU-145), which led to the upregulation of CD107a in T cells upto 31%. These results clearly show the possibility of using VHH-based CAR T cells for targeted immunotherapy, which may be developed to target virtually any tumor-associated antigen for adoptive T-cell immunotherapy of solid tumors.


Subject(s)
Immunotherapy, Adoptive/methods , Kallikreins/genetics , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/therapy , Receptors, Chimeric Antigen/genetics , Single-Domain Antibodies/chemistry , T-Lymphocytes/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/immunology , Biomarkers/metabolism , Camelus , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Cytotoxicity, Immunologic , Electroporation , Gene Expression , Humans , Interleukin-2/genetics , Interleukin-2/immunology , Kallikreins/immunology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lysosomal-Associated Membrane Protein 1/genetics , Lysosomal-Associated Membrane Protein 1/immunology , Male , Plasmids/chemistry , Plasmids/immunology , Primary Cell Culture , Prostate/immunology , Prostate/pathology , Prostate-Specific Antigen/immunology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Receptors, Chimeric Antigen/immunology , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/isolation & purification , T-Lymphocytes/cytology
12.
Prostate ; 79(9): 937-948, 2019 06.
Article in English | MEDLINE | ID: mdl-31017696

ABSTRACT

BACKGROUND: Intratumoral steroidogenesis and its potential relevance in castration-resistant prostate cancer (CRPC) and in cytochrome P450, family 17, subfamily A, polypeptide 1 (CYP17A1)-inhibitor treated hormone-naïve and patients with CRPC are not well established. In this study, we tested if substrates for de novo steroidogenesis accumulating during CYP17A1 inhibition may drive cell growth in relevant preclinical models. METHODS: PCa cell lines and their respective CRPC sublines were used to model CRPC in vitro. Precursor steroids pregnenolone (Preg) and progesterone (Prog) served as substrate for de novo steroid synthesis. TAK700 (orteronel), abiraterone, and small interfering RNA (siRNA) against CYP17A1 were used to block CYP17A1 enzyme activity. The antiandrogen RD162 was used to assess androgen receptor (AR) involvement. Cell growth was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. AR-target gene expression was quantified by reverse transcription polymerase chain reaction (RT-PCR). Nuclear import studies using cells with green fluorescent protein (GFP)-tagged AR were performed to assess the potential of precursor steroids to directly activate AR. RESULTS: Preg and Prog stimulated cell proliferation and AR target gene expression in VCaP, DuCaP, LNCaP, and their respective CRPC sublines. The antiandrogen RD162, but not CYP17A1 inhibition with TAK700, abiraterone or siRNA, was able to block Preg- and Prog-induced proliferation. In contrast to TAK700, abiraterone also affected dihydrotestosterone-induced cell growth, indicating direct AR binding. Furthermore, Prog-induced AR translocation was not affected by treatment with TAK700 or abiraterone, while it was effectively blocked by the AR antagonist enzalutamide, further demonstrating the direct AR activation by Prog. CONCLUSION: Activation of the AR by clinically relevant levels of Preg and Prog accumulating in abiraterone-treated patients may act as a driver for CRPC. These data provide a scientific rationale for combining CYP17A1 inhibitors with antiandrogens, particularly in patients with overexpressed or mutated-AR.


Subject(s)
Abiraterone Acetate/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Steroid 17-alpha-Hydroxylase/metabolism , Cell Growth Processes/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Male , Pregnenolone/metabolism , Progesterone/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Signal Transduction , Steroids/biosynthesis
13.
Prostate ; 79(4): 390-402, 2019 03.
Article in English | MEDLINE | ID: mdl-30520109

ABSTRACT

BACKGROUND: In vitro models of prostate cancer (PCa) are not always reliable to evaluate anticancer treatment efficacy. This limitation may be overcome by using viable tumor slice material. Here we report on the establishment of an optimized ex vivo method to culture tissue slices from patient-derived xenografts (PDX) of prostate cancer (PCa), to assess responses to PCa treatments. METHODS: Three PDX models were used that are characterized by different androgen receptor (AR) expression and different homology directed DNA repair capacities, due to a breast cancer associated two (BRCA2) wild-type or mutated status. Tumors were removed from mice, sliced using a vibratome and cultured for a maximum of 6 days. To test the sensitivity to androgen antagonist, tumor slices from the AR-expressing and AR-negative PDX tumors were treated with the anti-androgen enzalutamide. For sensitivity to DNA repair intervention, tumors slices from BRCA2 wild-type and mutated PDXs were treated with the poly (ADP-ribose) polymerase-1 inhibitor olaparib. Treatment response in these tumor slices was determined by measuring slice morphology, cell proliferation, apoptosis, AR expression level, and secretion of prostate specific antigen (PSA). RESULTS: We compared various culture conditions (support materials, growth media, and use of a 3D smooth rocking platform) to define the optimal condition to maintain tissue viability and proliferative capacity up to least 6 days. Under optimized conditions, enzalutamide treatment significantly decreased proliferation, increased apoptosis, and reduced AR-expression and PSA secretion of AR-expressing tumor slices compared to AR-negative slices, that did not respond to the intervention. Olaparib treatment significantly increased cell death in BRCA2 mutated tumors slices as compared to slices from BRCA2 wild type tumors. CONCLUSIONS: Ex vivo treatment of PCa PDX tumor slices with enzalutamide and olaparib recapitulates responses previously observed in vivo. The faithful retention of tissue structure and function in this ex vivo model offers an ideal opportunity for treatment efficacy screening, thereby reducing costs and numbers of experimental animals.


Subject(s)
Neoplasm Transplantation , Prostatic Neoplasms/drug therapy , Tissue Culture Techniques/methods , Androgen Receptor Antagonists/therapeutic use , Animals , Apoptosis/drug effects , BRCA2 Protein/genetics , Benzamides , Cell Proliferation/drug effects , DNA Repair/drug effects , Humans , Male , Mice , Mice, Nude , Mutation , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/therapeutic use , Phthalazines/therapeutic use , Piperazines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/analysis , Xenograft Model Antitumor Assays
14.
Prostate ; 78(13): 1013-1023, 2018 09.
Article in English | MEDLINE | ID: mdl-30133757

ABSTRACT

BACKGROUND: Prostate cancer is recognized as a heterogeneous disease demanding appropriate preclinical models that reflect tumor complexity. Previously, we established the PSA-Cre;PtenLoxP/LoxP genetic engineered mouse model (GEMM) for prostate cancer reflecting the various stages of tumor development. Prostate tumors in this Pten KO model slowly develop, requiring more than 10 months. In order to enhance its practical utility, we established a syngeneic panel of cell lines derived from PSA-Cre targeted Pten KO tumors, designated the mouse prostate cancer (MuCap) model. METHODS: Four different MuCaP epithelial cell lines were established from three independent primary Pten KO mouse prostate tumors. Tumorigenic capacity of the MuCaP cell lines was determined by subcutaneous inoculation of these cell lines in immunocompetent mice. Response to PI3K-targeted therapy was validated in ex vivo tissue slices of the established MuCaP tumors. RESULTS: The MuCaP cell lines were all tumorigenic in immunocompetent mice after subcutaneous inoculation. Interestingly, these syngrafted tumors represented different tumor growth rates and morphologies. Treatment with the specific PI3K inhibitor GDC0941 resulted in responses very similar between syngeneic MuCaP and primary Pten KO prostate tumors. Finally, immunoprofiling of the different syngeneic MuCaP tumors demonstrated differential numbers of tumor infiltrating lymphocytes and distinct immune gene profiles with expression of CD8, INFy, and PD1 being inversely related to tumor aggressiveness. CONCLUSIONS: Collectively, we present here a well-defined MuCaP platform of in vitro and in vivo mouse prostate cancer models that may support preclinical assessment of (immune)-therapies for prostate cancer.


Subject(s)
Neoplasm Invasiveness/pathology , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Disease Models, Animal , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Mice , Mice, Knockout , Neoplasm Invasiveness/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism
15.
Prostate ; 78(16): 1262-1282, 2018 12.
Article in English | MEDLINE | ID: mdl-30073676

ABSTRACT

BACKGROUND: While it has been challenging to establish prostate cancer patient-derived xenografts (PDXs), with a take rate of 10-40% and long latency time, multiple groups throughout the world have developed methods for the successful establishment of serially transplantable human prostate cancer PDXs using a variety of immune deficient mice. In 2014, the Movember Foundation launched a Global Action Plan 1 (GAP1) project to support an international collaborative prostate cancer PDX program involving eleven groups. Between these Movember consortium members, a total of 98 authenticated human prostate cancer PDXs were available for characterization. Eighty three of these were derived directly from patient material, and 15 were derived as variants of patient-derived material via serial passage in androgen deprived hosts. A major goal of the Movember GAP1 PDX project was to provide the prostate cancer research community with a summary of both the basic characteristics of the 98 available authenticated serially transplantable human prostate cancer PDX models and the appropriate contact information for collaborations. Herein, we report a summary of these PDX models. METHODS: PDX models were established in immunocompromised mice via subcutaneous or subrenal-capsule implantation. Dual-label species (ie, human vs mouse) specific centromere and telomere Fluorescence In Situ Hybridization (FISH) and immuno-histochemical (IHC) staining of tissue microarrays (TMAs) containing replicates of the PDX models were used for characterization of expression of a number of phenotypic markers important for prostate cancer including AR (assessed by IHC and FISH), Ki67, vimentin, RB1, P-Akt, chromogranin A (CgA), p53, ERG, PTEN, PSMA, and epithelial cytokeratins. RESULTS: Within this series of PDX models, the full spectrum of clinical disease stages is represented, including androgen-sensitive and castration-resistant primary and metastatic prostate adenocarcinomas as well as prostate carcinomas with neuroendocrine differentiation. The annotated clinical characteristics of these PDXs were correlated with their marker expression profile. CONCLUSION: Our results demonstrate the clinical relevance of this series of PDXs as a platform for both basic science studies and therapeutic discovery/drug development. The present report provides the prostate cancer community with a summary of the basic characteristics and a contact information for collaborations using these models.


Subject(s)
Heterografts , Neoplasm Transplantation/methods , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays , Animals , Biomarkers, Tumor/metabolism , Disease Models, Animal , Humans , Male , Mice , Prostatic Neoplasms/metabolism
17.
Br J Cancer ; 115(6): 674-81, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27537383

ABSTRACT

BACKGROUND: Both taxanes, docetaxel and cabazitaxel, are effective treatments for metastatic castration-resistant prostate cancer (mCRPC). However, resistance to taxanes is common. Our objective was to investigate mechanisms of taxane resistance in prostate cancer. METHODS: Two docetaxel-resistant patient-derived xenografts (PDXs) of CRPC were established (PC339-DOC and PC346C-DOC) in male athymic nude mice by frequent intraperitoneal administrations of docetaxel. Next-generation sequencing was performed on PDX tissue pre- and post-docetaxel resistance and gene expression profiles were compared. [(14)C]-docetaxel and [(14)C]-cabazitaxel uptake assays in vitro and cytotoxicity assays were performed to validate direct involvement of transporter genes in taxane sensitivity. RESULTS: Organic anion-transporting polypeptide (SLCO1B3), an influx transporter of docetaxel, was significantly downregulated in PC346C-DOC tumours. In accordance with this finding, intratumoural concentrations of docetaxel and cabazitaxel were significantly decreased in PC346C-DOC as compared with levels in chemotherapy-naive PC346C tumours. In addition, silencing of SLCO1B3 in chemo-naive PC346C resulted in a two-fold decrease in intracellular concentrations of both taxanes. Overexpression of SLCO1B3 showed higher sensitivity to docetaxel and cabazitaxel. CONCLUSIONS: The SLCO1B3 determines intracellular concentrations of docetaxel and cabazitaxel and consequently influences taxane efficacy. Loss of the drug transporter SLCO1B3 may drive taxane resistance in prostate cancer.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents, Phytogenic/pharmacology , Drug Resistance, Neoplasm/physiology , Neoplasm Proteins/physiology , Organic Anion Transporters, Sodium-Independent/physiology , Prostatic Neoplasms/drug therapy , Taxoids/pharmacology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Androgens , Androstenes/pharmacology , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/therapeutic use , Benzamides , Biological Transport , Cell Line, Tumor , Docetaxel , Drug Resistance, Multiple/genetics , Drug Resistance, Multiple/physiology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Heterografts , Humans , Male , Mice, Nude , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Nitriles , Organic Anion Transporters, Sodium-Independent/biosynthesis , Organic Anion Transporters, Sodium-Independent/genetics , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA Interference , RNA, Small Interfering/pharmacology , Solute Carrier Organic Anion Transporter Family Member 1B3 , Taxoids/pharmacokinetics , Taxoids/therapeutic use
18.
J Steroid Biochem Mol Biol ; 236: 106446, 2024 02.
Article in English | MEDLINE | ID: mdl-38104728

ABSTRACT

Prostate cancer (PC) is dependent on androgen receptor (AR) activation by testosterone and 5α-dihydrotestosterone (DHT). Intratumoral androgen accumulation and activation despite systemic androgen deprivation therapy underlies the development of castration-resistant PC (CRPC), but the precise pathways involved remain controversial. Here we investigated the differential contributions of de novo androgen biosynthesis and androgen precursor conversion to androgen accumulation. Steroid flux analysis by liquid chromatography-tandem mass spectrometry (LC-MS/MS) was performed on (CR)PC cell lines and fresh patient PC tissue slices after incubation with classic and alternative biosynthesis intermediates, alongside quantitative PCR analysis for steroidogenic enzyme expression. Activity of CYP17A1 was undetectable in all PC cell lines and patient PC tissue slices. Instead, steroid flux analysis confirmed the generation of testosterone and DHT from adrenal precursors and reactivation of androgen metabolites. Precursor steroids upstream of DHEA were converted down the first steps of the alternative DHT biosynthesis pathway, but did not proceed through to active androgen generation. Comprehensive steroid flux analysis of (CR)PC cells provides strong evidence against intratumoral de novo androgen biosynthesis and demonstrates that androgen precursor steroids downstream of CYP17A1 activities constitute the major source of intracrine androgen generation.


Subject(s)
Prostatic Neoplasms , Male , Humans , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Androgens/metabolism , Androgen Antagonists , Chromatography, Liquid , Tandem Mass Spectrometry , Testosterone/metabolism , Dihydrotestosterone/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Steroids/metabolism , Cell Line, Tumor , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism
19.
Prostate ; 73(15): 1636-50, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23996639

ABSTRACT

BACKGROUND: Despite an initial response to hormonal therapy, patients with advanced prostate cancer (PC) almost always progress to castration-resistant disease (CRPC). Although serum testosterone (T) is reduced by androgen deprivation therapy, intratumoral T levels in CRPC are comparable to those in prostate tissue of eugonadal men. These levels could originate from intratumoral conversion of adrenal androgens and/or from de novo steroid synthesis. However, the relative contribution of de novo steroidogenesis to AR-driven cell growth is unknown. METHODS: The relative contribution of androgen biosynthetic pathways to activate androgen receptor (AR)-regulated cell growth and expression of PSA, FKBP5, and TMPRSS2 was studied at physiologically relevant levels of adrenal androgen precursors and intermediates of de novo androgen biosynthesis in human prostate cancer cell lines, PC346C, VCaP, and LNCaP. RESULTS: In PC346C and VCaP, responses to pregnenolone and progesterone were absent or minimal, while large effects of adrenal androgen precursors were found. VCaP CRPC clones overexpressing CYP17A1 did not acquire an increased ability to use pregnenolone or progesterone to activate AR. In contrast, all precursors stimulated growth and gene expression in LNCaP cells, presumably resulting from the mutated AR in these cells. CONCLUSIONS: Our data indicate that at physiological levels of T precursors PC cells can generally convert adrenal androgens, while de novo steroidogenesis is not generally possible in PC cells and is not able to support AR transactivation and PC growth.


Subject(s)
Androgens/biosynthesis , Cell Proliferation , Prostate/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Cell Line, Tumor , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism
20.
Res Involv Engagem ; 9(1): 85, 2023 Sep 26.
Article in English | MEDLINE | ID: mdl-37752584

ABSTRACT

BACKGROUND: Metastatic cancer is often experienced by patients as a death sentence. At the same time, translational scientists approach metastasis also as an interesting phenomenon that they try to understand and prevent. These two sides of the same coin do not mask the considerable gap that exists between the laboratory world of scientists and the life world of patients. Funding agencies nowadays increasingly demand researchers to be responsive to the values and priorities of patients and public. One approach to bridge this gap and to increase the impact of science is patient and public involvement (PPI). A concise literature review of PPI research and practice in this paper revealed that although PPI is often deployed in translational health care research, its methodology is not settled, it is not sufficiently emancipatory, and its implementation in basic and translational science is lagging behind. Here, we illustrate the practical implementation of PPI in basic and translational science, namely in the context of HOUDINI, a multidisciplinary network with the ultimate goal to improve the management of metastatic disease. METHODS: This paper reports on a societal workshop that was organized to launch the holistic PPI approach of HOUDINI. During this workshop, societal partners, patients, and physicians discussed societal issues regarding cancer metastasis, and contributed to prioritization of research objectives for HOUDINI. In a later stage, the workshop results were discussed with scientists from the network to critically review its research strategy and objectives. RESULTS: Workshop participants chose the development of metastasis prediction tools, effective therapies which preserve good quality of life, and non-invasive tissue sampling methods as most important research objectives for HOUDINI. Importantly, during the discussions, mutual understanding about issues like economic feasibility of novel therapies, patient anxiety for metastases, and clear communication between stakeholders was further increased. CONCLUSIONS: In conclusion, the PPI workshop delivered valuable early-stage input and connections for HOUDINI, and may serve as example for similar basic and translational research projects.


Metastatic cancer is an aggravated form of cancer, that patients are afraid of. At the same time, cancer researchers are fascinated by this disease. Therefore, there is an apparent gap between how patients and researchers feel about cancer. If researchers wish to be most helpful to cancer patients, it is important to consult the patients and ask what they need and find important. This is also stimulated by agencies that financially support research projects. A possible way to do this is Patient and Public Involvement (PPI), in which not only scientists, but also patients and lay people are asked to provide input. It appears, however, that PPI is scarcely applied in basic science. In this article, we describe how a collaborative network of basic and translational cancer researchers, HOUDINI, intends to include the patients' voice throughout the research progress and actively asked for input from patients, societal partners and physicians at the start of their project. These people discussed what themes they found most important to be researched by HOUDINI. Later, the HOUDINI researchers reflected on this. This example shows how PPI can be applied and how HOUDINI received valuable input for their research goals.

SELECTION OF CITATIONS
SEARCH DETAIL